1
|
Yin J, Maggi L, Wiesner C, Affolter M, Belting HG. Oscillatory contractile forces refine endothelial cell-cell interactions for continuous lumen formation governed by Heg1/Ccm1. Angiogenesis 2024:10.1007/s10456-024-09945-5. [PMID: 39249713 DOI: 10.1007/s10456-024-09945-5] [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: 06/10/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024]
Abstract
The formation and organization of complex blood vessel networks rely on various biophysical forces, yet the mechanisms governing endothelial cell-cell interactions under different mechanical inputs are not well understood. Using the dorsal longitudinal anastomotic vessel (DLAV) in zebrafish as a model, we studied the roles of multiple biophysical inputs and cerebral cavernous malformation (CCM)-related genes in angiogenesis. Our research identifies heg1 and krit1 (ccm1) as crucial for the formation of endothelial cell-cell interfaces during anastomosis. In mutants of these genes, cell-cell interfaces are entangled with fragmented apical domains. A Heg1 live reporter demonstrated that Heg1 is dynamically involved in the oscillatory constrictions along cell-cell junctions, whilst a Myosin live reporter indicated that heg1 and krit1 mutants lack actomyosin contractility along these junctions. In wild-type embryos, the oscillatory contractile forces at junctions refine endothelial cell-cell interactions by straightening junctions and eliminating excessive cell-cell interfaces. Conversely, in the absence of junctional contractility, the cell-cell interfaces become entangled and prone to collapse in both mutants, preventing the formation of a continuous luminal space. By restoring junctional contractility via optogenetic activation of RhoA, contorted junctions are straightened and disentangled. Additionally, haemodynamic forces complement actomyosin contractile forces in resolving entangled cell-cell interfaces in both wild-type and mutant embryos. Overall, our study reveals that oscillatory contractile forces governed by Heg1 and Krit1 are essential for maintaining proper endothelial cell-cell interfaces and thus for the formation of a continuous luminal space, which is essential to generate a functional vasculature.
Collapse
Affiliation(s)
- Jianmin Yin
- Department of Cell Biology, Biozentrum, University of Basel, Spitalstrasse 41, Basel, 4056, Switzerland.
| | - Ludovico Maggi
- Department of Cell Biology, Biozentrum, University of Basel, Spitalstrasse 41, Basel, 4056, Switzerland
| | - Cora Wiesner
- Department of Cell Biology, Biozentrum, University of Basel, Spitalstrasse 41, Basel, 4056, Switzerland
| | - Markus Affolter
- Department of Cell Biology, Biozentrum, University of Basel, Spitalstrasse 41, Basel, 4056, Switzerland.
| | - Heinz-Georg Belting
- Department of Cell Biology, Biozentrum, University of Basel, Spitalstrasse 41, Basel, 4056, Switzerland.
| |
Collapse
|
2
|
Nguyen ML, Demri N, Lapin B, Di Federico F, Gropplero G, Cayrac F, Hennig K, Gomes ER, Wilhelm C, Roman W, Descroix S. Studying the impact of geometrical and cellular cues on myogenesis with a skeletal muscle-on-chip. LAB ON A CHIP 2024; 24:4147-4160. [PMID: 39072529 DOI: 10.1039/d4lc00417e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
In the skeletal muscle tissue, cells are organized following an anisotropic architecture, which is both required during myogenesis when muscle precursor cells fuse to generate myotubes and for its contractile function. To build an in vitro skeletal muscle tissue, it is therefore essential to develop methods to organize cells in an anisotropic fashion, which can be particularly challenging, especially in 3D. In this study, we present a versatile muscle-on-chip system with adjustable collagen hollow tubes that can be seeded with muscle precursor cells. The collagen acts both as a tube-shaped hollow mold and as an extracellular matrix scaffold that can house other cell types for co-culture. We found that the diameter of the channel affects the organization of the muscle cells and that proper myogenesis was obtained at a diameter of 75 μm. In these conditions, muscle precursor cells fused into long myotubes aligned along these collagen channels, resulting in a fascicle-like structure. These myotubes exhibited actin striations and upregulation of multiple myogenic genes, reflecting their maturation. Moreover, we showed that our chip allowed muscle tissue culture and maturation over a month, with the possibility of fibroblast co-culture embedding in the collagen matrix.
Collapse
Affiliation(s)
- M-L Nguyen
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - N Demri
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - B Lapin
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - F Di Federico
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - G Gropplero
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - F Cayrac
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - K Hennig
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Edgar R Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - C Wilhelm
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| | - W Roman
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Australian Regenerative Medicine Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - S Descroix
- Laboratoire Physico Chimie Curie, PCC, CNRS UMR168, Institut Curie, Sorbonne University, PSL University, 75005 Paris, France.
| |
Collapse
|
3
|
Kukharchuk O, Bopardikar A, Anand Baskaran PP, Kukharchuk A, Kulkarni R, Ranbhor R. Fetal progenitor cells for treatment of chronic limb ischemia. AMERICAN JOURNAL OF STEM CELLS 2024; 13:169-190. [PMID: 39021376 PMCID: PMC11249671 DOI: 10.62347/mzki8393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/06/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES This study investigated the therapeutic potential of fetal progenitor cells (FPCs) in the treatment of chronic non-healing wounds and ulcers associated with chronic limb ischemia (CLI). The research aimed to elucidate the mechanism of action of FPCs and evaluate their efficacy and safety in CLI patients. METHODS The researchers isolated FPCs from aborted human fetal liver, brain, and skin tissues and thoroughly characterized them. The preclinical phase of the study involved assessing the effects of FPCs in a rat model of CLI. Subsequently, a randomized controlled clinical trial was conducted to compare the efficacy of FPCs with standard treatment and autologous bone marrow mononuclear cells in CLI patients. The clinical trial lasted 12 months, with a follow-up period of 24-36 months. The primary outcomes included wound healing, frequency of major and minor amputations, pain reduction, and the incidence of complications. Secondary outcomes involved changes in local hemodynamics and histological, ultrastructural, and immunohistochemical assessments of angiogenesis. RESULTS In the animal model, FPC treatment significantly enhanced angiogenesis and accelerated healing of ischemic wounds compared to controls. The clinical trial in CLI patients demonstrated that the FPC therapy achieved substantially higher rates of complete wound closure, prevention of major amputation, pain reduction, and improvement in ankle-brachial index compared to control groups. Notably, the study reported no serious adverse events. CONCLUSIONS FPC therapy exhibited remarkable efficacy in promoting the healing of ischemic wounds, preventing amputation, and improving symptoms and quality of life in patients with CLI. The proangiogenic and provasculogenic effects of FPCs may be attributed to their ability to secrete specific growth factors. These findings provide new insights into the development of cellular therapeutic angiogenesis as a promising approach for the treatment of peripheral arterial diseases.
Collapse
Affiliation(s)
- Oleksandr Kukharchuk
- ReeLabs Pvt. Ltd.1st Floor, KK Chambers, Sir P.T. Rd., Azad Maidan, Fort, Mumbai 400001, India
| | | | | | - Andrii Kukharchuk
- ReeLabs Pvt. Ltd.1st Floor, KK Chambers, Sir P.T. Rd., Azad Maidan, Fort, Mumbai 400001, India
| | - Rohit Kulkarni
- ReeLabs Pvt. Ltd.1st Floor, KK Chambers, Sir P.T. Rd., Azad Maidan, Fort, Mumbai 400001, India
| | | |
Collapse
|
4
|
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
|
5
|
Duong VT, Lin CC. Digital Light Processing 3D Bioprinting of Gelatin-Norbornene Hydrogel for Enhanced Vascularization. Macromol Biosci 2023; 23:e2300213. [PMID: 37536347 PMCID: PMC10837335 DOI: 10.1002/mabi.202300213] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Indexed: 08/05/2023]
Abstract
Digital light processing (DLP) bioprinting can be used to fabricate volumetric scaffolds with intricate internal structures, such as perfusable vascular channels. The successful implementation of DLP bioprinting in tissue fabrication requires using suitable photo-reactive bioinks. Norbornene-based bioinks have emerged as an attractive alternative to (meth)acrylated macromers in 3D bioprinting owing to their mild and rapid reaction kinetics, high cytocompatibility for in situ cell encapsulation, and adaptability for post-printing modification or conjugation of bioactive motifs. In this contribution, the development of gelatin-norbornene (GelNB) is reported as a photo-cross-linkable bioink for DLP 3D bioprinting. Low concentrations of GelNB (2-5 wt.%) and poly(ethylene glycol)-tetra-thiol (PEG4SH) are DLP-printed with a wide range of stiffness (G' ≈120 to 4000 Pa) and with perfusable channels. DLP-printed GelNB hydrogels are highly cytocompatible, as demonstrated by the high viability of the encapsulated human umbilical vein endothelial cells (HUVECs). The encapsulated HUVECs formed an interconnected microvascular network with lumen structures. Notably, the GelNB bioink permitted both in situ tethering and secondary conjugation of QK peptide, a vascular endothelial growth factor (VEGF)-mimetic peptide. Incorporation of QK peptide significantly improved endothelialization and vasculogenesis of the DLP-printed GelNB hydrogels, reinforcing the applicability of this bioink system in diverse biofabrication applications.
Collapse
Affiliation(s)
- Van Thuy Duong
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
6
|
Rosowski S, Remmert C, Marder M, Akishiba M, Bushe J, Feuchtinger A, Platen A, Ussar S, Theis F, Wiedenmann S, Meier M. Single-cell characterization of neovascularization using hiPSC-derived endothelial cells in a 3D microenvironment. Stem Cell Reports 2023; 18:1972-1986. [PMID: 37714147 PMCID: PMC10656300 DOI: 10.1016/j.stemcr.2023.08.008] [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: 10/21/2022] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 09/17/2023] Open
Abstract
The formation of vascular structures is fundamental for in vitro tissue engineering. Vascularization can enable the nutrient supply within larger structures and increase transplantation efficiency. We differentiated human induced pluripotent stem cells toward endothelial cells in 3D suspension culture. To investigate in vitro neovascularization and various 3D microenvironmental approaches, we designed a comprehensive single-cell transcriptomic study. Time-resolved single-cell transcriptomics of the endothelial and co-evolving mural cells gave insights into cell type development, stability, and plasticity. Transfer to a 3D hydrogel microenvironment induced neovascularization and facilitated tracing of migrating, coalescing, and tubulogenic endothelial cell states. During maturation, we monitored two pericyte subtypes evolving mural cells. Profiling cell-cell interactions between pericytes and endothelial cells revealed angiogenic signals during tubulogenesis. In silico discovered ligands were tested for their capability to attract endothelial cells. Our data, analyses, and results provide an in vitro roadmap to guide vascularization in future tissue engineering.
