1
|
Thanh DD, Bich-Ngoc N, Paques C, Christian A, Herkenne S, Struman I, Muller M. The food dye Tartrazine disrupts vascular formation both in zebrafish larvae and in human primary endothelial cells. Sci Rep 2024; 14:30367. [PMID: 39639097 PMCID: PMC11621646 DOI: 10.1038/s41598-024-82076-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Tartrazine (E102) is a controversial coloring agent whose potential impacts on human health are not fully understood. Our study reveals the vascular disrupting effects of tartrazine (TTZ) on developing zebrafish embryos in vivo and on human umbilical vein endothelial cells in vitro. The dye was shown to cause dose-dependent hemorrhages in zebrafish embryos. Analyzing transgenic zebrafish harboring fluorescent endothelial cells revealed that TTZ treatment disrupted cell organization into vessels in both the sub-intestinal vein and the brain area. Assays on human umbilical vein endothelial cells demonstrated that TTZ inhibited endothelial proliferation, tube formation, and migration in a dose-dependent manner. Taken together, our results indicate for the first time that TTZ can affect endothelial cell properties, possibly by disrupting Rho family GTPase pathways which control the cytoskeleton. Our finding provides a credible explanation for many reported human health impacts and offers prospective applications for biomedicine.
Collapse
Affiliation(s)
- Dinh Duy Thanh
- Lab. for Organogenesis and Regeneration, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
- Department of Cell Biology, Faculty of Biology, VNU University of Science, Hanoi, 100000, Vietnam
| | - Nguyen Bich-Ngoc
- VNU School of Interdisciplinary Sciences and Arts, Vietnam National University, Hanoi, 100000, Vietnam
| | - Cécile Paques
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Aurélie Christian
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Stéphanie Herkenne
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Ingrid Struman
- Lab. of Molecular Angiogenesis, GIGA-Institute, Université de Liège, Liège, 4000, Belgium
| | - Marc Muller
- Lab. for Organogenesis and Regeneration, GIGA-Institute, Université de Liège, Liège, 4000, Belgium.
| |
Collapse
|
2
|
Huveneers S, Phng LK. Endothelial cell mechanics and dynamics in angiogenesis. Curr Opin Cell Biol 2024; 91:102441. [PMID: 39342870 DOI: 10.1016/j.ceb.2024.102441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024]
Abstract
The efficient distribution of oxygen and metabolites is critical for embryonic development and growth as well as tissue homeostasis. This is achieved by endothelial cells forming and maintaining a closed, circulatory network of tubular blood vessels. Endothelial cells are highly plastic cells with the capability to generate diverse dynamic responses at different stages of vessel development in order to build vessel networks of tissue-specific patterns and morphologies. In this review, we discuss new conceptual advances gained from in vitro and in vivo models of angiogenesis on the control of endothelial cell dynamics. We highlight the complex interplay between mechanical cues, actin cytoskeleton and endothelial behaviors, and the emerging importance of hydrostatic pressure in complementing actin-dependent mechanisms to regulate endothelial cell mechanics and angiogenesis. Understanding these processes provides insights into vascular repair and regeneration mechanisms.
Collapse
Affiliation(s)
- Stephan Huveneers
- Amsterdam UMC, Location University of Amsterdam, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| | - Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.
| |
Collapse
|
3
|
Cheng C, Chen J, Zhang L, Huang B, Ma J, Luo L, Yang Y. Migration and proliferation of ductal cells promote pancreatic repair after trauma. J Genet Genomics 2024; 51:1318-1321. [PMID: 39214453 DOI: 10.1016/j.jgg.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/18/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Chaoqing Cheng
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing 400715, China
| | - Jinzi Chen
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing 400715, China
| | - Liqi Zhang
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing 400715, China
| | - Bangzhuo Huang
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing 400715, China
| | - Jianlong Ma
- School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Lingfei Luo
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing 400715, China; School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yun Yang
- Institute of Development Biology and Regenerative Medicine, Southwest University, Chongqing 400715, China.
| |
Collapse
|
4
|
Alvarez YD, van der Spuy M, Wang JX, Noordstra I, Tan SZ, Carroll M, Yap AS, Serralbo O, White MD. A Lifeact-EGFP quail for studying actin dynamics in vivo. J Cell Biol 2024; 223:e202404066. [PMID: 38913324 PMCID: PMC11194674 DOI: 10.1083/jcb.202404066] [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: 04/12/2024] [Revised: 05/15/2024] [Accepted: 06/03/2024] [Indexed: 06/25/2024] Open
Abstract
Here, we report the generation of a transgenic Lifeact-EGFP quail line for the investigation of actin organization and dynamics during morphogenesis in vivo. This transgenic avian line allows for the high-resolution visualization of actin structures within the living embryo, from the subcellular filaments that guide cell shape to the supracellular assemblies that coordinate movements across tissues. The unique suitability of avian embryos to live imaging facilitates the investigation of previously intractable processes during embryogenesis. Using high-resolution live imaging approaches, we present the dynamic behaviors and morphologies of cellular protrusions in different tissue contexts. Furthermore, through the integration of live imaging with computational segmentation, we visualize cells undergoing apical constriction and large-scale actin structures such as multicellular rosettes within the neuroepithelium. These findings not only enhance our understanding of tissue morphogenesis but also demonstrate the utility of the Lifeact-EGFP transgenic quail as a new model system for live in vivo investigations of the actin cytoskeleton.
Collapse
Affiliation(s)
- Yanina D. Alvarez
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Marise van der Spuy
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Jian Xiong Wang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Ivar Noordstra
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Siew Zhuan Tan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Murron Carroll
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Alpha S. Yap
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Olivier Serralbo
- Commonwealth Scientific and Industrial Research (CSIRO) Health and Biosecurity, Geelong, Australia
| | - Melanie D. White
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| |
Collapse
|
5
|
Shah FH, Lee HW. Endothelial and macrophage interactions in the angiogenic niche. Cytokine Growth Factor Rev 2024; 78:64-76. [PMID: 39019663 DOI: 10.1016/j.cytogfr.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024]
Abstract
The interactions between vascular cells, especially endothelial cells, and macrophages play a pivotal role in maintaining the subtle balance of vascular biology, which is crucial for angiogenesis in both healthy and diseased states. These cells are central to ensuring a harmonious balance between tissue repair and preventing excessive angiogenic activity, which could lead to pathological conditions. Recent advances in sophisticated genetic engineering vivo models and novel sequencing approaches, such as single-cell RNA-sequencing, in immunobiology have significantly enhanced our understanding of the gene expression and behavior of macrophages. These insights offer new perspectives on the role macrophages play not only in development but also across various health conditions. In this review, we explore the complex interactions between multiple types of macrophages and endothelium, focusing on their impact on new blood vessel formation. By understanding these intricate interactions, we aim to provide insights into new methods for managing angiogenesis in various diseases, thereby offering hope for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Fahad Hassan Shah
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Heon-Woo Lee
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea.
| |
Collapse
|
6
|
Malchow J, Eberlein J, Li W, Hogan BM, Okuda KS, Helker CSM. Neural progenitor-derived Apelin controls tip cell behavior and vascular patterning. SCIENCE ADVANCES 2024; 10:eadk1174. [PMID: 38968355 PMCID: PMC11225789 DOI: 10.1126/sciadv.adk1174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 05/31/2024] [Indexed: 07/07/2024]
Abstract
During angiogenesis, vascular tip cells guide nascent vascular sprouts to form a vascular network. Apelin, an agonist of the G protein-coupled receptor Aplnr, is enriched in vascular tip cells, and it is hypothesized that vascular-derived Apelin regulates sprouting angiogenesis. We identify an apelin-expressing neural progenitor cell population in the dorsal neural tube. Vascular tip cells exhibit directed elongation and migration toward and along the apelin-expressing neural progenitor cells. Notably, restoration of neural but not vascular apelin expression in apelin mutants remedies the angiogenic defects of mutants. By functional analyses, we show the requirement of Apelin signaling for tip cell behaviors, like filopodia formation and cell elongation. Through genetic interaction studies and analysis of transgenic activity reporters, we identify Apelin signaling as a modulator of phosphoinositide 3-kinase and extracellular signal-regulated kinase signaling in tip cells in vivo. Our results suggest a previously unidentified neurovascular cross-talk mediated by Apelin signaling that is important for tip cell function during sprouting angiogenesis.
Collapse
Affiliation(s)
- Julian Malchow
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University of Marburg, Marburg, Germany
| | - Jean Eberlein
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University of Marburg, Marburg, Germany
| | - Wei Li
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University of Marburg, Marburg, Germany
| | - Benjamin M. Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria 3000, Australia
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Kazuhide S. Okuda
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria 3000, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Christian S. M. Helker
- Faculty of Biology, Cell Signaling and Dynamics, Philipps-University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus-Liebig-University Giessen, Marburg, Germany
| |
Collapse
|
7
|
Abbey CA, Duran CL, Chen Z, Chen Y, Roy S, Coffell A, Sveeggen TM, Chakraborty S, Wells GB, Chang J, Bayless KJ. Identification of New Markers of Angiogenic Sprouting Using Transcriptomics: New Role for RND3. Arterioscler Thromb Vasc Biol 2024; 44:e145-e167. [PMID: 38482696 PMCID: PMC11043006 DOI: 10.1161/atvbaha.123.320599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/28/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND New blood vessel formation requires endothelial cells to transition from a quiescent to an invasive phenotype. Transcriptional changes are vital for this switch, but a comprehensive genome-wide approach focused exclusively on endothelial cell sprout initiation has not been reported. METHODS Using a model of human endothelial cell sprout initiation, we developed a protocol to physically separate cells that initiate the process of new blood vessel formation (invading cells) from noninvading cells. We used this model to perform multiple transcriptomics analyses from independent donors to monitor endothelial gene expression changes. RESULTS Single-cell population analyses, single-cell cluster analyses, and bulk RNA sequencing revealed common transcriptomic changes associated with invading cells. We also found that collagenase digestion used to isolate single cells upregulated the Fos proto-oncogene transcription factor. Exclusion of Fos proto-oncogene expressing cells revealed a gene signature consistent with activation of signal transduction, morphogenesis, and immune responses. Many of the genes were previously shown to regulate angiogenesis and included multiple tip cell markers. Upregulation of SNAI1 (snail family transcriptional repressor 1), PTGS2 (prostaglandin synthase 2), and JUNB (JunB proto-oncogene) protein expression was confirmed in invading cells, and silencing JunB and SNAI1 significantly reduced invasion responses. Separate studies investigated rounding 3, also known as RhoE, which has not yet been implicated in angiogenesis. Silencing rounding 3 reduced endothelial invasion distance as well as filopodia length, fitting with a pathfinding role for rounding 3 via regulation of filopodial extensions. Analysis of in vivo retinal angiogenesis in Rnd3 heterozygous mice confirmed a decrease in filopodial length compared with wild-type littermates. CONCLUSIONS Validation of multiple genes, including rounding 3, revealed a functional role for this gene signature early in the angiogenic process. This study expands the list of genes associated with the acquisition of a tip cell phenotype during endothelial cell sprout initiation.
Collapse
Affiliation(s)
- Colette A. Abbey
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Camille L. Duran
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Zhishi Chen
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Houston, TX
| | - Yanping Chen
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Houston, TX
| | - Sukanya Roy
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
| | - Ashley Coffell
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Timothy M. Sveeggen
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| | - Sanjukta Chakraborty
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
| | - Gregg B. Wells
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
- Department of Cell Biology and Genetics, Texas A&M School of Medicine, Bryan, TX
| | - Jiang Chang
- Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Houston, TX
| | - Kayla J. Bayless
- Texas A&M Health, Department of Medical Physiology, Texas A&M School of Medicine, Bryan TX
- Department of Molecular & Cellular Medicine, Texas A&M School of Medicine, Bryan, TX
| |
Collapse
|
8
|
Wilsch-Bräuninger M, Peters J, Huttner WB. High-resolution 3D ultrastructural analysis of developing mouse neocortex reveals long slender processes of endothelial cells that enter neural cells. Front Cell Dev Biol 2024; 12:1344734. [PMID: 38500687 PMCID: PMC10945550 DOI: 10.3389/fcell.2024.1344734] [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: 11/26/2023] [Accepted: 01/31/2024] [Indexed: 03/20/2024] Open
Abstract
The development of the neocortex involves an interplay between neural cells and the vasculature. However, little is known about this interplay at the ultrastructural level. To gain a 3D insight into the ultrastructure of the developing neocortex, we have analyzed the embryonic mouse neocortex by serial block-face scanning electron microscopy (SBF-SEM). In this study, we report a first set of findings that focus on the interaction of blood vessels, notably endothelial tip cells (ETCs), and the neural cells in this tissue. A key observation was that the processes of ETCs, located either in the ventricular zone (VZ) or subventricular zone (SVZ)/intermediate zone (IZ), can enter, traverse the cytoplasm, and even exit via deep plasma membrane invaginations of the host cells, including apical progenitors (APs), basal progenitors (BPs), and newborn neurons. More than half of the ETC processes were found to enter the neural cells. Striking examples of this ETC process "invasion" were (i) protrusions of apical progenitors or newborn basal progenitors into the ventricular lumen that contained an ETC process inside and (ii) ETC process-containing protrusions of neurons that penetrated other neurons. Our observations reveal a - so far unknown - complexity of the ETC-neural cell interaction.
Collapse
Affiliation(s)
| | | | - Wieland B. Huttner
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
9
|
Furtado J, Eichmann A. Vascular development, remodeling and maturation. Curr Top Dev Biol 2024; 159:344-370. [PMID: 38729681 DOI: 10.1016/bs.ctdb.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The development of the vascular system is crucial in supporting the growth and health of all other organs in the body, and vascular system dysfunction is the major cause of human morbidity and mortality. This chapter discusses three successive processes that govern vascular system development, starting with the differentiation of the primitive vascular system in early embryonic development, followed by its remodeling into a functional circulatory system composed of arteries and veins, and its final maturation and acquisition of an organ specific semi-permeable barrier that controls nutrient uptake into tissues and hence controls organ physiology. Along these steps, endothelial cells forming the inner lining of all blood vessels acquire extensive heterogeneity in terms of gene expression patterns and function, that we are only beginning to understand. These advances contribute to overall knowledge of vascular biology and are predicted to unlock the unprecedented therapeutic potential of the endothelium as an avenue for treatment of diseases associated with dysfunctional vasculature.
Collapse
Affiliation(s)
- Jessica Furtado
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, United States; Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Anne Eichmann
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, United States; Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States; Paris Cardiovascular Research Center, Inserm U970, Université Paris, Paris, France.
| |
Collapse
|
10
|
Teichmann T, Malacarne P, Zehr S, Günther S, Pflüger-Müller B, Warwick T, Brandes RP. NCoR1 limits angiogenic capacity by altering Notch signaling. J Mol Cell Cardiol 2024; 188:65-78. [PMID: 38359551 DOI: 10.1016/j.yjmcc.2024.02.003] [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: 12/11/2023] [Revised: 01/15/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Abstract
Corepressors negatively regulate gene expression by chromatin compaction. Targeted regulation of gene expression could provide a means to control endothelial cell phenotype. We hypothesize that by targeting corepressor proteins, endothelial angiogenic function can be improved. To study this, the expression and function of nuclear corepressors in human umbilical vein endothelial cells (HUVEC) and in murine organ culture was studied. RNA-seq revealed that nuclear receptor corepressor 1 (NCoR1), silencing mediator of retinoid and thyroid hormone receptors (SMRT) and repressor element-1 silencing transcription factor (REST) are the highest expressed corepressors in HUVECs. Knockout and knockdown strategies demonstrated that the depletion of NCoR1 increased the angiogenic capacity of endothelial cells, whereas depletion of SMRT or REST did not. Interestingly, the effect was VEGF signaling independent. NCoR1 depletion significantly upregulated angiogenesis-associated genes, especially tip cell genes, including ESM1, DLL4 and NOTCH4, as observed by RNA- and ATAC-seq. Confrontation assays comparing cells with and without NCoR1-deficiency revealed that loss of NCoR1 promotes a tip-cell position during spheroid sprouting. Moreover, a proximity ligation assay identified NCoR1 as a direct binding partner of the Notch-signaling-related transcription factor RBPJk. Luciferase assays showed that siRNA-mediated knockdown of NCOR1 promotes RBPJk activity. Furthermore, NCoR1 depletion prompts upregulation of several elements in the Notch signaling cascade. Downregulation of NOTCH4, but not NOTCH1, prevented the positive effect of NCOR1 knockdown on spheroid outgrowth. Collectively, these data indicate that decreasing NCOR1 expression is an attractive approach to promote angiogenic function.
Collapse
Affiliation(s)
- Tom Teichmann
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main 60590, Germany; German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Pedro Malacarne
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main 60590, Germany; German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Simonida Zehr
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main 60590, Germany; German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Stefan Günther
- Max-Planck-Institute for Heart- and Lung Research (MPI-HLR), Bad Nauheim 61231, Germany
| | - Beatrice Pflüger-Müller
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main 60590, Germany; German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Timothy Warwick
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main 60590, Germany; German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany.
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main 60590, Germany; German Center for Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany.
| |
Collapse
|
11
|
Zhou J, Zhou P, Wang J, Song J. Roles of endothelial cell specific molecule‑1 in tumor angiogenesis (Review). Oncol Lett 2024; 27:137. [PMID: 38357478 PMCID: PMC10865172 DOI: 10.3892/ol.2024.14270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Angiogenesis plays a crucial role in tumor growth and metastasis, and is heavily influenced by the tumor microenvironment (TME). Endothelial cell dysfunction is a key factor in tumor angiogenesis and is characterized by the aberrant expression of pro-angiogenic factors. Endothelial cell specific molecule-1 (ESM1), also known as endocan, is a marker of endothelial cell dysfunction. Although ESM1 is primarily expressed in normal endothelial cells, dysregulated ESM1 expression has been observed in human tumors and animal tumor models, and implicated in tumor growth, metastasis and angiogenesis. The precise role of ESM1 in tumor angiogenesis and its potential regulatory mechanisms are not yet conclusively defined. However, the aim of the present review was to explore the involvement of ESM1 in the process of tumor angiogenesis in the TME and the characteristics of neovascularization. In addition, the present review discusses the interaction between ESM1 and angiogenic factors, as well as the mechanisms through which ESM1 contributes to tumor angiogenesis. Furthermore, the reciprocal regulation between ESM1 and the TME is explored. Finally, the potential of targeting ESM1 as a therapeutic strategy for tumor angiogenesis is presented.
Collapse
Affiliation(s)
- Jie Zhou
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Ping Zhou
- College of Chinese Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Jinfang Wang
- College of Nursing, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Jie Song
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| |
Collapse
|
12
|
Röss H, Aaldijk D, Vladymyrov M, Odriozola A, Djonov V. Transluminal Pillars-Their Origin and Role in the Remodelling of the Zebrafish Caudal Vein Plexus. Int J Mol Sci 2023; 24:16703. [PMID: 38069025 PMCID: PMC10706262 DOI: 10.3390/ijms242316703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Intussusceptive pillars, regarded as a hallmark of intussusceptive angiogenesis, have been described in developing vasculature of many organs and organisms. The aim of this study was to resolve the question about pillar formation and their further maturation employing zebrafish caudal vein plexus (CVP). The CVP development was monitored by in vivo confocal microscopy in high spatio-temporal resolution using the transgenic zebrafish model Fli1a:eGPF//Gata1:dsRed. We tracked back the formation of pillars (diameter ≤ 4 µm) and intercapillary meshes (diameter > 4 µm) and analysed their morphology and behaviour. Transluminal pillars in the CVP arose via a combination of sprouting, lumen expansion, and/or the creation of intraluminal folds, and those mechanisms were not associated directly with blood flow. The follow-up of pillars indicated that one-third of them disappeared between 28 and 48 h post fertilisation (hpf), and of the remaining ones, only 1/17 changed their cross-section area by >50%. The majority of the bigger meshes (39/62) increased their cross-section area by >50%. Plexus simplification and the establishment of hierarchy were dominated by the dynamics of intercapillary meshes, which formed mainly via sprouting angiogenesis. These meshes were observed to grow, reshape, and merge with each other. Our observations suggested an alternative view on intussusceptive angiogenesis in the CVP.
Collapse
Affiliation(s)
- Helena Röss
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (H.R.); (D.A.); (A.O.)
| | - Dea Aaldijk
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (H.R.); (D.A.); (A.O.)
| | | | - Adolfo Odriozola
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (H.R.); (D.A.); (A.O.)
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland; (H.R.); (D.A.); (A.O.)
| |
Collapse
|
13
|
Greysson-Wong J, Rode R, Ryu JR, Chan JL, Davari P, Rinker KD, Childs SJ. rasa1-related arteriovenous malformation is driven by aberrant venous signalling. Development 2023; 150:dev201820. [PMID: 37708300 DOI: 10.1242/dev.201820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
Arteriovenous malformations (AVMs) develop where abnormal endothelial signalling allows direct connections between arteries and veins. Mutations in RASA1, a Ras GTPase activating protein, lead to AVMs in humans and, as we show, in zebrafish rasa1 mutants. rasa1 mutants develop cavernous AVMs that subsume part of the dorsal aorta and multiple veins in the caudal venous plexus (CVP) - a venous vascular bed. The AVMs progressively enlarge and fill with slow-flowing blood. We show that the AVM results in both higher minimum and maximum flow velocities, resulting in increased pulsatility in the aorta and decreased pulsatility in the vein. These hemodynamic changes correlate with reduced expression of the flow-responsive transcription factor klf2a. Remodelling of the CVP is impaired with an excess of intraluminal pillars, which is a sign of incomplete intussusceptive angiogenesis. Mechanistically, we show that the AVM arises from ectopic activation of MEK/ERK in the vein of rasa1 mutants, and that cell size is also increased in the vein. Blocking MEK/ERK signalling prevents AVM initiation in mutants. Alterations in venous MEK/ERK therefore drive the initiation of rasa1 AVMs.
Collapse
Affiliation(s)
- Jasper Greysson-Wong
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
| | - Rachael Rode
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
- Department of Chemical and Petroleum Engineering, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
| | - Jae-Ryeon Ryu
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
| | - Jo Li Chan
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
| | - Paniz Davari
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
| | - Kristina D Rinker
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
- Department of Chemical and Petroleum Engineering, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
| | - Sarah J Childs
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, 3330 University Drive NW, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
14
|
Zhou X, Lai Y, Xu X, Wang Q, Sun L, Chen L, Li J, Li R, Luo D, Lin Y, Ding X. Tetrahedral framework nucleic acids inhibit pathological neovascularization and vaso-obliteration in ischaemic retinopathy via PI3K/AKT/mTOR signalling pathway. Cell Prolif 2023; 56:e13407. [PMID: 36694349 PMCID: PMC10334269 DOI: 10.1111/cpr.13407] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
This study aimed to explore the effect and the molecular mechanism of tetrahedral framework nucleic acids (tFNAs), a novel self-assembled nanomaterial with excellent biocompatibility and superior endocytosis ability, in inhibition of pathological retinal neovascularization (RNV) and more importantly, in amelioration of vaso-obliteration (VO) in ischaemic retinopathy. tFNAs were synthesized from four single-stranded DNAs (ssDNAs). Cell proliferation, wound healing and tube formation assays were performed to explore cellular angiogenic functions in vitro. The effects of tFNAs on reducing angiogenesis and inhibiting VO were explored by oxygen-induced retinopathy (OIR) model in vivo. In vitro, tFNAs were capable to enter endothelial cells (ECs), inhibit cell proliferation, tube formation and migration under hypoxic conditions. In vivo, tFNAs successfully reduce RNV and inhibit VO in OIR model via the PI3K/AKT/mTOR/S6K pathway, while vascular endothelial growth factor fusion protein, Aflibercept, could reduce RNV but not inhibit VO. This study provides a theoretical basis for the further understanding of RNV and suggests that tFNAs might be a novel promising candidate for the treatment of blind-causing RNV.