Collapse
Affiliation(s)
- Simon Rosowski
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Caroline Remmert
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Maren Marder
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Misao Akishiba
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Judith Bushe
- Research Unit Analytical Pathology, Helmholtz München, 85764 Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz München, 85764 Neuherberg, Germany
| | - Alina Platen
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Siegfried Ussar
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Fabian Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany; Department of Mathematics, Technical University of Munich, 85748 Garching bei München, Germany
| | - Sandra Wiedenmann
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Matthias Meier
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany; University Leipzig, Center for Biotechnology and Biomedicine, Institute of Biochemistry, Leipzig, Germany.
| |
Collapse
|
7
|
Jian H, Poetsch A. CASZ1: Current Implications in Cardiovascular Diseases and Cancers. Biomedicines 2023; 11:2079. [PMID: 37509718 PMCID: PMC10377389 DOI: 10.3390/biomedicines11072079] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/09/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Castor zinc finger 1 (CASZ1) is a C2H2 zinc finger family protein that has two splicing variants, CASZ1a and CASZ1b. It is involved in multiple physiological processes, such as tissue differentiation and aldosterone antagonism. Genetic and epigenetic alternations of CASZ1 have been characterized in multiple cardiovascular disorders, such as congenital heart diseases, chronic venous diseases, and hypertension. However, little is known about how CASZ1 mechanically participates in the pathogenesis of these diseases. Over the past decades, at first glance, paradoxical influences on cell behaviors and progressions of different cancer types have been discovered for CASZ1, which may be explained by a "double-agent" role for CASZ1. In this review, we discuss the physiological function of CASZ1, and focus on the association of CASZ1 aberrations with the pathogenesis of cardiovascular diseases and cancers.
Collapse
Affiliation(s)
- Heng Jian
- Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Ansgar Poetsch
- Queen Mary School, Nanchang University, Nanchang 330006, China
- School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| |
Collapse
|
8
|
Wang J, Song Y, Xie W, Zhao J, Wang Y, Yu W. Therapeutic angiogenesis based on injectable hydrogel for protein delivery in ischemic heart disease. iScience 2023; 26:106577. [PMID: 37192972 PMCID: PMC10182303 DOI: 10.1016/j.isci.2023.106577] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Ischemic heart disease (IHD) remains the leading cause of death and disability worldwide and leads to myocardial necrosis and negative myocardial remodeling, ultimately leading to heart failure. Current treatments include drug therapy, interventional therapy, and surgery. However, some patients with severe diffuse coronary artery disease, complex coronary artery anatomy, and other reasons are unsuitable for these treatments. Therapeutic angiogenesis stimulates the growth of the original blood vessels by using exogenous growth factors to generate more new blood vessels, which provides a new treatment for IHD. However, direct injection of these growth factors can cause a short half-life and serious side effects owing to systemic spread. Therefore, to overcome this problem, hydrogels have been developed for temporally and spatially controlled delivery of single or multiple growth factors to mimic the process of angiogenesis in vivo. This paper reviews the mechanism of angiogenesis, some important bioactive molecules, and natural and synthetic hydrogels currently being applied for bioactive molecule delivery to treat IHD. Furthermore, the current challenges of therapeutic angiogenesis in IHD and its potential solutions are discussed to facilitate real translation into clinical applications in the future.
Collapse
Affiliation(s)
- Junke Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Yancheng Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
| | - Wenjie Xie
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Shandong, Qingdao, Shandong 26000, China
| | - Jiang Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ying Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong 26000, China
- Corresponding author
| | - Wenzhou Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26003, China
- Corresponding author
| |
Collapse
|
9
|
Davis GE, Kemp SS. Extracellular Matrix Regulation of Vascular Morphogenesis, Maturation, and Stabilization. Cold Spring Harb Perspect Med 2023; 13:a041156. [PMID: 35817544 PMCID: PMC10578078 DOI: 10.1101/cshperspect.a041156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The extracellular matrix represents a critical regulator of tissue vascularization during embryonic development and postnatal life. In this perspective, we present key information and concepts that focus on how the extracellular matrix controls capillary assembly, maturation, and stabilization, and, in addition, contributes to tissue stability and health. In particular, we present and discuss mechanistic details underlying (1) the role of the extracellular matrix in controlling different steps of vascular morphogenesis, (2) the ability of endothelial cells (ECs) and pericytes to coassemble into elongated and narrow capillary EC-lined tubes with associated pericytes and basement membrane matrices, and (3) the identification of specific growth factor combinations ("factors") and peptides as well as coordinated "factor" and extracellular matrix receptor signaling pathways that are required to form stabilized capillary networks.
Collapse
Affiliation(s)
- George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida 33612, USA
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida 33612, USA
| |
Collapse
|
10
|
Cabral KA, Srivastava V, Graham AJ, Coyle MC, Stashko C, Weaver V, Gartner ZJ. Programming the Self-Organization of Endothelial Cells into Perfusable Microvasculature. Tissue Eng Part A 2023; 29:80-92. [PMID: 36181350 PMCID: PMC10266707 DOI: 10.1089/ten.tea.2022.0072] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/15/2022] [Indexed: 11/12/2022] Open
Abstract
The construction of three-dimensional (3D) microvascular networks with defined structures remains challenging. Emerging bioprinting strategies provide a means of patterning endothelial cells (ECs) into the geometry of 3D microvascular networks, but the microenvironmental cues necessary to promote their self-organization into cohesive and perfusable microvessels are not well known. To this end, we reconstituted microvessel formation in vitro by patterning thin lines of closely packed ECs fully embedded within a 3D extracellular matrix (ECM) and observed how different microenvironmental parameters influenced EC behaviors and their self-organization into microvessels. We found that the inclusion of fibrillar matrices, such as collagen I, into the ECM positively influenced cell condensation into extended geometries such as cords. We also identified the presence of a high-molecular-weight protein(s) in fetal bovine serum that negatively influenced EC condensation. This component destabilized cord structure by promoting cell protrusions and destabilizing cell-cell adhesions. Endothelial cords cultured in the presence of fibrillar collagen and in the absence of this protein activity were able to polarize, lumenize, incorporate mural cells, and support fluid flow. These optimized conditions allowed for the construction of branched and perfusable microvascular networks directly from patterned cells in as little as 3 days. These findings reveal important design principles for future microvascular engineering efforts based on bioprinting and micropatterning techniques. Impact statement Bioprinting is a potential strategy to achieve microvascularization in engineered tissues. However, the controlled self-organization of patterned endothelial cells into perfusable microvasculature remains challenging. We used DNA Programmed Assembly of Cells to create cell-dense, capillary-sized cords of endothelial cells with complete control over their structure. We optimized the matrix and media conditions to promote self-organization and maturation of these endothelial cords into stable and perfusable microvascular networks.
Collapse
Affiliation(s)
- Katelyn A. Cabral
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Austin J. Graham
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
| | - Maxwell C. Coyle
- Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, California, USA
| | - Connor Stashko
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, Berkeley, California, USA
| | - Valerie Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, University of California, San Francisco, San Francisco, California, USA
- Center for Cellular Construction, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
11
|
Yvernogeau L, Dainese G, Jaffredo T. Dorsal aorta polarization and haematopoietic stem cell emergence. Development 2023; 150:286251. [PMID: 36602140 DOI: 10.1242/dev.201173] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Recent studies have highlighted the crucial role of the aorta microenvironment in the generation of the first haematopoietic stem cells (HSCs) from specialized haemogenic endothelial cells (HECs). Despite more than two decades of investigations, we require a better understanding of the cellular and molecular events driving aorta formation and polarization, which will be pivotal to establish the mechanisms that operate during HEC specification and HSC competency. Here, we outline the early mechanisms involved in vertebrate aorta formation by comparing four different species: zebrafish, chicken, mouse and human. We highlight how this process, which is tightly controlled in time and space, requires a coordinated specification of several cell types, in particular endothelial cells originating from distinct mesodermal tissues. We also discuss how molecular signals originating from the aorta environment result in its polarization, creating a unique entity for HSC generation.
Collapse
Affiliation(s)
- Laurent Yvernogeau
- Sorbonne Université, IBPS, CNRS UMR7622, Inserm U1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Giovanna Dainese
- Sorbonne Université, IBPS, CNRS UMR7622, Inserm U1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Thierry Jaffredo
- Sorbonne Université, IBPS, CNRS UMR7622, Inserm U1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| |
Collapse
|
12
|
Arena AF, Escudero J, Shaye DD. A metazoan-specific C-terminal motif in EXC-4 and Gα-Rho/Rac signaling regulate cell outgrowth during tubulogenesis in C. elegans. Development 2022; 149:285944. [PMID: 36398726 PMCID: PMC10108608 DOI: 10.1242/dev.200748] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
Chloride intracellular channels (CLICs) are conserved proteins for which the cellular and molecular functions remain mysterious. An important insight into CLIC function came from the discovery that Caenorhabditis elegans EXC-4/CLIC regulates morphogenesis of the excretory canal (ExCa) cell, a single-cell tube. Subsequent work showed that mammalian CLICs regulate vascular development and angiogenesis, and human CLIC1 can rescue exc-4 mutants, suggesting conserved function in biological tube formation (tubulogenesis) and maintenance. However, the cell behaviors and signaling pathways regulated by EXC-4/CLICs during tubulogenesis in vivo remain largely unknown. We report a new exc-4 mutation, affecting a C-terminal residue conserved in virtually all metazoan CLICs, that reveals a specific role for EXC-4 in ExCa outgrowth. Cell culture studies suggest a function for CLICs in heterotrimeric G protein (Gα/β/γ)-Rho/Rac signaling, and Rho-family GTPases are common regulators of cell outgrowth. Using our new exc-4 mutant, we describe a previously unknown function for Gα-encoding genes (gpa-12/Gα12/13, gpa-7/Gαi, egl-30/Gαq and gsa-1/Gαs), ced-10/Rac and mig-2/RhoG in EXC-4-mediated ExCa outgrowth. Our results demonstrate that EXC-4/CLICs are primordial players in Gα-Rho/Rac-signaling, a pathway that is crucial for tubulogenesis in C. elegans and in vascular development.