Collapse
Affiliation(s)
- Xiaodi Zhou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| | - Yanting Lai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| | - Xiaoxiao Xu
- Innovative Institute of Chinese Medicine and PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Qiong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| | - Limei Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| | - Limei Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| | - Jiajie Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Department of Maxillofacial Surgery, West China Stomatological HospitalSichuan UniversityChengduChina
| | - Rong Li
- Innovative Institute of Chinese Medicine and PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Delun Luo
- Innovative Institute of Chinese Medicine and PharmacyChengdu University of Traditional Chinese MedicineChengduChina
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Department of Maxillofacial Surgery, West China Stomatological HospitalSichuan UniversityChengduChina
| | - Xiaoyan Ding
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| |
Collapse
|
15
|
Ferre-Torres J, Noguera-Monteagudo A, Lopez-Canosa A, Romero-Arias JR, Barrio R, Castaño O, Hernandez-Machado A. Modelling of chemotactic sprouting endothelial cells through an extracellular matrix. Front Bioeng Biotechnol 2023; 11:1145550. [PMID: 37362221 PMCID: PMC10285466 DOI: 10.3389/fbioe.2023.1145550] [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: 01/17/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Sprouting angiogenesis is a core biological process critical to vascular development. Its accurate simulation, relevant to multiple facets of human health, is of broad, interdisciplinary appeal. This study presents an in-silico model replicating a microfluidic assay where endothelial cells sprout into a biomimetic extracellular matrix, specifically, a large-pore, low-concentration fibrin-based porous hydrogel, influenced by chemotactic factors. We introduce a novel approach by incorporating the extracellular matrix and chemotactic factor effects into a unified term using a single parameter, primarily focusing on modelling sprouting dynamics and morphology. This continuous model naturally describes chemotactic-induced sprouting with no need for additional rules. In addition, we extended our base model to account for matrix sensing and degradation, crucial aspects of angiogenesis. We validate our model via a hybrid in-silico experimental method, comparing the model predictions with experimental results derived from the microfluidic setup. Our results underscore the intricate relationship between the extracellular matrix structure and angiogenic sprouting, proposing a promising method for predicting the influence of the extracellular matrix on angiogenesis.
Collapse
Affiliation(s)
- Josep Ferre-Torres
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | | | - Adrian Lopez-Canosa
- Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Spain
| | - J Roberto Romero-Arias
- Institute for Research in Applied Mathematics and Systems, National Autonomous University of Mexico , Mexico City, Mexico
| | - Rafael Barrio
- Institute of Physics, National Autonomous University of Mexico, Mexico City, Mexico
| | - Oscar Castaño
- Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain
- Biomaterials for Regenerative Therapies, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona (UB), Barcelona, Spain
| | - Aurora Hernandez-Machado
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona (UB), Barcelona, Spain
| |
Collapse
|
16
|
Wälchli T, Bisschop J, Carmeliet P, Zadeh G, Monnier PP, De Bock K, Radovanovic I. Shaping the brain vasculature in development and disease in the single-cell era. Nat Rev Neurosci 2023; 24:271-298. [PMID: 36941369 PMCID: PMC10026800 DOI: 10.1038/s41583-023-00684-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2023] [Indexed: 03/23/2023]
Abstract
The CNS critically relies on the formation and proper function of its vasculature during development, adult homeostasis and disease. Angiogenesis - the formation of new blood vessels - is highly active during brain development, enters almost complete quiescence in the healthy adult brain and is reactivated in vascular-dependent brain pathologies such as brain vascular malformations and brain tumours. Despite major advances in the understanding of the cellular and molecular mechanisms driving angiogenesis in peripheral tissues, developmental signalling pathways orchestrating angiogenic processes in the healthy and the diseased CNS remain incompletely understood. Molecular signalling pathways of the 'neurovascular link' defining common mechanisms of nerve and vessel wiring have emerged as crucial regulators of peripheral vascular growth, but their relevance for angiogenesis in brain development and disease remains largely unexplored. Here we review the current knowledge of general and CNS-specific mechanisms of angiogenesis during brain development and in brain vascular malformations and brain tumours, including how key molecular signalling pathways are reactivated in vascular-dependent diseases. We also discuss how these topics can be studied in the single-cell multi-omics era.
Collapse
Affiliation(s)
- Thomas Wälchli
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland.
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada.
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada.
| | - Jeroen Bisschop
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB & Department of Oncology, KU Leuven, Leuven, Belgium
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Gelareh Zadeh
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Philippe P Monnier
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Donald K. Johnson Research Institute, Krembil Research Institute, Krembil Discovery Tower, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Science and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ivan Radovanovic
- Group of Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|
17
|
Wu C, Zhang D, Chen J. Microtubules are essential for angiogenic sprout elongation in zebrafish. J Genet Genomics 2023; 50:126-129. [PMID: 36064182 DOI: 10.1016/j.jgg.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 08/20/2022] [Accepted: 08/21/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Chuan Wu
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Da Zhang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China
| | - Jingying Chen
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing 400715, China; University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei, Chongqing 400714, China.
| |
Collapse
|
18
|
Apeldoorn C, Safaei S, Paton J, Maso Talou GD. Computational models for generating microvascular structures: Investigations beyond medical imaging resolution. WIREs Mech Dis 2023; 15:e1579. [PMID: 35880683 PMCID: PMC10077909 DOI: 10.1002/wsbm.1579] [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: 12/15/2021] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 01/31/2023]
Abstract
Angiogenesis, arteriogenesis, and pruning are revascularization processes essential to our natural vascular development and adaptation, as well as central players in the onset and development of pathologies such as tumoral growth and stroke recovery. Computational modeling allows for repeatable experimentation and exploration of these complex biological processes. In this review, we provide an introduction to the biological understanding of the vascular adaptation processes of sprouting angiogenesis, intussusceptive angiogenesis, anastomosis, pruning, and arteriogenesis, discussing some of the more significant contributions made to the computational modeling of these processes. Each computational model represents a theoretical framework for how biology functions, and with rises in computing power and study of the problem these frameworks become more accurate and complete. We highlight physiological, pathological, and technological applications that can be benefit from the advances performed by these models, and we also identify which elements of the biology are underexplored in the current state-of-the-art computational models. This article is categorized under: Cancer > Computational Models Cardiovascular Diseases > Computational Models.
Collapse
Affiliation(s)
- Cameron Apeldoorn
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Soroush Safaei
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Julian Paton
- Cardiovascular Autonomic Research Cluster, Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Gonzalo D Maso Talou
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
19
|
Yadunandanan Nair N, Samuel V, Ramesh L, Marib A, David DT, Sundararaman A. Actin cytoskeleton in angiogenesis. Biol Open 2022; 11:bio058899. [PMID: 36444960 PMCID: PMC9729668 DOI: 10.1242/bio.058899] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
Actin, one of the most abundant intracellular proteins in mammalian cells, is a critical regulator of cell shape and polarity, migration, cell division, and transcriptional response. Angiogenesis, or the formation of new blood vessels in the body is a well-coordinated multi-step process. Endothelial cells lining the blood vessels acquire several new properties such as front-rear polarity, invasiveness, rapid proliferation and motility during angiogenesis. This is achieved by changes in the regulation of the actin cytoskeleton. Actin remodelling underlies the switch between the quiescent and angiogenic state of the endothelium. Actin forms endothelium-specific structures that support uniquely endothelial functions. Actin regulators at endothelial cell-cell junctions maintain the integrity of the blood-tissue barrier while permitting trans-endothelial leukocyte migration. This review focuses on endothelial actin structures and less-recognised actin-mediated endothelial functions. Readers are referred to other recent reviews for the well-recognised roles of actin in endothelial motility, barrier functions and leukocyte transmigration. Actin generates forces that are transmitted to the extracellular matrix resulting in vascular matrix remodelling. In this review, we attempt to synthesize our current understanding of the roles of actin in vascular morphogenesis. We speculate on the vascular bed specific differences in endothelial actin regulation and its role in the vast heterogeneity in endothelial morphology and function across the various tissues of our body.
Collapse
Affiliation(s)
- Nidhi Yadunandanan Nair
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Victor Samuel
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Lariza Ramesh
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Areeba Marib
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Deena T. David
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| | - Ananthalakshmy Sundararaman
- Cardiovascular Diseases and Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India695014
| |
Collapse
|
20
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
21
|
Chau TCY, Keyser MS, Da Silva JA, Morris EK, Yordanov TE, Duscyz KP, Paterson S, Yap AS, Hogan BM, Lagendijk AK. Dynamically regulated focal adhesions coordinate endothelial cell remodelling in developing vasculature. Development 2022; 149:285926. [PMID: 36314606 DOI: 10.1242/dev.200454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 10/20/2022] [Indexed: 12/13/2022]
Abstract
The assembly of a mature vascular network involves coordinated endothelial cell (EC) shape changes, including the process of EC elongation. How EC elongation is dynamically regulated in vivo is not fully understood. Here, we have generated a zebrafish mutant that is deficient for the integrin adaptor protein Talin 1 (Tln1). Using a new focal adhesion (FA) marker line expressing endothelial Vinculinb-eGFP, we demonstrate that EC FAs function dynamically and are lost in our tln1 mutants, allowing us to uncouple the primary roles of FAs in EC morphogenesis from the secondary effects that occur due to systemic vessel failure or loss of blood flow. Tln1 loss led to compromised F-actin rearrangements, perturbed EC elongation and disrupted cell-cell junction linearisation in vessel remodelling. Finally, chemical induction of actin polymerisation restored actin dynamics and EC elongation during vascular morphogenesis. Together, we identify that FAs are essential for EC elongation and junction linearisation in flow-pressured vessels and that they influence actin polymerisation in cellular morphogenesis. These observations can explain the severely compromised vessel beds and vascular leakage observed in mutant models that lack integrin signalling. This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Tevin C Y Chau
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Mikaela S Keyser
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Jason A Da Silva
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Elysse K Morris
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Teodor E Yordanov
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kinga P Duscyz
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Scott Paterson
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre and The PeterMac Callum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Alpha S Yap
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Benjamin M Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre and The PeterMac Callum Department of Oncology, The University of Melbourne, Melbourne, Victoria 3000, Australia.,Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anne Karine Lagendijk
- Division of Cell and Developmental Biology, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
22
|
Abstract
Pten is one of the most frequently mutated tumour suppressor gene in cancer. PTEN is generally altered in invasive cancers such as glioblastomas, but its function in collective cell migration and invasion is not fully characterised. Herein, we report that the loss of PTEN increases cell speed during collective migration of non-tumourous cells both in vitro and in vivo. We further show that loss of PTEN promotes LKB1-dependent phosphorylation and activation of the major metabolic regulator AMPK. In turn AMPK increases VASP phosphorylation, reduces VASP localisation at cell-cell junctions and decreases the interjunctional transverse actin arcs at the leading front, provoking a weakening of cell-cell contacts and increasing migration speed. Targeting AMPK activity not only slows down PTEN-depleted cells, it also limits PTEN-null glioblastoma cell invasion, opening new opportunities to treat glioblastoma lethal invasiveness. Pten is a tumour suppressor gene that is associated with highly invasive cancers such as glioblastoma. Here the authors show that PTEN loss results in increased migratory behaviour, which can be countered by targeting AMPK activity.
Collapse
|
23
|
Panara V, Monteiro R, Koltowska K. Epigenetic Regulation of Endothelial Cell Lineages During Zebrafish Development-New Insights From Technical Advances. Front Cell Dev Biol 2022; 10:891538. [PMID: 35615697 PMCID: PMC9125237 DOI: 10.3389/fcell.2022.891538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/10/2022] [Indexed: 01/09/2023] Open
Abstract
Epigenetic regulation is integral in orchestrating the spatiotemporal regulation of gene expression which underlies tissue development. The emergence of new tools to assess genome-wide epigenetic modifications has enabled significant advances in the field of vascular biology in zebrafish. Zebrafish represents a powerful model to investigate the activity of cis-regulatory elements in vivo by combining technologies such as ATAC-seq, ChIP-seq and CUT&Tag with the generation of transgenic lines and live imaging to validate the activity of these regulatory elements. Recently, this approach led to the identification and characterization of key enhancers of important vascular genes, such as gata2a, notch1b and dll4. In this review we will discuss how the latest technologies in epigenetics are being used in the zebrafish to determine chromatin states and assess the function of the cis-regulatory sequences that shape the zebrafish vascular network.
Collapse
Affiliation(s)
- Virginia Panara
- Immunology Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rui Monteiro
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Birmingham Centre of Genome Biology, University of Birmingham, Birmingham, United Kingdom
| | | |
Collapse
|
24
|
Kotini MP, van der Stoel MM, Yin J, Han MK, Kirchmaier B, de Rooij J, Affolter M, Huveneers S, Belting HG. Vinculin controls endothelial cell junction dynamics during vascular lumen formation. Cell Rep 2022; 39:110658. [PMID: 35417696 DOI: 10.1016/j.celrep.2022.110658] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 02/01/2022] [Accepted: 03/20/2022] [Indexed: 12/20/2022] Open
Abstract
Blood vessel morphogenesis is driven by coordinated endothelial cell behaviors. Active remodeling of cell-cell junctions promotes cellular plasticity while preserving vascular integrity. Here, we analyze the dynamics of endothelial adherens junctions during lumen formation in angiogenic sprouts in vivo. Live imaging in zebrafish reveals that lumen expansion is accompanied by the formation of transient finger-shaped junctions. Junctional fingers are positively regulated by blood pressure, whereas flow inhibition prevents their formation. Using fluorescent reporters, we show that junctional fingers contain the mechanotransduction protein vinculin. Furthermore, genetic deletion of vinculin prevents finger formation, a junctional defect that could be rescued by transient endothelial expression of vinculin. Our findings suggest a mechanism whereby lumen expansion leads to an increase in junctional tension, triggering recruitment of vinculin and formation of junctional fingers. We propose that endothelial cells employ force-dependent junctional remodeling to counteract external forces in order to maintain vascular integrity during sprouting angiogenesis.
Collapse
Affiliation(s)
- Maria P Kotini
- Biozentrum der Universität Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Miesje M van der Stoel
- Amsterdam UMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Jianmin Yin
- Biozentrum der Universität Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Mitchell K Han
- Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Stratenum 3.231, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Bettina Kirchmaier
- Institute for Cell Biology and Neuroscience, Biologicum/Campus Riedberg, Goethe-Universität Frankfurt Am Main, 60438 Frankfurt Am Main, Germany
| | - Johan de Rooij
- Department of Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Stratenum 3.231, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Markus Affolter
- Biozentrum der Universität Basel, Spitalstrasse 41, 4056 Basel, Switzerland
| | - Stephan Huveneers
- Amsterdam UMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| | - Heinz-Georg Belting
- Biozentrum der Universität Basel, Spitalstrasse 41, 4056 Basel, Switzerland.
| |
Collapse
|
25
|
Coxam B, Collins RT, Hußmann M, Huisman Y, Meier K, Jung S, Bartels-Klein E, Szymborska A, Finotto L, Helker CSM, Stainier DYR, Schulte-Merker S, Gerhardt H. Svep1 stabilises developmental vascular anastomosis in reduced flow conditions. Development 2022; 149:274823. [PMID: 35312765 PMCID: PMC8977097 DOI: 10.1242/dev.199858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 02/14/2022] [Indexed: 11/24/2022]
Abstract
Molecular mechanisms controlling the formation, stabilisation and maintenance of blood vessel connections remain poorly defined. Here, we identify blood flow and the large extracellular protein Svep1 as co-modulators of vessel anastomosis during developmental angiogenesis in zebrafish embryos. Both loss of Svep1 and blood flow reduction contribute to defective anastomosis of intersegmental vessels. The reduced formation and lumenisation of the dorsal longitudinal anastomotic vessel (DLAV) is associated with a compensatory increase in Vegfa/Vegfr pERK signalling, concomittant expansion of apelin-positive tip cells, but reduced expression of klf2a. Experimentally, further increasing Vegfa/Vegfr signalling can rescue the DLAV formation and lumenisation defects, whereas its inhibition dramatically exacerbates the loss of connectivity. Mechanistically, our results suggest that flow and Svep1 co-regulate the stabilisation of vascular connections, in part by modulating the Vegfa/Vegfr signalling pathway. Summary: Blood flow and the large extracellular matrix protein Svep1 jointly regulate vessel anastomosis during developmental angiogenesis in zebrafish embryos partly by modulating the Vegfa/Vegfr signalling pathway.
Collapse
Affiliation(s)
- Baptiste Coxam
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, Berlin 13125, Germany
- DZHK (German Center for Cardiovascular Research), Berlin 10785, Germany
| | - Russell T. Collins
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, Berlin 13125, Germany
- DZHK (German Center for Cardiovascular Research), Berlin 10785, Germany
| | - Melina Hußmann
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Mendelstraße 7, 48149 Münster, Germany
| | - Yvonne Huisman
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Mendelstraße 7, 48149 Münster, Germany
| | - Katja Meier
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, Berlin 13125, Germany
| | - Simone Jung
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, Berlin 13125, Germany
| | - Eireen Bartels-Klein
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, Berlin 13125, Germany
| | - Anna Szymborska
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, Berlin 13125, Germany
| | - Lise Finotto
- Vascular Patterning Laboratory, Center for Cancer Biology, VIB, Leuven 3000, Belgium
- Vascular Patterning Laboratory, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Christian S. M. Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, WWU Münster, Mendelstraße 7, 48149 Münster, Germany
| | - Holger Gerhardt
- Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, Berlin 13125, Germany
- DZHK (German Center for Cardiovascular Research), Berlin 10785, Germany
- Berlin Institute of Health (BIH), Berlin 10178, Germany
| |
Collapse
|
26
|
Puebla M, Tapia PJ, Espinoza H. Key Role of Astrocytes in Postnatal Brain and Retinal Angiogenesis. Int J Mol Sci 2022; 23:ijms23052646. [PMID: 35269788 PMCID: PMC8910249 DOI: 10.3390/ijms23052646] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 01/27/2023] Open
Abstract
Angiogenesis is a key process in various physiological and pathological conditions in the nervous system and in the retina during postnatal life. Although an increasing number of studies have addressed the role of endothelial cells in this event, the astrocytes contribution in angiogenesis has received less attention. This review is focused on the role of astrocytes as a scaffold and in the stabilization of the new blood vessels, through different molecules release, which can modulate the angiogenesis process in the brain and in the retina. Further, differences in the astrocytes phenotype are addressed in glioblastoma, one of the most devastating types of brain cancer, in order to provide potential targets involved in the cross signaling between endothelial cells, astrocytes and glioma cells, that mediate tumor progression and pathological angiogenesis. Given the relevance of astrocytes in angiogenesis in physiological and pathological conditions, future studies are required to better understand the interrelation between endothelial and astrocyte signaling pathways during this process.
Collapse
Affiliation(s)
- Mariela Puebla
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina-Clínica Alemana, Universidad del Desarrollo, Av. Plaza 680, Las Condes, Santiago 7550000, Chile;
| | - Pablo J. Tapia
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Av. Lota 2465, Providencia, Santiago 7500000, Chile;
- Facultad de Medicina Veterinaria y Agronomía, Universidad de las Américas, Av. República 71, Santiago 8320000, Chile
| | - Hilda Espinoza
- Facultad de Ciencias de la Salud, Universidad del Alba, Av. Ejército Libertador 171, Santiago 8320000, Chile
- Correspondence:
| |
Collapse
|
27
|
Dynamics of Endothelial Engagement and Filopodia Formation in Complex 3D Microscaffolds. Int J Mol Sci 2022; 23:ijms23052415. [PMID: 35269558 PMCID: PMC8910162 DOI: 10.3390/ijms23052415] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 11/28/2022] Open
Abstract
The understanding of endothelium–extracellular matrix interactions during the initiation of new blood vessels is of great medical importance; however, the mechanobiological principles governing endothelial protrusive behaviours in 3D microtopographies remain imperfectly understood. In blood capillaries submitted to angiogenic factors (such as vascular endothelial growth factor, VEGF), endothelial cells can transiently transdifferentiate in filopodia-rich cells, named tip cells, from which angiogenesis processes are locally initiated. This protrusive state based on filopodia dynamics contrasts with the lamellipodia-based endothelial cell migration on 2D substrates. Using two-photon polymerization, we generated 3D microstructures triggering endothelial phenotypes evocative of tip cell behaviour. Hexagonal lattices on pillars (“open”), but not “closed” hexagonal lattices, induced engagement from the endothelial monolayer with the generation of numerous filopodia. The development of image analysis tools for filopodia tracking allowed to probe the influence of the microtopography (pore size, regular vs. elongated structures, role of the pillars) on orientations, engagement and filopodia dynamics, and to identify MLCK (myosin light-chain kinase) as a key player for filopodia-based protrusive mode. Importantly, these events occurred independently of VEGF treatment, suggesting that the observed phenotype was induced through microtopography. These microstructures are proposed as a model research tool for understanding endothelial cell behaviour in 3D fibrillary networks.
Collapse
|
28
|
Mentor S, Makhathini KB, Fisher D. The Role of Cytoskeletal Proteins in the Formation of a Functional In Vitro Blood-Brain Barrier Model. Int J Mol Sci 2022; 23:ijms23020742. [PMID: 35054928 PMCID: PMC8775705 DOI: 10.3390/ijms23020742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/27/2021] [Accepted: 01/07/2022] [Indexed: 02/05/2023] Open
Abstract
The brain capillary endothelium is highly regulatory, maintaining the chemical stability of the brain’s microenvironment. The role of cytoskeletal proteins in tethering nanotubules (TENTs) during barrier-genesis was investigated using the established immortalized mouse brain endothelial cell line (bEnd5) as an in vitro blood-brain barrier (BBB) model. The morphology of bEnd5 cells was evaluated using both high-resolution scanning electron microscopy and immunofluorescence to evaluate treatment with depolymerizing agents Cytochalasin D for F-actin filaments and Nocodazole for α-tubulin microtubules. The effects of the depolymerizing agents were investigated on bEnd5 monolayer permeability by measuring the transendothelial electrical resistance (TEER). The data endorsed that during barrier-genesis, F-actin and α-tubulin play a cytoarchitectural role in providing both cell shape dynamics and cytoskeletal structure to TENTs forming across the paracellular space to provide cell-cell engagement. Western blot analysis of the treatments suggested a reduced expression of both proteins, coinciding with a reduction in the rates of cellular proliferation and decreased TEER. The findings endorsed that TENTs provide alignment of the paracellular (PC) spaces and tight junction (TJ) zones to occlude bEnd5 PC spaces. The identification of specific cytoskeletal structures in TENTs endorsed the postulate of their indispensable role in barrier-genesis and the maintenance of regulatory permeability across the BBB.