Collapse
Affiliation(s)
- Anthony F Arena
- Department of Physiology and Biophysics, University of Illinois at Chicago - College of Medicine, Chicago, IL 60612, USA.,Graduate Education in Biomedical Sciences program, University of Illinois at Chicago - College of Medicine, Chicago, IL 60612, USA
| | - Julianna Escudero
- Department of Physiology and Biophysics, University of Illinois at Chicago - College of Medicine, Chicago, IL 60612, USA
| | - Daniel D Shaye
- Department of Physiology and Biophysics, University of Illinois at Chicago - College of Medicine, Chicago, IL 60612, USA.,Center for Cardiovascular Research, University of Illinois at Chicago - College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Bats ML, Peghaire C, Delobel V, Dufourcq P, Couffinhal T, Duplàa C. Wnt/frizzled Signaling in Endothelium: A Major Player in Blood-Retinal- and Blood-Brain-Barrier Integrity. Cold Spring Harb Perspect Med 2022; 12:a041219. [PMID: 35074794 PMCID: PMC9121893 DOI: 10.1101/cshperspect.a041219] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The Wnt/frizzled signaling pathway is one of the major regulators of endothelial biology, controlling key cellular activities. Many secreted Wnt ligands have been identified and can initiate diverse signaling via binding to a complex set of Frizzled (Fzd) transmembrane receptors and coreceptors. Roughly, Wnt signaling is subdivided into two pathways: the canonical Wnt/β-catenin signaling pathway whose main downstream effector is the transcriptional coactivator β-catenin, and the noncanonical Wnt signaling pathway, which is subdivided into the Wnt/Ca2+ pathway and the planar cell polarity pathway. Here, we will focus on its cross talk with other angiogenic pathways and on its role in blood-retinal- and blood-brain-barrier formation and its maintenance in a differentiated state. We will unravel how retinal vascular pathologies and neurovascular degenerative diseases result from disruption of the Wnt pathway related to vascular instability, and highlight current research into therapeutic options.
Collapse
Affiliation(s)
- Marie-Lise Bats
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
- Department of Biochemistry, Pellegrin Hospital, University Hospital of Bordeaux, 33076 Bordeaux Cedex, France
| | - Claire Peghaire
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
| | - Valentin Delobel
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
| | - Pascale Dufourcq
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
| | - Thierry Couffinhal
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
- Centre d'exploration, de prévention et de traitement de l'athérosclérose (CEPTA), CHU Bordeaux, 33000 Bordeaux, France
| | - Cécile Duplàa
- Univ. Bordeaux, Inserm, UMR1034, Biology of Cardiovascular Diseases, F-33600 Pessac, France
| |
Collapse
|
14
|
The basement membrane controls size and integrity of the Drosophila tracheal tubes. Cell Rep 2022; 39:110734. [PMID: 35476979 DOI: 10.1016/j.celrep.2022.110734] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 11/20/2022] Open
Abstract
Biological tubes are fundamental units of most metazoan organs. Their defective morphogenesis can cause malformations and pathologies. An integral component of biological tubes is the extracellular matrix, present apically (aECM) and basally (BM). Studies using the Drosophila tracheal system established an essential function for the aECM in tubulogenesis. Here, we demonstrate that the BM also plays a critical role in this process. We find that BM components are deposited in a spatial-temporal manner in the trachea. We show that laminins, core BM components, control size and shape of tracheal tubes and their topology within the embryo. At a cellular level, laminins control cell shape changes and distribution of the cortical cytoskeleton component α-spectrin. Finally, we report that the BM and aECM act independently-yet cooperatively-to control tube elongation and together to guarantee tissue integrity. Our results unravel key roles for the BM in shaping, positioning, and maintaining biological tubes.
Collapse
|
15
|
Francis CR, Kushner EJ. Trafficking in blood vessel development. Angiogenesis 2022; 25:291-305. [PMID: 35449244 PMCID: PMC9249721 DOI: 10.1007/s10456-022-09838-5] [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: 02/02/2022] [Accepted: 04/03/2022] [Indexed: 02/17/2023]
Abstract
Blood vessels demonstrate a multitude of complex signaling programs that work in concert to produce functional vasculature networks during development. A known, but less widely studied, area of endothelial cell regulation is vesicular trafficking, also termed sorting. After moving through the Golgi apparatus, proteins are shuttled to organelles, plugged into membranes, recycled, or degraded depending on the internal and extrinsic cues. A snapshot of these protein-sorting systems can be viewed as a trafficking signature that is not only unique to endothelial tissue, but critically important for blood vessel form and function. In this review, we will cover how vesicular trafficking impacts various aspects of angiogenesis, such as sprouting, lumen formation, vessel stabilization, and secretion, emphasizing the role of Rab GTPase family members and their various effectors.
Collapse
Affiliation(s)
- Caitlin R Francis
- Department of Biological Sciences, University of Denver, Denver, CO, 80210, USA
| | - Erich J Kushner
- Department of Biological Sciences, University of Denver, Denver, CO, 80210, USA.
| |
Collapse
|
16
|
Endothelial Cell Metabolism in Vascular Functions. Cancers (Basel) 2022; 14:cancers14081929. [PMID: 35454836 PMCID: PMC9031281 DOI: 10.3390/cancers14081929] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Recent findings in the field of vascular biology are nourishing the idea that targeting the endothelial cell metabolism may be an alternative strategy to antiangiogenic therapy, as well as a novel therapeutic approach for cardiovascular disease. Deepening the molecular mechanisms regulating how ECs re-adapt their metabolic status in response to the changeable conditions of the tissue microenvironment may be beneficial to develop novel innovative treatments to counteract the aberrant growth of vasculature. Abstract The endothelium is the innermost layer of all blood and lymphatic vessels composed of a monolayer of specialized endothelial cells (ECs). It is regarded as a dynamic and multifunctional endocrine organ that takes part in essential processes, such as the control of blood fluidity, the modulation of vascular tone, the regulation of immune response and leukocyte trafficking into perivascular tissues, and angiogenesis. The inability of ECs to perform their normal biological functions, known as endothelial dysfunction, is multi-factorial; for instance, it implicates the failure of ECs to support the normal antithrombotic and anti-inflammatory status, resulting in the onset of unfavorable cardiovascular conditions such as atherosclerosis, coronary artery disease, hypertension, heart problems, and other vascular pathologies. Notably, it is emerging that the ability of ECs to adapt their metabolic status to persistent changes of the tissue microenvironment could be vital for the maintenance of vascular functions and to prevent adverse vascular events. The main purpose of the present article is to shed light on the unique metabolic plasticity of ECs as a prospective therapeutic target; this may lead to the development of novel strategies for cardiovascular diseases and cancer.
Collapse
|
17
|
Seymour AJ, Westerfield AD, Cornelius VC, Skylar-Scott MA, Heilshorn SC. Bioprinted microvasculature: progressing from structure to function. Biofabrication 2022; 14:10.1088/1758-5090/ac4fb5. [PMID: 35086069 PMCID: PMC8988885 DOI: 10.1088/1758-5090/ac4fb5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/27/2022] [Indexed: 11/12/2022]
Abstract
Three-dimensional (3D) bioprinting seeks to unlock the rapid generation of complex tissue constructs, but long-standing challenges with efficientin vitromicrovascularization must be solved before this can become a reality. Microvasculature is particularly challenging to biofabricate due to the presence of a hollow lumen, a hierarchically branched network topology, and a complex signaling milieu. All of these characteristics are required for proper microvascular-and, thus, tissue-function. While several techniques have been developed to address distinct portions of this microvascularization challenge, no single approach is capable of simultaneously recreating all three microvascular characteristics. In this review, we present a three-part framework that proposes integration of existing techniques to generate mature microvascular constructs. First, extrusion-based 3D bioprinting creates a mesoscale foundation of hollow, endothelialized channels. Second, biochemical and biophysical cues induce endothelial sprouting to create a capillary-mimetic network. Third, the construct is conditioned to enhance network maturity. Across all three of these stages, we highlight the potential for extrusion-based bioprinting to become a central technique for engineering hierarchical microvasculature. We envision that the successful biofabrication of functionally engineered microvasculature will address a critical need in tissue engineering, and propel further advances in regenerative medicine andex vivohuman tissue modeling.
Collapse
Affiliation(s)
- Alexis J. Seymour
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Ashley D. Westerfield
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Vincent C. Cornelius
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Mark A. Skylar-Scott
- Department of Bioengineering, Stanford University, 443 Via Ortega, Shriram Center Room 119, Stanford, CA 94305, USA
| | - Sarah C. Heilshorn
- Department of Materials Science & Engineering, Stanford University, 476 Lomita Mall, McCullough Room 246, Stanford, CA 94305, USA
| |
Collapse
|
18
|
Sun Z, Kemp SS, Lin PK, Aguera KN, Davis GE. Endothelial k-RasV12 Expression Induces Capillary Deficiency Attributable to Marked Tube Network Expansion Coupled to Reduced Pericytes and Basement Membranes. Arterioscler Thromb Vasc Biol 2022; 42:205-222. [PMID: 34879709 PMCID: PMC8792373 DOI: 10.1161/atvbaha.121.316798] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE We sought to determine how endothelial cell (EC) expression of the activating k-Ras (kirsten rat sarcoma 2 viral oncogene homolog) mutation, k-RasV12, affects their ability to form lumens and tubes and interact with pericytes during capillary assembly Approach and Results: Using defined bioassays where human ECs undergo observable tubulogenesis, sprouting behavior, pericyte recruitment to EC-lined tubes, and pericyte-induced EC basement membrane deposition, we assessed the impact of EC k-RasV12 expression on these critical processes that are necessary for proper capillary network formation. This mutation, which is frequently seen in human ECs within brain arteriovenous malformations, was found to markedly accentuate EC lumen formation mechanisms, with strongly accelerated intracellular vacuole formation, vacuole fusion, and lumen expansion and with reduced sprouting behavior, leading to excessively widened tube networks compared with control ECs. These abnormal tubes demonstrate strong reductions in pericyte recruitment and pericyte-induced EC basement membranes compared with controls, with deficiencies in fibronectin, collagen type IV, and perlecan deposition. Analyses of signaling during tube formation from these k-RasV12 ECs reveals strong enhancement of Src (Src proto-oncogene, non-receptor tyrosine kinase), Pak2 (P21 [RAC1 (Rac family small GTPase 1)] activated kinase 2), b-Raf (v-raf murine sarcoma viral oncogene homolog B1), Erk (extracellular signal-related kinase), and Akt (AK strain transforming) activation and increased expression of PKCε (protein kinase C epsilon), MT1-MMP (membrane-type 1 matrix metalloproteinase), acetylated tubulin and CDCP1 (CUB domain-containing protein 1; most are known EC lumen regulators). Pharmacological blockade of MT1-MMP, Src, Pak, Raf, Mek (mitogen-activated protein kinase) kinases, Cdc42 (cell division cycle 42)/Rac1, and Notch markedly interferes with lumen and tube formation from these ECs. CONCLUSIONS Overall, this novel work demonstrates that EC expression of k-RasV12 disrupts capillary assembly due to markedly excessive lumen formation coupled with strongly reduced pericyte recruitment and basement membrane deposition, which are critical pathogenic features predisposing the vasculature to develop arteriovenous malformations.
Collapse
Affiliation(s)
- Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - Scott S. Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - Prisca K. Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - Kalia N. Aguera
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| | - George E. Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL 33612
| |
Collapse
|
19
|
Vakhrushev IV, Nezhurina EK, Karalkin PA, Tsvetkova AV, Sergeeva NS, Majouga AG, Yarygin KN. Heterotypic Multicellular Spheroids as Experimental and Preclinical Models of Sprouting Angiogenesis. BIOLOGY 2021; 11:18. [PMID: 35053016 PMCID: PMC8772844 DOI: 10.3390/biology11010018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022]
Abstract
Sprouting angiogenesis is the common response of live tissues to physiological and pathological angiogenic stimuli. Its accurate evaluation is of utmost importance for basic research and practical medicine and pharmacology and requires adequate experimental models. A variety of assays for angiogenesis were developed, none of them perfect. In vitro approaches are generally less physiologically relevant due to the omission of essential components regulating the process. However, only in vitro models can be entirely non-xenogeneic. The limitations of the in vitro angiogenesis assays can be partially overcome using 3D models mimicking tissue O2 and nutrient gradients, the influence of the extracellular matrix (ECM), and enabling cell-cell interactions. Here we present a review of the existing models of sprouting angiogenesis that are based on the use of endothelial cells (ECs) co-cultured with perivascular or other stromal cells. This approach provides an excellent in vitro platform for further decoding of the cellular and molecular mechanisms of sprouting angiogenesis under conditions close to the in vivo conditions, as well as for preclinical drug testing and preclinical research in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Igor V. Vakhrushev
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| | - Elizaveta K. Nezhurina
- P.A. Hertsen Moscow Oncology Research Center, National Medical Research Radiological Center, 125284 Moscow, Russia;
| | - Pavel A. Karalkin
- Institute for Cluster Oncology, Sechenov University, 119435 Moscow, Russia;
| | | | - Nataliya S. Sergeeva
- Department of Biology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Alexander G. Majouga
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Products, D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia;
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| |
Collapse
|
20
|
Chen D, Schwartz MA, Simons M. Developmental Perspectives on Arterial Fate Specification. Front Cell Dev Biol 2021; 9:691335. [PMID: 34249941 PMCID: PMC8269928 DOI: 10.3389/fcell.2021.691335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
Blood vessel acquisition of arterial or venous fate is an adaptive phenomenon in response to increasing blood circulation during vascular morphogenesis. The past two decades of effort in this field led to development of a widely accepted paradigm of molecular regulators centering on VEGF and Notch signaling. More recent findings focused on shear stress-induced cell cycle arrest as a prerequisite for arterial specification substantially modify this traditional understanding. This review aims to summarize key molecular mechanisms that work in concert to drive the acquisition of arterial fate in two distinct developmental settings of vascular morphogenesis: de novo vasculogenesis of the dorsal aorta and postnatal retinal angiogenesis. We will also discuss the questions and conceptual controversies that potentially point to novel directions of investigation and possible clinical relevance.
Collapse
Affiliation(s)
- Dongying Chen
- Yale Cardiovascular Research Center, Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Martin A. Schwartz
- Yale Cardiovascular Research Center, Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Michael Simons
- Yale Cardiovascular Research Center, Departments of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
21
|
Mandrycky CJ, Howard CC, Rayner SG, Shin YJ, Zheng Y. Organ-on-a-chip systems for vascular biology. J Mol Cell Cardiol 2021; 159:1-13. [PMID: 34118217 DOI: 10.1016/j.yjmcc.2021.06.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/03/2021] [Accepted: 06/06/2021] [Indexed: 12/18/2022]
Abstract
Organ-on-a-chip (OOC) platforms involve the miniaturization of cell culture systems and enable a variety of novel experimental approaches. These range from modeling the independent effects of biophysical forces on cells to screening novel drugs in multi-organ microphysiological systems, all within microscale devices. As in living systems, the incorporation of vascular structure is a key feature common to almost all organ-on-a-chip systems. In this review we highlight recent advances in organ-on-a-chip technologies with a focus on the vasculature. We first present the developmental process of the blood vessels through which vascular cells assemble into networks and remodel to form complex vascular beds under flow. We then review self-assembled vascular models and flow systems for the study of vascular development and biology as well as pre-patterned vascular models for the generation of perfusable microvessels for modeling vascular and tissue function. We finally conclude with a perspective on developing future OOC approaches for studying different aspects of vascular biology. We highlight the fit for purpose selection of OOC models towards either simple but powerful testbeds for therapeutic development, or complex vasculature to accurately replicate human physiology for specific disease modeling and tissue regeneration.
Collapse
Affiliation(s)
- Christian J Mandrycky
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA.
| | - Caitlin C Howard
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA.
| | - Samuel G Rayner
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA; Department of Medicine; Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA 98195, USA.
| | - Yu Jung Shin
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA.
| | - Ying Zheng
- Department of Bioengineering, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA; Institute for Stem Cell and Regenerative Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
22
|
Ricolo D, Castro-Ribera J, Araújo SJ. Cytoskeletal players in single-cell branching morphogenesis. Dev Biol 2021; 477:22-34. [PMID: 34004181 DOI: 10.1016/j.ydbio.2021.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/25/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022]
Abstract
Branching networks are a very common feature of multicellular animals and underlie the formation and function of numerous organs including the nervous system, the respiratory system, the vasculature and many internal glands. These networks range from subcellular structures such as dendritic trees to large multicellular tissues such as the lungs. The production of branched structures by single cells, so called subcellular branching, which has been better described in neurons and in cells of the respiratory and vascular systems, involves complex cytoskeletal remodelling events. In Drosophila, tracheal system terminal cells (TCs) and nervous system dendritic arborisation (da) neurons are good model systems for these subcellular branching processes. During development, the generation of subcellular branches by single-cells is characterized by extensive remodelling of the microtubule (MT) network and actin cytoskeleton, followed by vesicular transport and membrane dynamics. In this review, we describe the current knowledge on cytoskeletal regulation of subcellular branching, based on the terminal cells of the Drosophila tracheal system, but drawing parallels with dendritic branching and vertebrate vascular subcellular branching.
Collapse
Affiliation(s)
- Delia Ricolo
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain
| | - Judith Castro-Ribera
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain
| | - Sofia J Araújo
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona, 08028, Barcelona, Spain; Institute of Biomedicine University of Barcelona (IBUB), Barcelona, Spain.
| |
Collapse
|
23
|
Roly ZY, Godini R, Estermann MA, Major AT, Pocock R, Smith CA. Transcriptional landscape of the embryonic chicken Müllerian duct. BMC Genomics 2020; 21:688. [PMID: 33008304 PMCID: PMC7532620 DOI: 10.1186/s12864-020-07106-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Background Müllerian ducts are paired embryonic tubes that give rise to the female reproductive tract in vertebrates. Many disorders of female reproduction can be attributed to anomalies of Müllerian duct development. However, the molecular genetics of Müllerian duct formation is poorly understood and most disorders of duct development have unknown etiology. In this study, we describe for the first time the transcriptional landscape of the embryonic Müllerian duct, using the chicken embryo as a model system. RNA sequencing was conducted at 1 day intervals during duct formation to identify developmentally-regulated genes, validated by in situ hybridization. Results This analysis detected hundreds of genes specifically up-regulated during duct morphogenesis. Gene ontology and pathway analysis revealed enrichment for developmental pathways associated with cell adhesion, cell migration and proliferation, ERK and WNT signaling, and, interestingly, axonal guidance. The latter included factors linked to neuronal cell migration or axonal outgrowth, such as Ephrin B2, netrin receptor, SLIT1 and class A semaphorins. A number of transcriptional modules were identified that centred around key hub genes specifying matrix-associated signaling factors; SPOCK1, HTRA3 and ADGRD1. Several novel regulators of the WNT and TFG-β signaling pathway were identified in Müllerian ducts, including APCDD1 and DKK1, BMP3 and TGFBI. A number of novel transcription factors were also identified, including OSR1, FOXE1, PRICKLE1, TSHZ3 and SMARCA2. In addition, over 100 long non-coding RNAs (lncRNAs) were expressed during duct formation. Conclusions This study provides a rich resource of new candidate genes for Müllerian duct development and its disorders. It also sheds light on the molecular pathways engaged during tubulogenesis, a fundamental process in embryonic development.
Collapse
Affiliation(s)
- Zahida Yesmin Roly
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Rasoul Godini
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Martin A Estermann
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Andrew T Major
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Roger Pocock
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Craig A Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia.
| |
Collapse
|
24
|
Nazarnezhad S, Baino F, Kim HW, Webster TJ, Kargozar S. Electrospun Nanofibers for Improved Angiogenesis: Promises for Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1609. [PMID: 32824491 PMCID: PMC7466668 DOI: 10.3390/nano10081609] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/27/2022]
Abstract
Angiogenesis (or the development of new blood vessels) is a key event in tissue engineering and regenerative medicine; thus, a number of biomaterials have been developed and combined with stem cells and/or bioactive molecules to produce three-dimensional (3D) pro-angiogenic constructs. Among the various biomaterials, electrospun nanofibrous scaffolds offer great opportunities for pro-angiogenic approaches in tissue repair and regeneration. Nanofibers made of natural and synthetic polymers are often used to incorporate bioactive components (e.g., bioactive glasses (BGs)) and load biomolecules (e.g., vascular endothelial growth factor (VEGF)) that exert pro-angiogenic activity. Furthermore, seeding of specific types of stem cells (e.g., endothelial progenitor cells) onto nanofibrous scaffolds is considered as a valuable alternative for inducing angiogenesis. The effectiveness of these strategies has been extensively examined both in vitro and in vivo and the outcomes have shown promise in the reconstruction of hard and soft tissues (mainly bone and skin, respectively). However, the translational of electrospun scaffolds with pro-angiogenic molecules or cells is only at its beginning, requiring more research to prove their usefulness in the repair and regeneration of other highly-vascularized vital tissues and organs. This review will cover the latest progress in designing and developing pro-angiogenic electrospun nanofibers and evaluate their usefulness in a tissue engineering and regenerative medicine setting.