Collapse
Affiliation(s)
- Shireen Mentor
- Neurobiology Research Group, Department of Medical Biosciences, University of the Western Cape, Cape Town 7535, South Africa; (S.M.); (K.B.M.)
| | - Khayelihle Brian Makhathini
- Neurobiology Research Group, Department of Medical Biosciences, University of the Western Cape, Cape Town 7535, South Africa; (S.M.); (K.B.M.)
| | - David Fisher
- Neurobiology Research Group, Department of Medical Biosciences, University of the Western Cape, Cape Town 7535, South Africa; (S.M.); (K.B.M.)
- School of Health Professions, University of Missouri, Columbia, MO 65211, USA
- Correspondence: ; Tel.: +27-21-959-2185
| |
Collapse
|
29
|
Rosa A, Giese W, Meier K, Alt S, Klaus-Bergmann A, Edgar LT, Bartels E, Collins R, Szymborska A, Coxam B, Bernabeu MO, Gerhardt H. Wasp controls oriented migration of endothelial cells to achieve functional vascular patterning. Development 2021; 149:273808. [PMID: 34931661 PMCID: PMC8918813 DOI: 10.1242/dev.200195] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/10/2021] [Indexed: 11/21/2022]
Abstract
Endothelial cell migration and proliferation are essential for the establishment of a hierarchical organization of blood vessels and optimal distribution of blood. However, how these cellular processes are quantitatively coordinated to drive vascular network morphogenesis remains unknown. Here, using the zebrafish vasculature as a model system, we demonstrate that the balanced distribution of endothelial cells, as well as the resulting regularity of vessel calibre, is a result of cell migration from veins towards arteries and cell proliferation in veins. We identify the Wiskott-Aldrich Syndrome protein (WASp) as an important molecular regulator of this process and show that loss of coordinated migration from veins to arteries upon wasb depletion results in aberrant vessel morphology and the formation of persistent arteriovenous shunts. We demonstrate that WASp achieves its function through the coordination of junctional actin assembly and PECAM1 recruitment and provide evidence that this is conserved in humans. Overall, we demonstrate that functional vascular patterning in the zebrafish trunk is established through differential cell migration regulated by junctional actin, and that interruption of differential migration may represent a pathomechanism in vascular malformations. Summary: Regular diameter of developing veins and arteries in the zebrafish trunk is controlled by differential endothelial cell proliferation and WASp-driven directed cell migration.
Collapse
Affiliation(s)
- André Rosa
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Wolfgang Giese
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Katja Meier
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Silvanus Alt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Alexandra Klaus-Bergmann
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Lowell T Edgar
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Eireen Bartels
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Russell Collins
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Anna Szymborska
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Baptiste Coxam
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Miguel O Bernabeu
- Usher Institute, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK.,The Bayes Centre, The University of Edinburgh, Edinburgh, United Kingdom. 5 Berlin Institute of Health (BIH), Berlin, Germany
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
30
|
Abstract
Cerebral ischemic injury may lead to a series of serious brain diseases, death or different degrees of disability. Hypoxia-inducible factor-1α (HIF-1α) is an oxygen-sensitive transcription factor, which mediates the adaptive metabolic response to hypoxia and serves a key role in cerebral ischemia. HIF-1α is the main molecule that responds to hypoxia. HIF-1α serves an important role in the development of cerebral ischemia by participating in numerous processes, including metabolism, proliferation and angiogenesis. The present review focuses on the endogenous protective mechanism of cerebral ischemia and elaborates on the role of HIF-1α in cerebral ischemia. In addition, it focuses on cerebral ischemia interventions that act on the HIF-1α target, including biological factors, non-coding RNA, hypoxic-ischemic preconditioning and drugs, and expands upon the measures to strengthen the endogenous compensatory response to support HIF-1α as a therapeutic target, thus providing novel suggestions for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Peiliang Dong
- Institute of Traditional Chinese Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Qingna Li
- College of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Hua Han
- College of Pharmacy, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
31
|
Xu R, Du S. Overexpression of Lifeact-GFP Disrupts F-Actin Organization in Cardiomyocytes and Impairs Cardiac Function. Front Cell Dev Biol 2021; 9:746818. [PMID: 34765602 PMCID: PMC8576398 DOI: 10.3389/fcell.2021.746818] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 10/07/2021] [Indexed: 11/28/2022] Open
Abstract
Lifeact-GFP is a frequently used molecular probe to study F-actin structure and dynamic assembly in living cells. In this study, we generated transgenic zebrafish models expressing Lifeact-GFP specifically in cardiac muscles to investigate the effect of Lifeact-GFP on heart development and its application to study cardiomyopathy. The data showed that transgenic zebrafish with low to moderate levels of Lifeact-GFP expression could be used as a good model to study contractile dynamics of actin filaments in cardiac muscles in vivo. Using this model, we demonstrated that loss of Smyd1b, a lysine methyltransferase, disrupted F-actin filament organization in cardiomyocytes of zebrafish embryos. Our studies, however, also demonstrated that strong Lifeact-GFP expression in cardiomyocytes was detrimental to actin filament organization in cardiomyocytes that led to pericardial edema and early embryonic lethality of zebrafish embryos. Collectively, these data suggest that although Lifeact-GFP is a good probe for visualizing F-actin dynamics, transgenic models need to be carefully evaluated to avoid artifacts induced by Lifeact-GFP overexpression.
Collapse
Affiliation(s)
| | - Shaojun Du
- Department of Biochemistry and Molecular Biology, Institute of Marine and Environmental Technology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
32
|
Watterston C, Halabi R, McFarlane S, Childs SJ. Endothelial Semaphorin 3fb regulates Vegf pathway-mediated angiogenic sprouting. PLoS Genet 2021; 17:e1009769. [PMID: 34424892 PMCID: PMC8412281 DOI: 10.1371/journal.pgen.1009769] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 09/02/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Vessel growth integrates diverse extrinsic signals with intrinsic signaling cascades to coordinate cell migration and sprouting morphogenesis. The pro-angiogenic effects of Vascular Endothelial Growth Factor (VEGF) are carefully controlled during sprouting to generate an efficiently patterned vascular network. We identify crosstalk between VEGF signaling and that of the secreted ligand Semaphorin 3fb (Sema3fb), one of two zebrafish paralogs of mammalian Sema3F. The sema3fb gene is expressed by endothelial cells in actively sprouting vessels. Loss of sema3fb results in abnormally wide and stunted intersegmental vessel artery sprouts. Although the sprouts initiate at the correct developmental time, they have a reduced migration speed. These sprouts have persistent filopodia and abnormally spaced nuclei suggesting dysregulated control of actin assembly. sema3fb mutants show simultaneously higher expression of pro-angiogenic (VEGF receptor 2 (vegfr2) and delta-like 4 (dll4)) and anti-angiogenic (soluble VEGF receptor 1 (svegfr1)/ soluble Fms Related Receptor Tyrosine Kinase 1 (sflt1)) pathway components. We show increased phospho-ERK staining in migrating angioblasts, consistent with enhanced Vegf activity. Reducing Vegfr2 kinase activity in sema3fb mutants rescues angiogenic sprouting. Our data suggest that Sema3fb plays a critical role in promoting endothelial sprouting through modulating the VEGF signaling pathway, acting as an autocrine cue that modulates intrinsic growth factor signaling.
Collapse
Affiliation(s)
- Charlene Watterston
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Rami Halabi
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Sarah McFarlane
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Canada
| | - Sarah J. Childs
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
33
|
The blood flow-klf6a-tagln2 axis drives vessel pruning in zebrafish by regulating endothelial cell rearrangement and actin cytoskeleton dynamics. PLoS Genet 2021; 17:e1009690. [PMID: 34319989 PMCID: PMC8318303 DOI: 10.1371/journal.pgen.1009690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
Recent studies have focused on capillary pruning in various organs and species. However, the way in which large-diameter vessels are pruned remains unclear. Here we show that pruning of the zebrafish caudal vein (CV) from ventral capillaries of the CV plexus in different transgenic embryos is driven by endothelial cell (EC) rearrangement, which involves EC nucleus migration, junction remodeling, and actin cytoskeleton remodeling. Further observation reveals a growing difference in blood flow velocity between the two vessels in CV pruning in zebrafish embryos. With this model, we identify the critical role of Kruppel-like factor 6a (klf6a) in CV pruning. Disruption of klf6a functioning impairs CV pruning in zebrafish. klf6a is required for EC nucleus migration, junction remodeling, and actin cytoskeleton dynamics in zebrafish embryos. Moreover, actin-related protein transgelin 2 (tagln2) is a direct downstream target of klf6a in CV pruning in zebrafish embryos. Together these results demonstrate that the klf6a-tagln2 axis regulates CV pruning by promoting EC rearrangement. Vascular remodeling is critical for vascular physiology and pathology. The primitive vascular plexus formed by angiogenesis, subsequently undergoes extensive vascular remodeling to establish a functionally and hierarchically branched network of blood vessels. Vascular remodeling mainly consists of vessel pruning and fusion. Endothelial cell rearrangement plays an essential role in vessel pruning, which involves endothelial cell migration and polarity. Dysfunction of flow-induced vascular remodeling will cause arteriovenous malformation and impair reperfusion of ischemia stroke. In this study, we show that the large-diameter vessel of the caudal vein is pruned from ventral capillaries of the caudal vein plexus in zebrafish embryos. With this model, we observe a growing difference in blood flow velocity between two branches in vessel pruning. We identify that the klf6a-tagln2 axis regulates CV pruning by promoting endothelial cell rearrangement and junction remodeling. Our results suggest that the caudal vein formation is an ideal model for screening the potential genes involved in vascular remodeling-related disease.
Collapse
|
34
|
Phng LK, Belting HG. Endothelial cell mechanics and blood flow forces in vascular morphogenesis. Semin Cell Dev Biol 2021; 120:32-43. [PMID: 34154883 DOI: 10.1016/j.semcdb.2021.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 12/21/2022]
Abstract
The vertebrate cardiovascular system is made up by a hierarchically structured network of highly specialised blood vessels. This network emerges during early embryogenesis and evolves in size and complexity concomitant with embryonic growth and organ formation. Underlying this plasticity are actin-driven endothelial cell behaviours, which allow endothelial cells to change their shape and move within the vascular network. In this review, we discuss the cellular and molecular mechanisms involved in vascular network formation and how these intrinsic mechanisms are influenced by haemodynamic forces provided by pressurized blood flow. While most of this review focusses on in vivo evidence from zebrafish embryos, we also mention complementary findings obtained in other experimental systems.