Collapse
Affiliation(s)
- Simin Nazarnezhad
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran;
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Hae-Won Kim
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Korea;
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan 31116, Korea
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA;
| | - Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran;
| |
Collapse
|
25
|
Kirkegaard JB, Nielsen BF, Trusina A, Sneppen K. Self-assembly, buckling and density-invariant growth of three-dimensional vascular networks. J R Soc Interface 2019; 16:20190517. [PMID: 31640503 PMCID: PMC6833333 DOI: 10.1098/rsif.2019.0517] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/02/2019] [Indexed: 12/19/2022] Open
Abstract
The experimental actualization of organoids modelling organs from brains to pancreases has revealed that much of the diverse morphologies of organs are emergent properties of simple intercellular 'rules' and not the result of top-down orchestration. In contrast to other organs, the initial plexus of the vascular system is formed by aggregation of cells in the process known as vasculogenesis. Here we study this self-assembling process of blood vessels in three dimensions through a set of simple rules that align intercellular apical-basal and planar cell polarity. We demonstrate that a fully connected network of tubes emerges above a critical initial density of cells. Through planar cell polarity, our model demonstrates convergent extension, and this polarity furthermore allows for both morphology-maintaining growth and growth-induced buckling. We compare this buckling with the special vasculature of the islets of Langerhans in the pancreas and suggest that the mechanism behind the vascular density-maintaining growth of these islets could be the result of growth-induced buckling.
Collapse
|
26
|
Mittal N, Yoon SH, Enomoto H, Hiroshi M, Shimizu A, Kawakami A, Fujita M, Watanabe H, Fukuda K, Makino S. Versican is crucial for the initiation of cardiovascular lumen development in medaka (Oryzias latipes). Sci Rep 2019; 9:9475. [PMID: 31263118 PMCID: PMC6603046 DOI: 10.1038/s41598-019-45851-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 06/13/2019] [Indexed: 12/17/2022] Open
Abstract
Versican is an evolutionary conserved extracellular matrix proteoglycan, and versican expression loss in mice results in embryonic lethality owing to cardiovascular defects. However, the in utero development of mammals limits our understanding of the precise role of versican during cardiovascular development. Therefore, the use of evolutionarily distant species that develop ex utero is more suitable for studying the mechanistic basis of versican activity. We performed ENU mutagenesis screening to identify medaka mutants with defects in embryonic cardiovascular development. In this study, we described a recessive point mutation in the versican 3'UTR resulting in reduced versican protein expression. The fully penetrant homozygous mutant showed termination of cardiac development at the linear heart tube stage and exhibited absence of cardiac looping, a constricted outflow tract, and no cardiac jelly. Additionally, progenitor cells did not migrate from the secondary source towards the arterial pole of the linear heart tube, resulting in a constricted outflow tract. Furthermore, mutants lacked blood flow and vascular lumen despite continuous peristaltic heartbeats. These results enhance our understanding of the mechanistic basis of versican in cardiac development, and this mutant represents a novel genetic model to investigate the mechanisms of vascular tubulogenesis.
Collapse
Affiliation(s)
- Nishant Mittal
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Sung Han Yoon
- Department of Interventional Cardiology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, AHSP A9229, Los Angeles, CA, 90048, USA
| | - Hirokazu Enomoto
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Miyama Hiroshi
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Atsushi Shimizu
- Division of Biomedical Information Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-cho, Shiwa-gun, Iwate, 028-3694, Japan
| | - Atsushi Kawakami
- Department of Biological Information, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama, 226-8501, Japan
| | - Misato Fujita
- Department of Biological Science, Graduate School of Science, Kanagawa University, 2946 Tsuchiya, Hiratsuka-Shi, Kanagawa-Ken, 259-1293, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, 1-, Yazakokarimata, Nagakute, Aichi, 480-1195, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shinji Makino
- Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan.
- Keio University Health Centre, 35-Shinanomachi Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
27
|
Barlow HR, Cleaver O. Building Blood Vessels-One Rho GTPase at a Time. Cells 2019; 8:cells8060545. [PMID: 31174284 PMCID: PMC6627795 DOI: 10.3390/cells8060545] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/01/2019] [Accepted: 06/03/2019] [Indexed: 01/09/2023] Open
Abstract
Blood vessels are required for the survival of any organism larger than the oxygen diffusion limit. Blood vessel formation is a tightly regulated event and vessel growth or changes in permeability are linked to a number of diseases. Elucidating the cell biology of endothelial cells (ECs), which are the building blocks of blood vessels, is thus critical to our understanding of vascular biology and to the development of vascular-targeted disease treatments. Small GTPases of the Rho GTPase family are known to regulate several processes critical for EC growth and maintenance. In fact, many of the 21 Rho GTPases in mammals are known to regulate EC junctional remodeling, cell shape changes, and other processes. Rho GTPases are thus an attractive target for disease treatments, as they often have unique functions in specific vascular cell types. In fact, some Rho GTPases are even expressed with relative specificity in diseased vessels. Interestingly, many Rho GTPases are understudied in ECs, despite their known expression in either developing or mature vessels, suggesting an even greater wealth of knowledge yet to be gleaned from these complex signaling pathways. This review aims to provide an overview of Rho GTPase signaling contributions to EC vasculogenesis, angiogenesis, and mature vessel barrier function. A particular emphasis is placed on so-called "alternative" Rho GTPases, as they are largely understudied despite their likely important contributions to EC biology.
Collapse
Affiliation(s)
- Haley Rose Barlow
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Ondine Cleaver
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
28
|
Possible cooption of a VEGF-driven tubulogenesis program for biomineralization in echinoderms. Proc Natl Acad Sci U S A 2019; 116:12353-12362. [PMID: 31152134 DOI: 10.1073/pnas.1902126116] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Biomineralization is the process by which living organisms use minerals to form hard structures that protect and support them. Biomineralization is believed to have evolved rapidly and independently in different phyla utilizing preexisting components. The mechanistic understanding of the regulatory networks that drive biomineralization and their evolution is far from clear. Sea urchin skeletogenesis is an excellent model system for studying both gene regulation and mineral uptake and deposition. The sea urchin calcite spicules are formed within a tubular cavity generated by the skeletogenic cells controlled by vascular endothelial growth factor (VEGF) signaling. The VEGF pathway is essential for biomineralization in echinoderms, while in many other phyla, across metazoans, it controls tubulogenesis and vascularization. Despite the critical role of VEGF signaling in sea urchin spiculogenesis, the downstream program it activates was largely unknown. Here we study the cellular and molecular machinery activated by the VEGF pathway during sea urchin spiculogenesis and reveal multiple parallels to the regulation of vertebrate vascularization. Human VEGF rescues sea urchin VEGF knockdown, vesicle deposition into an internal cavity plays a significant role in both systems, and sea urchin VEGF signaling activates hundreds of genes, including biomineralization and interestingly, vascularization genes. Moreover, five upstream transcription factors and three signaling genes that drive spiculogenesis are homologous to vertebrate factors that control vascularization. Overall, our findings suggest that sea urchin spiculogenesis and vertebrate vascularization diverged from a common ancestral tubulogenesis program, broadly adapted for vascularization and specifically coopted for biomineralization in the echinoderm phylum.
Collapse
|
29
|
Augustine R, Prasad P, Khalaf IMN. Therapeutic angiogenesis: From conventional approaches to recent nanotechnology-based interventions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 97:994-1008. [DOI: 10.1016/j.msec.2019.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/06/2018] [Accepted: 01/02/2019] [Indexed: 12/27/2022]
|
30
|
Abu Rmilah A, Zhou W, Nelson E, Lin L, Amiot B, Nyberg SL. Understanding the marvels behind liver regeneration. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 8:e340. [PMID: 30924280 DOI: 10.1002/wdev.340] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/18/2019] [Accepted: 02/22/2019] [Indexed: 02/06/2023]
Abstract
Tissue regeneration is a process by which the remaining cells of an injured organ regrow to offset the missed cells. This field is relatively a new discipline that has been a focus of intense research by clinicians, surgeons, and scientists for decades. It constitutes the cornerstone of tissue engineering, creation of artificial organs, and generation and utilization of therapeutic stem cells to undergo transformation to different types of mature cells. Many medical experts, scientists, biologists, and bioengineers have dedicated their efforts to deeply comprehend the process of liver regeneration, striving for harnessing it to invent new therapies for liver failure. Liver regeneration after partial hepatectomy in rodents has been extensively studied by researchers for many years. It is divided into three important distinctive phases including (a) Initiation or priming phase which includes an overexpression of specific genes to prepare the liver cells for replication, (b) Proliferation phase in which the liver cells undergo a series of cycles of cell division and expansion and finally, (c) termination phase which acts as brake to stop the regenerative process and prevent the liver tissue overgrowth. These events are well controlled by cytokines, growth factors, and signaling pathways. In this review, we describe the function, embryology, and anatomy of human liver, discuss the molecular basis of liver regeneration, elucidate the hepatocyte and cholangiocyte lineages mediating this process, explain the role of hepatic progenitor cells and elaborate the developmental signaling pathways and regulatory molecules required to procure a complete restoration of hepatic lobule. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration Signaling Pathways > Global Signaling Mechanisms Gene Expression and Transcriptional Hierarchies > Cellular Differentiation.
Collapse
Affiliation(s)
- Anan Abu Rmilah
- Department of Surgery, Division of Transplant Surgery, Mayo Clinic, Rochester, Minnesota
| | - Wei Zhou
- Department of Surgery, Division of Transplant Surgery, Mayo Clinic, Rochester, Minnesota
| | - Erek Nelson
- Department of Surgery, Division of Transplant Surgery, Mayo Clinic, Rochester, Minnesota
| | - Li Lin
- Department of Surgery, Division of Transplant Surgery, Mayo Clinic, Rochester, Minnesota
| | - Bruce Amiot
- Department of Surgery, Division of Transplant Surgery, Mayo Clinic, Rochester, Minnesota
| | - Scott L Nyberg
- Department of Surgery, Division of Transplant Surgery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
31
|
Bezenah JR, Rioja AY, Juliar B, Friend N, Putnam AJ. Assessing the ability of human endothelial cells derived from induced-pluripotent stem cells to form functional microvasculature in vivo. Biotechnol Bioeng 2019; 116:415-426. [PMID: 30414271 PMCID: PMC6322937 DOI: 10.1002/bit.26860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/22/2018] [Accepted: 11/07/2018] [Indexed: 12/26/2022]
Abstract
Forming functional blood vessel networks is a major clinical challenge in the fields of tissue engineering and therapeutic angiogenesis. Cell-based strategies to promote neovascularization have been widely explored, but cell sourcing remains a significant limitation. Induced-pluripotent stem cell-derived endothelial cells (iPSC-ECs) are a promising, potentially autologous, alternative cell source. However, it is unclear whether iPSC-ECs form the same robust microvasculature in vivo documented for other EC sources. In this study, we utilized a well-established in vivo model, in which ECs (iPSC-EC or human umbilical vein endothelial cells [HUVEC]) were coinjected with normal human lung fibroblasts (NHLFs) and a fibrin matrix into the dorsal flank of severe combined immunodeficiency mice to assess their ability to form functional microvasculature. Qualitatively, iPSC-ECs were capable of vessel formation and perfusion and demonstrated similar vessel morphologies to HUVECs. However, quantitatively, iPSC-ECs exhibited a two-fold reduction in vessel density and a three-fold reduction in the number of perfused vessels compared with HUVECs. Further analysis revealed the presence of collagen-IV and α-smooth muscle actin were significantly lower around iPSC-EC/NHLF vasculature than in HUVEC/NHLF implants, suggesting reduced vessel maturity. Collectively, these results demonstrate the need for increased iPSC-EC maturation for clinical translation to be realized.
Collapse
Affiliation(s)
- Jonathan R. Bezenah
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| | - Ana Y. Rioja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| | - Benjamin Juliar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| | - Nicole Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| | - Andrew J. Putnam
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA 48109
| |
Collapse
|
32
|
De Moor L, Merovci I, Baetens S, Verstraeten J, Kowalska P, Krysko DV, De Vos WH, Declercq H. High-throughput fabrication of vascularized spheroids for bioprinting. Biofabrication 2018; 10:035009. [PMID: 29798932 DOI: 10.1088/1758-5090/aac7e6] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Overcoming the problem of vascularization remains the main challenge in the field of tissue engineering. As three-dimensional (3D) bioprinting is the rising technique for the fabrication of large tissue constructs, small prevascularized building blocks were generated that can be incorporated throughout a printed construct, answering the need for a microvasculature within the small micron range (<10 μm). Uniform spheroids with an ideal geometry and diameter for bioprinting were formed, using a high-throughput non-adhesive agarose microwell system. Since monoculture spheroids of endothelial cells were unable to remain stable, coculture spheroids combining endothelial cells with fibroblasts and/or adipose tissue derived mesenchymal stem cells (ADSC) as supporting cells, were created. When applying the favorable coculture ratio, viable spheroids were obtained and endothelial cells spontaneously formed a capillary-like network and lumina, as shown by immunohistochemistry and transmission electron microscopy. Especially the presence of ADSC led to a higher vascularization and extracellular matrix production of the microtissue. Moreover, spheroids were able to assemble at random in suspension and in a hydrogel, creating a macrotissue. During at random assembly, cells reorganized, creating a branched capillary-network throughout the entire fused construct by inoculating with capillaries of adjacent spheroids. Combining the advantage of this natural capacity of microtissues to self-assemble and the controlled organization by bioprinting technologies, these prevascularized spheroids can be useful as building blocks for the engineering of large vascularized 3D tissues.
Collapse
Affiliation(s)
- Lise De Moor
- Tissue engineering and Biomaterials Group, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Paragangliomas arise through an autonomous vasculo-angio-neurogenic program inhibited by imatinib. Acta Neuropathol 2018; 135:779-798. [PMID: 29305721 PMCID: PMC5904229 DOI: 10.1007/s00401-017-1799-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 12/16/2017] [Accepted: 12/21/2017] [Indexed: 12/26/2022]
Abstract
Tumours can be viewed as aberrant tissues or organs sustained by tumorigenic stem-like cells that engage into dysregulated histo/organogenetic processes. Paragangliomas, prototypical organoid tumours constituted by dysmorphic variants of the vascular and neural tissues found in normal paraganglia, provide a model to test this hypothesis. To understand the origin of paragangliomas, we built a biobank comprising 77 cases, 18 primary cultures, 4 derived cell lines, 80 patient-derived xenografts and 11 cell-derived xenografts. We comparatively investigated these unique complementary materials using morphofunctional, ultrastructural and flow cytometric assays accompanied by microRNA studies. We found that paragangliomas contain stem-like cells with hybrid mesenchymal/vasculoneural phenotype, stabilized and expanded in the derived cultures. The viability and growth of such cultures depended on the downregulation of the miR-200 and miR-34 families, which allowed high PDGFRA and ZEB1 protein expression levels. Both tumour tissue- and cell culture-derived xenografts recapitulated the vasculoneural paraganglioma structure and arose from mesenchymal-like cells through a fixed developmental sequence. First, vasculoangiogenesis organized the microenvironment, building a perivascular niche which in turn supported neurogenesis. Neuroepithelial differentiation was associated with severe mitochondrial dysfunction, not present in cultured paraganglioma cells, but acquired in vivo during xenograft formation. Vasculogenesis was the Achilles’ heel of xenograft development. In fact, imatinib, that targets endothelial-mural signalling, blocked paraganglioma xenograft formation (11 xenografts from 12 cell transplants in the control group versus 2 out of 10 in the treated group, P = 0.0015). Overall our key results were unaffected by the SDHx gene carrier status of the patient, characterized for 70 out of 77 cases. In conclusion, we explain the biphasic vasculoneural structure of paragangliomas and identify an early and pharmacologically actionable phase of paraganglioma organization.
Collapse
|
34
|
Abstract
The zebrafish is an outstanding model for studying vascular biology in vivo. Pericytes and vascular smooth muscle cells can be imaged as they associate with vessels and provide stability and integrity to the vasculature. In zebrafish, pericytes associate with the cerebral and trunk vasculature on the second day of development, as assayed by pdgfrβ and notch3 markers. In the head, cerebral pericytes are neural crest derived, except for the pericytes of the hindbrain vasculature, which are mesoderm derived. Similar to the hindbrain, pericytes on the trunk vasculature are also mesoderm derived. Regardless of their location, pericyte development depends on a complex interaction between blood flow and signalling pathways, such as Notch, SONIC HEDGEHOG and BMP signalling, all of which positively regulate pericyte numbers.Pericyte numbers rapidly increase as development proceeds in order to stabilize both the blood-brain barrier and the vasculature and hence, prevent haemorrhage. Consequently, compromised pericyte development results in compromised vascular integrity, which then evolves into detrimental pathologies. Some of these pathologies have been modelled in zebrafish by inducing mutations in the notch3, foxc1 and foxf2 genes. These zebrafish models provide insights into the mechanisms of disease as associated with pericyte biology. Going forward, these models may be key contributors in elucidating the role of vascular mural cells in regulating vessel diameter and hence, blood flow.
Collapse
Affiliation(s)
- Nabila Bahrami
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Sarah J Childs
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
35
|
Nesmith JE, Chappell JC, Cluceru JG, Bautch VL. Blood vessel anastomosis is spatially regulated by Flt1 during angiogenesis. Development 2017; 144:889-896. [PMID: 28246215 DOI: 10.1242/dev.145672] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022]
Abstract
Blood vessel formation is essential for vertebrate development and is primarily achieved by angiogenesis - endothelial cell sprouting from pre-existing vessels. Vessel networks expand when sprouts form new connections, a process whose regulation is poorly understood. Here, we show that vessel anastomosis is spatially regulated by Flt1 (VEGFR1), a VEGFA receptor that acts as a decoy receptor. In vivo, expanding vessel networks favor interactions with Flt1 mutant mouse endothelial cells. Live imaging in human endothelial cells in vitro revealed that stable connections are preceded by transient contacts from extending sprouts, suggesting sampling of potential target sites, and lowered Flt1 levels reduced transient contacts and increased VEGFA signaling. Endothelial cells at target sites with reduced Flt1 and/or elevated protrusive activity were more likely to form stable connections with incoming sprouts. Target cells with reduced membrane-localized Flt1 (mFlt1), but not soluble Flt1, recapitulated the bias towards stable connections, suggesting that relative mFlt1 expression spatially influences the selection of stable connections. Thus, sprout anastomosis parameters are regulated by VEGFA signaling, and stable connections are spatially regulated by endothelial cell-intrinsic modulation of mFlt1, suggesting new ways to manipulate vessel network formation.
Collapse
Affiliation(s)
- Jessica E Nesmith
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John C Chappell
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Julia G Cluceru
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Victoria L Bautch
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA .,Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
36
|
Kim DJ, Norden PR, Salvador J, Barry DM, Bowers SLK, Cleaver O, Davis GE. Src- and Fyn-dependent apical membrane trafficking events control endothelial lumen formation during vascular tube morphogenesis. PLoS One 2017; 12:e0184461. [PMID: 28910325 PMCID: PMC5598984 DOI: 10.1371/journal.pone.0184461] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/24/2017] [Indexed: 01/10/2023] Open
Abstract
Here we examine the question of how endothelial cells (ECs) develop their apical membrane surface domain during lumen and tube formation. We demonstrate marked apical membrane targeting of activated Src kinases to this apical domain during early and late stages of this process. Immunostaining for phosphotyrosine or phospho-Src reveals apical membrane staining in intracellular vacuoles initially. This is then followed by vacuole to vacuole fusion events to generate an apical luminal membrane, which is similarly decorated with activated phospho-Src kinases. Functional blockade of Src kinases completely blocks EC lumen and tube formation, whether this occurs during vasculogenic tube assembly or angiogenic sprouting events. Multiple Src kinases participate in this apical membrane formation process and siRNA suppression of Src, Fyn and Yes, but not Lyn, blocks EC lumen formation. We also demonstrate strong apical targeting of Src-GFP and Fyn-GFP fusion proteins and increasing their expression enhances lumen formation. Finally, we show that Src- and Fyn-associated vacuoles track and fuse along a subapically polarized microtubule cytoskeleton, which is highly acetylated. These vacuoles generate the apical luminal membrane in a stereotypically polarized, perinuclear position. Overall, our study identifies a critical role for Src kinases in creating and decorating the EC apical membrane surface during early and late stages of lumen and tube formation, a central event in the molecular control of vascular morphogenesis.