Collapse
Affiliation(s)
- Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan.
| | - Heinz-Georg Belting
- Department of Cell Biology, Biozentrum, University of Basel, Basel 4056, Switzerland.
| |
Collapse
|
35
|
Cabrerizo-Granados D, Peña R, Palacios L, Carrillo-Bosch L, Lloreta-Trull J, Comerma L, Iglesias M, de Herreros AG. Snail1 expression in endothelial cells controls growth, angiogenesis and differentiation of breast tumors. Theranostics 2021; 11:7671-7684. [PMID: 34335957 PMCID: PMC8315050 DOI: 10.7150/thno.61881] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/26/2021] [Indexed: 12/26/2022] Open
Abstract
Snail1 is a transcriptional factor required for epithelial to mesenchymal transition and activation of cancer-associated fibroblasts (CAF). Apart from that, tumor endothelial cells also express Snail1. Here, we have unraveled the role of Snail1 in this tissue in a tumorigenic context. Methods: We generated transgenic mice with an endothelial-specific and inducible Snail1 depletion. This murine line was crossed with MMTV-PyMT mice that develop mammary gland tumors and the consequence of Snail1 depletion in the endothelium were investigated. We also interfere Snail1 expression in cultured endothelial cells. Results: Specific Snail1 depletion in the endothelium of adult mice does not promote an overt phenotype; however, it delays the formation of mammary gland tumors in MMTV-PyMT mice. These effects are associated to the inability of Snail1-deficient endothelial cells to undergo angiogenesis and to enhance CAF activation in a paracrine manner. Moreover, tumors generated in mice with endothelium-specific Snail1 depletion are less advanced and show a papillary phenotype. Similar changes on onset and tumor morphology are observed by pretreatment of MMTV-PyMT mice with the angiogenic inhibitor Bevacizumab. Human breast papillary carcinomas exhibit a lower angiogenesis and present lower staining of Snail1, both in endothelial and stromal cells, compared with other breast neoplasms. Furthermore, human breast tumors datasets show a strong correlation between Snail1 expression and high angiogenesis. Conclusion: These findings show a novel role for Snail1 in endothelial cell activation and demonstrate that these cells impact not only on angiogenesis, but also on tumor onset and phenotype.
Collapse
|
36
|
Zhang T, Xu Z, Wen L, Lei D, Li S, Wang J, Huang J, Wang N, Durkan C, Liao X, Wang G. Cadmium-induced dysfunction of the blood-brain barrier depends on ROS-mediated inhibition of PTPase activity in zebrafish. JOURNAL OF HAZARDOUS MATERIALS 2021; 412:125198. [PMID: 33550130 DOI: 10.1016/j.jhazmat.2021.125198] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/04/2021] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
Increasing evidence has demonstrated that cadmium accumulation in the blood increases the risk of neurological diseases. However, how cadmium breaks through the blood-brain barrier (BBB) and is transferred from the blood circulation into the central nervous system is still unclear. In this study, we examined the toxic effect of cadmium chloride (CdCl2) on the development and function of BBB in zebrafish. CdCl2 exposure induced cerebral hemorrhage, increased BBB permeability and promoted abnormal vascular formation by promoting VEGF production in zebrafish brain. Furthermore, in vivo and in vitro experiments showed that CdCl2 altered cell-cell junctional morphology by disrupting the proper localization of VE-cadherin and ZO-1. The potential mechanism involved in the inhibition of protein tyrosine phosphatase (PTPase) mediated by cadmium-induced ROS was confirmed with diphenylene iodonium (DPI), a ROS production inhibitor. Together, these data indicate that BBB is a critical target of cadmium toxicity and provide in vivo etiological evidence of cadmium-induced neurovascular disease in a zebrafish BBB model.
Collapse
Affiliation(s)
- Tao Zhang
- 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; Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Zichen Xu
- 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.
| | - 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.
| | - Daoxi Lei
- 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.
| | - Shuyu Li
- 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.
| | - Jinxuan 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.
| | - Jinxia Huang
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB30FF, UK.
| | - Colm Durkan
- The Nanoscience Centre, University of Cambridge, 11 JJ Thomson Avenue, Cambridge CB30FF, UK.
| | - Xiaoling Liao
- Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing 401331, 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.
| |
Collapse
|
37
|
Taberner L, Bañón A, Alsina B. Sensory Neuroblast Quiescence Depends on Vascular Cytoneme Contacts and Sensory Neuronal Differentiation Requires Initiation of Blood Flow. Cell Rep 2021; 32:107903. [PMID: 32668260 DOI: 10.1016/j.celrep.2020.107903] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 04/02/2020] [Accepted: 06/23/2020] [Indexed: 02/08/2023] Open
Abstract
In many organs, stem cell function depends on communication with their niche partners. Cranial sensory neurons develop in close proximity to blood vessels; however, whether vasculature is an integral component of their niches is yet unknown. Here, two separate roles for vasculature in cranial sensory neurogenesis in zebrafish are uncovered. The first involves precise spatiotemporal endothelial-neuroblast cytoneme contacts and Dll4-Notch signaling to restrain neuroblast proliferation. The second, instead, requires blood flow to trigger a transcriptional response that modifies neuroblast metabolic status and induces sensory neuron differentiation. In contrast, no role of sensory neurogenesis in vascular development is found, suggesting unidirectional signaling from vasculature to sensory neuroblasts. Altogether, we demonstrate that the cranial vasculature constitutes a niche component of the sensory ganglia that regulates the pace of their growth and differentiation dynamics.
Collapse
Affiliation(s)
- Laura Taberner
- Developmental Biology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Aitor Bañón
- Developmental Biology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Berta Alsina
- Developmental Biology Unit, Department of Experimental and Health Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica de Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain.
| |
Collapse
|
38
|
Abstract
In this report, we describe how endothelial cells, the cells lining the interior of blood vessels, invade into tissues to form new vessels through sprouting angiogenesis. We found that endothelial cells use a specific lamellipodia-related membrane protrusion for invasion, which we termed dactylopodia. These protrusions have a special morphology, originate from filopodia, are linked to membrane-ruffling activity, and are specialized in invading into avascular extracellular matrix. Our work lays the foundations for drug discovery targeting sprouting angiogenesis. Sprouting angiogenesis is fundamental for development and contributes to cancer, diabetic retinopathy, and cardiovascular diseases. Sprouting angiogenesis depends on the invasive properties of endothelial tip cells. However, there is very limited knowledge on how tip cells invade into tissues. Here, we show that endothelial tip cells use dactylopodia as the main cellular protrusion for invasion into nonvascular extracellular matrix. We show that dactylopodia and filopodia protrusions are balanced by myosin IIA (NMIIA) and actin-related protein 2/3 (Arp2/3) activity. Endothelial cell-autonomous ablation of NMIIA promotes excessive dactylopodia formation in detriment of filopodia. Conversely, endothelial cell-autonomous ablation of Arp2/3 prevents dactylopodia development and leads to excessive filopodia formation. We further show that NMIIA inhibits Rac1-dependent activation of Arp2/3 by regulating the maturation state of focal adhesions. Our discoveries establish a comprehensive model of how endothelial tip cells regulate its protrusive activity and will pave the way toward strategies to block invasive tip cells during sprouting angiogenesis.
Collapse
|
39
|
Zakirov B, Charalambous G, Thuret R, Aspalter IM, Van-Vuuren K, Mead T, Harrington K, Regan ER, Herbert SP, Bentley K. Active perception during angiogenesis: filopodia speed up Notch selection of tip cells in silico and in vivo. Philos Trans R Soc Lond B Biol Sci 2021; 376:20190753. [PMID: 33550953 PMCID: PMC7934951 DOI: 10.1098/rstb.2019.0753] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2020] [Indexed: 12/19/2022] Open
Abstract
How do cells make efficient collective decisions during tissue morphogenesis? Humans and other organisms use feedback between movement and sensing known as 'sensorimotor coordination' or 'active perception' to inform behaviour, but active perception has not before been investigated at a cellular level within organs. Here we provide the first proof of concept in silico/in vivo study demonstrating that filopodia (actin-rich, dynamic, finger-like cell membrane protrusions) play an unexpected role in speeding up collective endothelial decisions during the time-constrained process of 'tip cell' selection during blood vessel formation (angiogenesis). We first validate simulation predictions in vivo with live imaging of zebrafish intersegmental vessel growth. Further simulation studies then indicate the effect is due to the coupled positive feedback between movement and sensing on filopodia conferring a bistable switch-like property to Notch lateral inhibition, ensuring tip selection is a rapid and robust process. We then employ measures from computational neuroscience to assess whether filopodia function as a primitive (basal) form of active perception and find evidence in support. By viewing cell behaviour through the 'basal cognitive lens' we acquire a fresh perspective on the tip cell selection process, revealing a hidden, yet vital time-keeping role for filopodia. Finally, we discuss a myriad of new and exciting research directions stemming from our conceptual approach to interpreting cell behaviour. This article is part of the theme issue 'Basal cognition: multicellularity, neurons and the cognitive lens'.
Collapse
Affiliation(s)
- Bahti Zakirov
- Cellular Adaptive Behaviour Lab, Francis Crick Institute, London, NW1 1AT, UK
- Department of Informatics, King's College London, London, UK
| | - Georgios Charalambous
- Division of Developmental Biology and Medicine, University of Manchester, Manchester, UK
| | - Raphael Thuret
- Division of Developmental Biology and Medicine, University of Manchester, Manchester, UK
| | - Irene M. Aspalter
- Cellular Adaptive Behaviour Lab, Francis Crick Institute, London, NW1 1AT, UK
| | - Kelvin Van-Vuuren
- Cellular Adaptive Behaviour Lab, Francis Crick Institute, London, NW1 1AT, UK
| | - Thomas Mead
- Cellular Adaptive Behaviour Lab, Francis Crick Institute, London, NW1 1AT, UK
- Department of Informatics, King's College London, London, UK
| | - Kyle Harrington
- Virtual Technology and Design, University of Idaho, Moscow, ID, USA
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Erzsébet Ravasz Regan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Department of Pathology, Harvard Medical School, Boston, MA, USA
- Department of Biology, The College of Wooster, Wooster, OH, USA
| | - Shane Paul Herbert
- Division of Developmental Biology and Medicine, University of Manchester, Manchester, UK
| | - Katie Bentley
- Cellular Adaptive Behaviour Lab, Francis Crick Institute, London, NW1 1AT, UK
- Department of Informatics, King's College London, London, UK
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Department of Pathology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Yin J, Heutschi D, Belting HG, Affolter M. Building the complex architectures of vascular networks: Where to branch, where to connect and where to remodel? Curr Top Dev Biol 2021; 143:281-297. [PMID: 33820624 DOI: 10.1016/bs.ctdb.2021.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The cardiovascular system is the first organ to become functional during vertebrate embryogenesis and is responsible for the distribution of oxygen and nutrients to all cells of the body. The cardiovascular system constitutes a circulatory loop in which blood flows from the heart through arteries into the microvasculature and back through veins to the heart. The vasculature is characterized by the heterogeneity of blood vessels with respect to size, cellular architecture and function, including both larger vessels that are found at defined positions within the body and smaller vessels or vascular beds that are organized in a less stereotyped manner. Recent studies have shed light on how the vascular tree is formed and how the interconnection of all branches is elaborated and maintained. In contrast to many other branched organs such as the lung or the kidney, vessel connection (also called anastomosis) is a key process underlying the formation of vascular networks; each outgrowing angiogenic sprout must anastomose in order to allow blood flow in the newly formed vessel segment. It turns out that during this "sprouting and anastomosis" process, too many vessels are generated, and that blood flow is subsequently optimized through the removal (pruning) of low flow segments. Here, we reflect on the cellular and molecular mechanisms involved in forming the complex architecture of the vasculature through sprouting, anastomosis and pruning, and raise some questions that remain to be addressed in future studies.