Collapse
Affiliation(s)
- Dae Joong Kim
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - Pieter R Norden
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - Jocelynda Salvador
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - David M Barry
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas TX, United States of America
| | - Stephanie L K Bowers
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - Ondine Cleaver
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas TX, United States of America
| | - George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| |
Collapse
|
37
|
Boutin ME, Kramer LL, Livi LL, Brown T, Moore C, Hoffman-Kim D. A three-dimensional neural spheroid model for capillary-like network formation. J Neurosci Methods 2017; 299:55-63. [PMID: 28143748 DOI: 10.1016/j.jneumeth.2017.01.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 01/19/2017] [Accepted: 01/24/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND In vitro three-dimensional neural spheroid models have an in vivo-like cell density, and have the potential to reduce animal usage and increase experimental throughput. The aim of this study was to establish a spheroid model to study the formation of capillary-like networks in a three-dimensional environment that incorporates both neuronal and glial cell types, and does not require exogenous vasculogenic growth factors. NEW METHOD We created self-assembled, scaffold-free cellular spheroids using primary-derived postnatal rodent cortex as a cell source. The interactions between relevant neural cell types, basement membrane proteins, and endothelial cells were characterized by immunohistochemistry. Transmission electron microscopy was used to determine if endothelial network structures had lumens. RESULTS Endothelial cells within cortical spheroids assembled into capillary-like networks with lumens. Networks were surrounded by basement membrane proteins, including laminin, fibronectin and collagen IV, as well as key neurovascular cell types. COMPARISON WITH EXISTING METHOD(S) Existing in vitro models of the cortical neurovascular environment study monolayers of endothelial cells, either on transwell inserts or coating cellular spheroids. These models are not well suited to study vasculogenesis, a process hallmarked by endothelial cell cord formation and subsequent lumenization. CONCLUSIONS The neural spheroid is a new model to study the formation of endothelial cell capillary-like structures in vitro within a high cell density three-dimensional environment that contains both neuronal and glial populations. This model can be applied to investigate vascular assembly in healthy or disease states, such as stroke, traumatic brain injury, or neurodegenerative disorders.
Collapse
Affiliation(s)
- Molly E Boutin
- Center for Biomedical Engineering, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Department of Molecular Pharmacology, Physiology, and Biotechnology, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Liana L Kramer
- Department of Molecular Pharmacology, Physiology, and Biotechnology, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Liane L Livi
- Center for Biomedical Engineering, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Department of Molecular Pharmacology, Physiology, and Biotechnology, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Tyler Brown
- Department of Neuroscience, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Brown Institute for Brain Science, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Christopher Moore
- Department of Neuroscience, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Brown Institute for Brain Science, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| | - Diane Hoffman-Kim
- Center for Biomedical Engineering, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Department of Molecular Pharmacology, Physiology, and Biotechnology, 175 Meeting Street, Brown University, Providence, RI, 02912, United States; Brown Institute for Brain Science, 175 Meeting Street, Brown University, Providence, RI, 02912, United States.
| |
Collapse
|
38
|
Calderon GA, Thai P, Hsu CW, Grigoryan B, Gibson SM, Dickinson ME, Miller JS. Tubulogenesis of co-cultured human iPS-derived endothelial cells and human mesenchymal stem cells in fibrin and gelatin methacrylate gels. Biomater Sci 2017; 5:1652-1660. [DOI: 10.1039/c7bm00223h] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Here, we investigate the tubulogenic potential of commercially-sourced iPS-ECs with and without supporting commercially-sourced hMSCs within 3D natural fibrin or semi-synthetic gelatin methacrylate (GelMA) hydrogels.
Collapse
Affiliation(s)
| | - P. Thai
- Department of Bioengineering
- Rice University
- Houston
- USA
| | - C. W. Hsu
- Department of Molecular Physiology and Biophysics
- Baylor College of Medicine
- Houston
- USA
| | - B. Grigoryan
- Department of Bioengineering
- Rice University
- Houston
- USA
| | - S. M. Gibson
- Department of Bioengineering
- Rice University
- Houston
- USA
- Department of Molecular Physiology and Biophysics
| | - M. E. Dickinson
- Department of Molecular Physiology and Biophysics
- Baylor College of Medicine
- Houston
- USA
| | - J. S. Miller
- Department of Bioengineering
- Rice University
- Houston
- USA
| |
Collapse
|
39
|
Chappell JC, Cluceru JG, Nesmith JE, Mouillesseaux KP, Bradley VB, Hartland CM, Hashambhoy-Ramsay YL, Walpole J, Peirce SM, Mac Gabhann F, Bautch VL. Flt-1 (VEGFR-1) coordinates discrete stages of blood vessel formation. Cardiovasc Res 2016; 111:84-93. [PMID: 27142980 DOI: 10.1093/cvr/cvw091] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 03/03/2016] [Indexed: 01/09/2023] Open
Abstract
AIMS In developing blood vessel networks, the overall level of vessel branching often correlates with angiogenic sprout initiations, but in some pathological situations, increased sprout initiations paradoxically lead to reduced vessel branching and impaired vascular function. We examine the hypothesis that defects in the discrete stages of angiogenesis can uniquely contribute to vessel branching outcomes. METHODS AND RESULTS Time-lapse movies of mammalian blood vessel development were used to define and quantify the dynamics of angiogenic sprouting. We characterized the formation of new functional conduits by classifying discrete sequential stages-sprout initiation, extension, connection, and stability-that are differentially affected by manipulation of vascular endothelial growth factor-A (VEGF-A) signalling via genetic loss of the receptor flt-1 (vegfr1). In mouse embryonic stem cell-derived vessels genetically lacking flt-1, overall branching is significantly decreased while sprout initiations are significantly increased. Flt-1(-/-) mutant sprouts are less likely to retract, and they form increased numbers of connections with other vessels. However, loss of flt-1 also leads to vessel collapse, which reduces the number of new stable conduits. Computational simulations predict that loss of flt-1 results in ectopic Flk-1 signalling in connecting sprouts post-fusion, causing protrusion of cell processes into avascular gaps and collapse of branches. Thus, defects in stabilization of new vessel connections offset increased sprout initiations and connectivity in flt-1(-/-) vascular networks, with an overall outcome of reduced numbers of new conduits. CONCLUSIONS These results show that VEGF-A signalling has stage-specific effects on vascular morphogenesis, and that understanding these effects on dynamic stages of angiogenesis and how they integrate to expand a vessel network may suggest new therapeutic strategies.
Collapse
Affiliation(s)
- John C Chappell
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, Roanoke, VA 24014, USA
| | - Julia G Cluceru
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica E Nesmith
- Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kevin P Mouillesseaux
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Vanessa B Bradley
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, Roanoke, VA 24014, USA
| | - Caitlin M Hartland
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, Roanoke, VA 24014, USA
| | - Yasmin L Hashambhoy-Ramsay
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Joseph Walpole
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Feilim Mac Gabhann
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Victoria L Bautch
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
40
|
Park-Windhol C, D'Amore PA. Disorders of Vascular Permeability. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:251-81. [PMID: 26907525 DOI: 10.1146/annurev-pathol-012615-044506] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The endothelial barrier maintains vascular and tissue homeostasis and modulates many physiological processes, such as angiogenesis. Vascular barrier integrity can be disrupted by a variety of soluble permeability factors, and changes in barrier function can exacerbate tissue damage during disease progression. Understanding endothelial barrier function is critical for vascular homeostasis. Many of the signaling pathways promoting vascular permeability can also be triggered during disease, resulting in prolonged or uncontrolled vascular leak. It is believed that recovery of the normal vasculature requires diminishing this hyperpermeable state. Although the molecular mechanisms governing vascular leak have been studied over the last few decades, recent advances have identified new therapeutic targets that have begun to show preclinical and clinical promise. These approaches have been successfully applied to an increasing number of disease conditions. New perspectives regarding how vascular leak impacts the progression of various diseases are highlighted in this review.
Collapse
Affiliation(s)
- Cindy Park-Windhol
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114; , .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Patricia A D'Amore
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114; , .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115.,Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
41
|
Rasip1 is essential to blood vessel stability and angiogenic blood vessel growth. Angiogenesis 2016; 19:173-90. [PMID: 26897025 DOI: 10.1007/s10456-016-9498-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/03/2016] [Indexed: 01/09/2023]
Abstract
Cardiovascular function depends on patent, continuous and stable blood vessel formation by endothelial cells (ECs). Blood vessel development initiates by vasculogenesis, as ECs coalesce into linear aggregates and organize to form central lumens that allow blood flow. Molecular mechanisms underlying in vivo vascular 'tubulogenesis' are only beginning to be unraveled. We previously showed that the GTPase-interacting protein called Rasip1 is required for the formation of continuous vascular lumens in the early embryo. Rasip1(-/-) ECs exhibit loss of proper cell polarity and cell shape, disrupted localization of EC-EC junctions and defects in adhesion of ECs to extracellular matrix. In vitro studies showed that Rasip1 depletion in cultured ECs blocked tubulogenesis. Whether Rasip1 is required in blood vessels after their initial formation remained unclear. Here, we show that Rasip1 is essential for vessel formation and maintenance in the embryo, but not in quiescent adult vessels. Rasip1 is also required for angiogenesis in three models of blood vessel growth: in vitro matrix invasion, retinal blood vessel growth and directed in vivo angiogenesis assays. Rasip1 is thus necessary in growing embryonic blood vessels, postnatal angiogenic sprouting and remodeling, but is dispensable for maintenance of established blood vessels, making it a potential anti-angiogenic therapeutic target.
Collapse
|
42
|
Norden PR, Kim DJ, Barry DM, Cleaver OB, Davis GE. Cdc42 and k-Ras Control Endothelial Tubulogenesis through Apical Membrane and Cytoskeletal Polarization: Novel Stimulatory Roles for GTPase Effectors, the Small GTPases, Rac2 and Rap1b, and Inhibitory Influence of Arhgap31 and Rasa1. PLoS One 2016; 11:e0147758. [PMID: 26812085 PMCID: PMC4728208 DOI: 10.1371/journal.pone.0147758] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/07/2016] [Indexed: 01/18/2023] Open
Abstract
A critical and understudied property of endothelial cells is their ability to form lumens and tube networks. Although considerable information has been obtained concerning these issues, including the role of Cdc42 and Rac1 and their effectors such as Pak2, Pak4, Par6b, and co-regulators such as integrins, MT1-MMP and Par3; many key questions remain that are necessary to elucidate molecular and signaling requirements for this fundamental process. In this work, we identify new small GTPase regulators of EC tubulogenesis including k-Ras, Rac2 and Rap1b that act in conjunction with Cdc42 as well as the key downstream effectors, IQGAP1, MRCKβ, beta-Pix, GIT1, and Rasip1 (which can assemble into multiprotein complexes with key regulators including α2β1 integrin and MT1-MMP). In addition, we identify the negative regulators, Arhgap31 (by inactivating Cdc42 and Rac) and Rasa1 (by inactivating k-Ras) and the positive regulator, Arhgap29 (by inactivating RhoA) which play a major functional role during the EC tubulogenic process. Human EC siRNA suppression or mouse knockout of Rasip1 leads to identical phenotypes where ECs form extensive cord networks, but cannot generate lumens or tubes. Essential roles for these molecules during EC tubulogenesis include; i) establishment of asymmetric EC cytoskeletal polarization (subapical distribution of acetylated tubulin and basal membrane distribution of F-actin); and ii) directed membrane trafficking of pinocytic vacuoles or other intracellular vesicles along acetylated tubulin tracks to the developing apical membrane surface. Cdc42 co-localizes subapically with acetylated tubulin, while Rac1 and k-Ras strongly label vacuole/ vesicle membranes which accumulate and fuse together in a polarized, perinuclear manner. We observe polarized apical membrane and subapical accumulation of key GTPases and effectors regulating EC lumen formation including Cdc42, Rac1, Rac2, k-Ras, Rap1b, activated c-Raf and Rasip1 to control EC tube network assembly. Overall, this work defines novel key regulators and their functional roles during human EC tubulogenesis.