Collapse
Affiliation(s)
- Jianmin Yin
- Biozentrum der Universität Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
41
|
Zink J, Frye M, Frömel T, Carlantoni C, John D, Schreier D, Weigert A, Laban H, Salinas G, Stingl H, Günther L, Popp R, Hu J, Vanhollebeke B, Schmidt H, Acker-Palmer A, Renné T, Fleming I, Benz PM. EVL regulates VEGF receptor-2 internalization and signaling in developmental angiogenesis. EMBO Rep 2021; 22:e48961. [PMID: 33512764 PMCID: PMC7857432 DOI: 10.15252/embr.201948961] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
Endothelial tip cells are essential for VEGF‐induced angiogenesis, but underlying mechanisms are elusive. The Ena/VASP protein family, consisting of EVL, VASP, and Mena, plays a pivotal role in axon guidance. Given that axonal growth cones and endothelial tip cells share many common features, from the morphological to the molecular level, we investigated the role of Ena/VASP proteins in angiogenesis. EVL and VASP, but not Mena, are expressed in endothelial cells of the postnatal mouse retina. Global deletion of EVL (but not VASP) compromises the radial sprouting of the vascular plexus in mice. Similarly, endothelial‐specific EVL deletion compromises the radial sprouting of the vascular plexus and reduces the endothelial tip cell density and filopodia formation. Gene sets involved in blood vessel development and angiogenesis are down‐regulated in EVL‐deficient P5‐retinal endothelial cells. Consistently, EVL deletion impairs VEGF‐induced endothelial cell proliferation and sprouting, and reduces the internalization and phosphorylation of VEGF receptor 2 and its downstream signaling via the MAPK/ERK pathway. Together, we show that endothelial EVL regulates sprouting angiogenesis via VEGF receptor‐2 internalization and signaling.
Collapse
Affiliation(s)
- Joana Zink
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Frömel
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Claudia Carlantoni
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David John
- German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Insitute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Danny Schreier
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Weigert
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Hebatullah Laban
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit (NIG), Institute of Human Genetics, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Heike Stingl
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Lea Günther
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Rüdiger Popp
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Jiong Hu
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, ULB Neuroscience Institute Department of Molecular Biology, University of Brussels, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium
| | - Hannes Schmidt
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt am Main, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Fleming
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Peter M Benz
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| |
Collapse
|
42
|
Zhang L, Wei F, Bai Q, Song D, Zheng Z, Wang Y, Liu X, Abdulrahman AA, Bian Y, Xu X, Chen C, Zhang H, Sun D. Oscillating Magnetic Field Regulates Cell Adherence and Endothelialization Based on Magnetic Nanoparticle-Modified Bacterial Cellulose. ACS APPLIED MATERIALS & INTERFACES 2020; 12:52467-52478. [PMID: 33170636 DOI: 10.1021/acsami.0c17213] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Despite the widely explored biomaterial scaffolds in vascular tissue engineering applications lately, no ideal platform has been provided for small diameter synthetic vascular grafts mainly due to the thrombosis issue. Endothelium is the only known completely non-thrombogenic material; so, functional endothelialization onto vascular biomaterials is critical in maintaining the patency of vascular networks. Bacterial cellulose (BC) is a natural biomaterial with superior biocompatibility and appropriate hydrophilicity as potential vascular grafts. In previous studies, surface modification of active peptides such as Arg-Gly-Asp (RGD) sequences onto biomaterials has been proven to achieve accelerated and selective endothelial cell (EC) adhesion. In our study, we demonstrated a new strategy to remotely regulate the adhesion of endothelial cells based on an oscillating magnetic field and achieve successful endothelialization on the modified BC membranes. In details, we synthesized bacterial cellulose (BC), magnetic BC (MBC), and RGD peptide-grafted magnetic BC (RMBC), modified with the HOOC-PEG-COOH-coated iron oxide nanoparticles (PEG-IONs). The endothelial cells were cultured on the three materials under different frequencies of an oscillating magnetic field, including "stationary" (0 Hz), "slow" (0.1 Hz), and "fast" (2 Hz) groups. Compared to BC and MBC membranes, the cells on RMBC membranes generally show better adhesion and proliferation. Meanwhile, the "slow" frequency of a magnetic field promotes this phenomenon on RMBC and achieves endothelialization after culture for 4 days, whereas "fast" inhibits the cellular attachment. Overall, we demonstrate a non-invasive and convenient method to regulate the endothelialization process, with promising applications in vascular tissue engineering.
Collapse
Affiliation(s)
- Lei Zhang
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Feng Wei
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Qianqian Bai
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, 999077 Hong Kong SAR, P.R. China
| | - Danhong Song
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Zhuofan Zheng
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Yafei Wang
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Xin Liu
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Al-Ammari Abdulrahman
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Yingxin Bian
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Xuran Xu
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Chuntao Chen
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| | - Hongsong Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing 210029, P.R. China
| | - Dongping Sun
- Chemicobiology and Functional Materials Institute, School of Chemical Engineering, Nanjing University of Science and Technology, Nanjing 210094, P.R. China
| |
Collapse
|
43
|
Marcksl1 modulates endothelial cell mechanoresponse to haemodynamic forces to control blood vessel shape and size. Nat Commun 2020; 11:5476. [PMID: 33127887 PMCID: PMC7603353 DOI: 10.1038/s41467-020-19308-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 10/02/2020] [Indexed: 11/16/2022] Open
Abstract
The formation of vascular tubes is driven by extensive changes in endothelial cell (EC) shape. Here, we have identified a role of the actin-binding protein, Marcksl1, in modulating the mechanical properties of EC cortex to regulate cell shape and vessel structure during angiogenesis. Increasing and depleting Marcksl1 expression level in vivo results in an increase and decrease, respectively, in EC size and the diameter of microvessels. Furthermore, endothelial overexpression of Marcksl1 induces ectopic blebbing on both apical and basal membranes, during and after lumen formation, that is suppressed by reduced blood flow. High resolution imaging reveals that Marcksl1 promotes the formation of linear actin bundles and decreases actin density at the EC cortex. Our findings demonstrate that a balanced network of linear and branched actin at the EC cortex is essential in conferring cortical integrity to resist the deforming forces of blood flow to regulate vessel structure. During lumen formation in blood vessels, endothelial cells become exposed to hemodynamic forces that induce membrane blebbing and changes in cell shape. Here, the authors show endothelial cells develop an actin-based protective mechanism in the cell cortex that prevents excessive blebbing to control cell shape and vessel diameter.
Collapse
|
44
|
Klems A, van Rijssel J, Ramms AS, Wild R, Hammer J, Merkel M, Derenbach L, Préau L, Hinkel R, Suarez-Martinez I, Schulte-Merker S, Vidal R, Sauer S, Kivelä R, Alitalo K, Kupatt C, van Buul JD, le Noble F. The GEF Trio controls endothelial cell size and arterial remodeling downstream of Vegf signaling in both zebrafish and cell models. Nat Commun 2020; 11:5319. [PMID: 33087700 PMCID: PMC7578835 DOI: 10.1038/s41467-020-19008-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Arterial networks enlarge in response to increase in tissue metabolism to facilitate flow and nutrient delivery. Typically, the transition of a growing artery with a small diameter into a large caliber artery with a sizeable diameter occurs upon the blood flow driven change in number and shape of endothelial cells lining the arterial lumen. Here, using zebrafish embryos and endothelial cell models, we describe an alternative, flow independent model, involving enlargement of arterial endothelial cells, which results in the formation of large diameter arteries. Endothelial enlargement requires the GEF1 domain of the guanine nucleotide exchange factor Trio and activation of Rho-GTPases Rac1 and RhoG in the cell periphery, inducing F-actin cytoskeleton remodeling, myosin based tension at junction regions and focal adhesions. Activation of Trio in developing arteries in vivo involves precise titration of the Vegf signaling strength in the arterial wall, which is controlled by the soluble Vegf receptor Flt1. Arterial flow regulates artery diameter but other mechanisms may also affect this. Here, the authors show that the guanine nucleotide exchange factor Trio and GTPases Rac1 and RhoG, triggers F-actin remodeling in arterial endothelial cells, independent of flow, to enhance lumen diameter in zebrafish and cell models.
Collapse
Affiliation(s)
- Alina Klems
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Jos van Rijssel
- Molecular Cell Biology lab, Department Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center at the University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands
| | - Anne S Ramms
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany.,Institute for Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021, Karlsruhe, Germany
| | - Raphael Wild
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Julia Hammer
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Melanie Merkel
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Laura Derenbach
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Laetitia Préau
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany
| | - Rabea Hinkel
- Laboratory Animal Science Unit, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany and DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Irina Suarez-Martinez
- Institute of Cardiovascular Organogenesis and Regeneration WWU Münster, Münster, Germany & Faculty of Medicine, WWU Münster, Münster, Germany & Cells in Motion Cluster of Excellence, Münster, Münster, Germany
| | - Stefan Schulte-Merker
- Institute of Cardiovascular Organogenesis and Regeneration WWU Münster, Münster, Germany & Faculty of Medicine, WWU Münster, Münster, Germany & Cells in Motion Cluster of Excellence, Münster, Münster, Germany
| | - Ramon Vidal
- Max Delbrück Center for Molecular Medicine (MDC), Berlin Institute of Medical Systems Biology & Berlin Institute of Health, Robert Rössle Strasse 10, 13092, Berlin, Germany
| | - Sascha Sauer
- Max Delbrück Center for Molecular Medicine (MDC), Berlin Institute of Medical Systems Biology & Berlin Institute of Health, Robert Rössle Strasse 10, 13092, Berlin, Germany
| | - Riikka Kivelä
- Stem Cells and Metabolism Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Helsinki, Finland
| | - Kari Alitalo
- Translational Cancer Medicine Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, and Wihuri Research Institute, Helsinki, Finland
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, TUM Munich, Germany, and DZHK, (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Jaap D van Buul
- Molecular Cell Biology lab, Department Molecular and Cellular Hemostasis, Sanquin Research and Landsteiner Laboratory, Academic Medical Center at the University of Amsterdam, Plesmanlaan 125, 1066CX, Amsterdam, The Netherlands.,Leeuwenhoek Centre for Advanced Microscopy, section Molecular Cytology at Swammerdam Institute for Life Sciences at University of Amsterdam, Amsterdam, The Netherlands
| | - Ferdinand le Noble
- Department of Cell and Developmental Biology, Institute of Zoology (ZOO), Karlsruhe Institute of Technology (KIT), Fritz Haber Weg 4, 76131, Karlsruhe, Germany. .,Institute for Biological and Chemical Systems-Biological Information Processing, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021, Karlsruhe, Germany. .,Institute of Experimental Cardiology, University of Heidelberg, Heidelberg Germany and DZHK (German Center for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
45
|
Mühleder S, Fernández-Chacón M, Garcia-Gonzalez I, Benedito R. Endothelial sprouting, proliferation, or senescence: tipping the balance from physiology to pathology. Cell Mol Life Sci 2020; 78:1329-1354. [PMID: 33078209 PMCID: PMC7904752 DOI: 10.1007/s00018-020-03664-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/05/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Therapeutic modulation of vascular cell proliferation and migration is essential for the effective inhibition of angiogenesis in cancer or its induction in cardiovascular disease. The general view is that an increase in vascular growth factor levels or mitogenic stimulation is beneficial for angiogenesis, since it leads to an increase in both endothelial proliferation and sprouting. However, several recent studies showed that an increase in mitogenic stimuli can also lead to the arrest of angiogenesis. This is due to the existence of intrinsic signaling feedback loops and cell cycle checkpoints that work in synchrony to maintain a balance between endothelial proliferation and sprouting. This balance is tightly and effectively regulated during tissue growth and is often deregulated or impaired in disease. Most therapeutic strategies used so far to promote vascular growth simply increase mitogenic stimuli, without taking into account its deleterious effects on this balance and on vascular cells. Here, we review the main findings on the mechanisms controlling physiological vascular sprouting, proliferation, and senescence and how those mechanisms are often deregulated in acquired or congenital cardiovascular disease leading to a diverse range of pathologies. We also discuss alternative approaches to increase the effectiveness of pro-angiogenic therapies in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Severin Mühleder
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Macarena Fernández-Chacón
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Irene Garcia-Gonzalez
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain.
| |
Collapse
|
46
|
Changchien C, Chen Y, Chang H, Chang S, Tsai W, Tsai H, Wang C, Lee H, Tsai C. Effect of malignant-associated pleural effusion on endothelial viability, motility and angiogenesis in lung cancer. Cancer Sci 2020; 111:3747-3758. [PMID: 32706142 PMCID: PMC7541005 DOI: 10.1111/cas.14584] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 12/30/2022] Open
Abstract
Malignant pleural effusion (MPE) and paramalignant pleural effusion (PPE) remain debilitating complications in lung cancer patients with poor prognosis and limited treatment options. The role of vascular endothelial cells has not been explored in the pleural environment of lung cancer. By integrating MPE and PPE as malignant-associated pleural fluid (MAPF), the current study aimed to evaluate the effect of MAPF on cell proliferation, migration and angiogenesis of HUVEC. First, increased capillaries were identified in the subpleural layer of lung adenocarcinoma. Compatible with pathological observations, the ubiquitous elevation of HUVEC survival was identified in MAPF culture regardless of the underlying cancer type, the driver gene mutation, prior treatments and evidence of malignant cells in pleural fluid. Moreover, MAPF enhanced HUVEC motility with the formation of lamellipodia and filopodia and focal adhesion complex. Tube formation assay revealed angiogenic behavior with the observation of sheet-like structures. HUVEC cultured with MAPF resulted in a significant increase in MAPK phosphorylation. Accompanied with VEGFR2 upregulation in MAPF culture, there was increased expressions of p-STAT3, HIF-1α and Nf-kB. VEGF/VEGFR2 blockade regressed endothelial migration and angiogenesis but not cell proliferation. Our data indicate the angiogenic activities of MAPF on vascular endothelial cells that revealed increased pleural capillaries in lung cancer. Targeting the VEGF/VEGFR2 pathway might modulate the angiogenic propensity of MAPF in future clinical investigations.
Collapse
MESH Headings
- Aged
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation/genetics
- Cell Survival/genetics
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Human Umbilical Vein Endothelial Cells
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Lung Neoplasms/complications
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Male
- NF-kappa B/genetics
- Neovascularization, Pathologic/complications
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/pathology
- Pleural Effusion/genetics
- Pleural Effusion, Malignant/complications
- Pleural Effusion, Malignant/genetics
- Pleural Effusion, Malignant/pathology
- STAT3 Transcription Factor/genetics
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor Receptor-2/genetics
Collapse
Affiliation(s)
- Chih‐Ying Changchien
- Department of Internal MedicineTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
- Department of Biology and AnatomyNational Defense Medical CenterTaipeiTaiwan
| | - Ying Chen
- Department of Biology and AnatomyNational Defense Medical CenterTaipeiTaiwan
| | - Hsin‐Han Chang
- Department of Biology and AnatomyNational Defense Medical CenterTaipeiTaiwan
| | - Shan‐Yueh Chang
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| | - Wen‐Chiuan Tsai
- Department of PathologyTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| | - Hao‐Chung Tsai
- Division of Chest MedicineDepartment of Internal MedicineTri‐Service General Hospital Songshan Branch, National Defense Medical CenterTaipeiTaiwan
| | - Chieh‐Yung Wang
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| | - Herng‐Sheng Lee
- Department of Pathology and Laboratory MedicineKaohsiung Veterans General HospitalKaohsiungTaiwan
| | - Chen‐Liang Tsai
- Division of Pulmonary and Critical Care MedicineDepartment of Internal MedicineTri‐Service General Hospital, National Defense Medical CenterTaipeiTaiwan
| |
Collapse
|
47
|
Wisniewski L, French V, Lockwood N, Valdivia LE, Frankel P. P130Cas/bcar1 mediates zebrafish caudal vein plexus angiogenesis. Sci Rep 2020; 10:15589. [PMID: 32973180 PMCID: PMC7518251 DOI: 10.1038/s41598-020-71753-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
P130CAS/BCAR1 belongs to the CAS family of adaptor proteins, with important regulatory roles in cell migration, cell cycle control, and apoptosis. Previously, we and others showed that P130CAS mediates VEGF-A and PDGF signalling in vitro, but its cardiovascular function in vivo remains relatively unexplored. We characterise here a novel deletion model of P130CAS in zebrafish. Using in vivo microscopy and transgenic vascular reporters, we observed that while bcar1−/− zebrafish showed no arterial angiogenic or heart defects during development, they strikingly failed to form the caudal vein plexus (CVP). Endothelial cells (ECs) within the CVP of bcar1−/− embryos produced fewer filopodial structures and did not detach efficiently from neighbouring cells, resulting in a significant reduction in ventral extension and overall CVP area. Mechanistically, we show that P130Cas mediates Bmp2b-induced ectopic angiogenic sprouting of ECs in the developing embryo and provide pharmacological evidence for a role of Src family kinases in CVP development.
Collapse
Affiliation(s)
- Laura Wisniewski
- Division of Medicine, University College London, 5 University Street, London, WC1E 6JF, UK. .,Queen Mary University of London, London, EC1M 6BQ, UK.
| | - Vanessa French
- Institute of Cardiovascular Science, University College London, 5 University Street, London, WC1E 6JF, UK
| | - Nicola Lockwood
- Division of Medicine, University College London, 5 University Street, London, WC1E 6JF, UK.,The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Leonardo E Valdivia
- Center for Integrative Biology, Faculty of Sciences, Universidad Mayor, Santiago, Chile
| | - Paul Frankel
- Institute of Cardiovascular Science, University College London, 5 University Street, London, WC1E 6JF, UK.
| |
Collapse
|
48
|
Helker CS, Eberlein J, Wilhelm K, Sugino T, Malchow J, Schuermann A, Baumeister S, Kwon HB, Maischein HM, Potente M, Herzog W, Stainier DY. Apelin signaling drives vascular endothelial cells toward a pro-angiogenic state. eLife 2020; 9:55589. [PMID: 32955436 PMCID: PMC7567607 DOI: 10.7554/elife.55589] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 09/19/2020] [Indexed: 12/18/2022] Open
Abstract
To form new blood vessels (angiogenesis), endothelial cells (ECs) must be activated and acquire highly migratory and proliferative phenotypes. However, the molecular mechanisms that govern these processes are incompletely understood. Here, we show that Apelin signaling functions to drive ECs into such an angiogenic state. Zebrafish lacking Apelin signaling exhibit defects in endothelial tip cell morphology and sprouting. Using transplantation experiments, we find that in mosaic vessels, wild-type ECs leave the dorsal aorta (DA) and form new vessels while neighboring ECs defective in Apelin signaling remain in the DA. Mechanistically, Apelin signaling enhances glycolytic activity in ECs at least in part by increasing levels of the growth-promoting transcription factor c-Myc. Moreover, APELIN expression is regulated by Notch signaling in human ECs, and its function is required for the hypersprouting phenotype in Delta-like 4 (Dll4) knockdown zebrafish embryos. These data provide new insights into fundamental principles of blood vessel formation and Apelin signaling, enabling a better understanding of vascular growth in health and disease.
Collapse
Affiliation(s)
- Christian Sm Helker
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | - Jean Eberlein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | - Kerstin Wilhelm
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Toshiya Sugino
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Julian Malchow
- Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | | | - Stefan Baumeister
- Philipps-University Marburg, Faculty of Biology, Cell Signaling and Dynamics, Marburg, Germany
| | - Hyouk-Bum Kwon
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hans-Martin Maischein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Michael Potente
- Angiogenesis and Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK (German Center for Cardiovascular Research), partner site Frankfurt Rhine-Main, Berlin, Germany
| | - Wiebke Herzog
- University of Muenster, Muenster, Germany.,Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DZHK (German Center for Cardiovascular Research), partner site Frankfurt Rhine-Main, Berlin, Germany
| |
Collapse
|
49
|
Muscarella AM, Dai W, Mitchell PG, Zhang W, Wang H, Jia L, Stossi F, Mancini MA, Chiu W, Zhang XHF. Unique cellular protrusions mediate breast cancer cell migration by tethering to osteogenic cells. NPJ Breast Cancer 2020; 6:42. [PMID: 32964116 PMCID: PMC7477119 DOI: 10.1038/s41523-020-00183-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 08/11/2020] [Indexed: 12/13/2022] Open
Abstract
Migration and invasion are key properties of metastatic cancer cells. These properties can be acquired through intrinsic reprogramming processes such as epithelial-mesenchymal transition. In this study, we discovered an alternative "migration-by-tethering" mechanism through which cancer cells gain the momentum to migrate by adhering to mesenchymal stem cells or osteoblasts. This tethering is mediated by both heterotypic adherens junctions and gap junctions, and leads to a unique cellular protrusion supported by cofilin-coated actin filaments. Inhibition of gap junctions or depletion of cofilin reduces migration-by-tethering. We observed evidence of these protrusions in bone segments harboring experimental and spontaneous bone metastasis in animal models. These data exemplify how cancer cells may acquire migratory ability without intrinsic reprogramming. Furthermore, given the important roles of osteogenic cells in early-stage bone colonization, our observations raise the possibility that migration-by-tethering may drive the relocation of disseminated tumor cells between different niches in the bone microenvironment.
Collapse
Affiliation(s)
- Aaron M. Muscarella
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Wei Dai
- Department of Cell Biology and Neuroscience, Institute for Quantitative Biomedicine, Rutgers University, 174 Frelinghuysen Road, Piscataway, NJ 08854 USA
| | - Patrick G. Mitchell
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- National Center for Macromolecular Imaging, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305 USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Luyu Jia
- Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Fabio Stossi
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Michael A. Mancini
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Wah Chiu
- Department of Bioengineering, Stanford University, Stanford, CA 94305 USA
| | - Xiang H.-F. Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| |
Collapse
|
50
|
Hiepen C, Mendez PL, Knaus P. It Takes Two to Tango: Endothelial TGFβ/BMP Signaling Crosstalk with Mechanobiology. Cells 2020; 9:E1965. [PMID: 32858894 PMCID: PMC7564048 DOI: 10.3390/cells9091965] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-beta (TGFβ) superfamily of cytokines. While some ligand members are potent inducers of angiogenesis, others promote vascular homeostasis. However, the precise understanding of the molecular mechanisms underlying these functions is still a growing research field. In bone, the tissue in which BMPs were first discovered, crosstalk of TGFβ/BMP signaling with mechanobiology is well understood. Likewise, the endothelium represents a tissue that is constantly exposed to multiple mechanical triggers, such as wall shear stress, elicited by blood flow or strain, and tension from the surrounding cells and to the extracellular matrix. To integrate mechanical stimuli, the cytoskeleton plays a pivotal role in the transduction of these forces in endothelial cells. Importantly, mechanical forces integrate on several levels of the TGFβ/BMP pathway, such as receptors and SMADs, but also global cell-architecture and nuclear chromatin re-organization. Here, we summarize the current literature on crosstalk mechanisms between biochemical cues elicited by TGFβ/BMP growth factors and mechanical cues, as shear stress or matrix stiffness that collectively orchestrate endothelial function. We focus on the different subcellular compartments in which the forces are sensed and integrated into the TGFβ/BMP growth factor signaling.
Collapse
Affiliation(s)
| | | | - Petra Knaus
- Knaus-Lab/Signal Transduction, Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, 14195 Berlin, Germany; (C.H.); (P.-L.M.)
| |
Collapse
|