Collapse
Affiliation(s)
- Pieter R. Norden
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - Dae Joong Kim
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| | - David M. Barry
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - Ondine B. Cleaver
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, United States of America
| | - George E. Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Dalton Cardiovascular Research Center, Columbia, MO, United States of America
| |
Collapse
|
43
|
Freiman A, Shandalov Y, Rozenfeld D, Shor E, Segal S, Ben-David D, Meretzki S, Egozi D, Levenberg S. Adipose-derived endothelial and mesenchymal stem cells enhance vascular network formation on three-dimensional constructs in vitro. Stem Cell Res Ther 2016; 7:5. [PMID: 26753517 PMCID: PMC4709933 DOI: 10.1186/s13287-015-0251-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/23/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022] Open
Abstract
Background Adipose-derived mesenchymal stem cells (MSCs) have been gaining fame mainly due to their vast clinical potential, simple isolation methods and minimal donor site morbidity. Adipose-derived MSCs and microvascular endothelial cells have been shown to bear angiogenic and vasculogenic capabilities. We hypothesized that co-culture of human adipose-derived MSCs with human adipose-derived microvascular endothelial cells (HAMECs) will serve as an effective cell pair to induce angiogenesis and vessel-like network formation in three-dimensional scaffolds in vitro. Methods HAMECs or human umbilical vein endothelial cells (HUVECs) were co-cultured on scaffolds with either MSCs or human neonatal dermal fibroblasts. Cells were immunofluorescently stained within the scaffolds at different time points post-seeding. Various analyses were performed to determine vessel length, complexity and degree of maturity. Results The HAMEC:MSC combination yielded the most organized and complex vascular elements within scaffolds, and in the shortest period of time, when compared to the other tested cell combinations. These differences were manifested by higher network complexity, more tube alignment and higher α-smooth muscle actin expression. Moreover, these generated microvessels further matured and developed during the 14-day incubation period within the three-dimensional microenvironment. Conclusions These data demonstrate optimal vascular network formation upon co-culture of microvascular endothelial cells and adipose-derived MSCs in vitro and constitute a significant step in appreciation of the potential of microvascular endothelial cells and MSCs in different tissue engineering applications that can also be advantageous in in vivo studies. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0251-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alina Freiman
- Inter-departmental Program in Biotechnology, Technion-Israel Institute of Technology, Haifa, 32000, Israel. .,Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | - Yulia Shandalov
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | - Dekel Rozenfeld
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | - Erez Shor
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | - Sofia Segal
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| | | | | | - Dana Egozi
- Department of Plastic and Reconstructive Surgery, Kaplan Hospital, Rehovot, Israel.
| | - Shulamit Levenberg
- Biomedical Engineering Department, Technion-Israel Institute of Technology, Haifa, 32000, Israel.
| |
Collapse
|
44
|
Bowers S, Norden P, Davis G. Molecular Signaling Pathways Controlling Vascular Tube Morphogenesis and Pericyte-Induced Tube Maturation in 3D Extracellular Matrices. ADVANCES IN PHARMACOLOGY 2016; 77:241-80. [DOI: 10.1016/bs.apha.2016.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
45
|
Fish JE, Wythe JD. The molecular regulation of arteriovenous specification and maintenance. Dev Dyn 2015; 244:391-409. [PMID: 25641373 DOI: 10.1002/dvdy.24252] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/02/2015] [Accepted: 01/04/2015] [Indexed: 12/21/2022] Open
Abstract
The formation of a hierarchical vascular network, composed of arteries, veins, and capillaries, is essential for embryogenesis and is required for the production of new functional vasculature in the adult. Elucidating the molecular mechanisms that orchestrate the differentiation of vascular endothelial cells into arterial and venous cell fates is requisite for regenerative medicine, as the directed formation of perfused vessels is desirable in a myriad of pathological settings, such as in diabetes and following myocardial infarction. Additionally, this knowledge will enhance our understanding and treatment of vascular anomalies, such as arteriovenous malformations (AVMs). From studies in vertebrate model organisms, such as mouse, zebrafish, and chick, a number of key signaling pathways have been elucidated that are required for the establishment and maintenance of arterial and venous fates. These include the Hedgehog, Vascular Endothelial Growth Factor (VEGF), Transforming Growth Factor-β (TGF-β), Wnt, and Notch signaling pathways. In addition, a variety of transcription factor families acting downstream of, or in concert with, these signaling networks play vital roles in arteriovenous (AV) specification. These include Notch and Notch-regulated transcription factors (e.g., HEY and HES), SOX factors, Forkhead factors, β-Catenin, ETS factors, and COUP-TFII. It is becoming apparent that AV specification is a highly coordinated process that involves the intersection and carefully orchestrated activity of multiple signaling cascades and transcriptional networks. This review will summarize the molecular mechanisms that are involved in the acquisition and maintenance of AV fate, and will highlight some of the limitations in our current knowledge of the molecular machinery that directs AV morphogenesis.
Collapse
Affiliation(s)
- Jason E Fish
- Toronto General Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Canada
| | | |
Collapse
|
46
|
Investigating human vascular tube morphogenesis and maturation using endothelial cell-pericyte co-cultures and a doxycycline-inducible genetic system in 3D extracellular matrices. Methods Mol Biol 2015; 1189:171-89. [PMID: 25245694 DOI: 10.1007/978-1-4939-1164-6_12] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Considerable progress has occurred toward our understanding of the molecular basis for vascular morphogenesis, maturation, and stabilization. A major reason for this progress has been the development of novel in vitro systems to investigate these processes in 3D extracellular matrices. In this chapter, we present models of human endothelial cell (EC) tube formation and EC-pericyte tube co-assembly using serum-free defined conditions in 3D collagen matrices. We utilize both human venous and arterial ECs and show that both cell types readily form tubes and induce pericyte recruitment and both ECs and pericytes work together to remodel the extracellular matrix environment by assembling the vascular basement membrane, a key step in capillary tube network maturation and stabilization. Importantly, we have shown that these events occur under serum-free defined conditions using the hematopoietic stem cell cytokines, SCF, IL-3, and SDF-1α and also including FGF-2. In contrast, the combination of VEGF and FGF-2 fails to support vascular tube morphogenesis or pericyte-induced tube maturation under the same serum-free defined conditions. Furthermore, we present novel assays whereby we have developed both human ECs and pericytes to induce specific genes using a doxycycline-regulated lentiviral system. In this manner, we can upregulate the expression of wild-type or mutant gene products at any stage of vascular morphogenesis or maturation in 3D matrices. These in vitro experimental approaches will continue to identify key molecular requirements and signaling pathways that control fundamental events in tissue vascularization under normal or pathologic conditions. Furthermore, these models will provide new insights into the development of novel disease therapeutic approaches where vascularization is an important pathogenic component and create new ways to assemble capillary tube networks with associated pericytes for tissue engineering applications.
Collapse
|
47
|
Bichsel CA, Hall SR, Schmid RA, Guenat OT, Geiser T. Primary Human Lung Pericytes Support and Stabilize In Vitro Perfusable Microvessels. Tissue Eng Part A 2015; 21:2166-76. [DOI: 10.1089/ten.tea.2014.0545] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Colette A. Bichsel
- Lung Regeneration Technologies, ARTORG Center, University of Bern, Bern, Switzerland
- Division of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Sean R.R. Hall
- Division of Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Ralph A. Schmid
- Division of Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Olivier T. Guenat
- Lung Regeneration Technologies, ARTORG Center, University of Bern, Bern, Switzerland
- Division of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
- Division of Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
| | - Thomas Geiser
- Division of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
- Department of Clinical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
48
|
Simons M, Alitalo K, Annex BH, Augustin HG, Beam C, Berk BC, Byzova T, Carmeliet P, Chilian W, Cooke JP, Davis GE, Eichmann A, Iruela-Arispe ML, Keshet E, Sinusas AJ, Ruhrberg C, Woo YJ, Dimmeler S. State-of-the-Art Methods for Evaluation of Angiogenesis and Tissue Vascularization: A Scientific Statement From the American Heart Association. Circ Res 2015; 116:e99-132. [PMID: 25931450 DOI: 10.1161/res.0000000000000054] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
49
|
Charpentier MS, Tandon P, Trincot CE, Koutleva EK, Conlon FL. A distinct mechanism of vascular lumen formation in Xenopus requires EGFL7. PLoS One 2015; 10:e0116086. [PMID: 25705891 PMCID: PMC4338030 DOI: 10.1371/journal.pone.0116086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 12/04/2014] [Indexed: 01/03/2023] Open
Abstract
During vertebrate blood vessel development, lumen formation is the critical process by which cords of endothelial cells transition into functional tubular vessels. Here, we use Xenopus embryos to explore the cellular and molecular mechanisms underlying lumen formation of the dorsal aorta and the posterior cardinal veins, the primary major vessels that arise via vasculogenesis within the first 48 hours of life. We demonstrate that endothelial cells are initially found in close association with one another through the formation of tight junctions expressing ZO-1. The emergence of vascular lumens is characterized by elongation of endothelial cell shape, reorganization of junctions away from the cord center to the periphery of the vessel, and onset of Claudin-5 expression within tight junctions. Furthermore, unlike most vertebrate vessels that exhibit specialized apical and basal domains, we show that early Xenopus vessels are not polarized. Moreover, we demonstrate that in embryos depleted of the extracellular matrix factor Epidermal Growth Factor-Like Domain 7 (EGFL7), an evolutionarily conserved factor associated with vertebrate vessel development, vascular lumens fail to form. While Claudin-5 localizes to endothelial tight junctions of EGFL7-depleted embryos in a timely manner, endothelial cells of the aorta and veins fail to undergo appropriate cell shape changes or clear junctions from the cell-cell contact. Taken together, we demonstrate for the first time the mechanisms by which lumens are generated within the major vessels in Xenopus and implicate EGFL7 in modulating cell shape and cell-cell junctions to drive proper lumen morphogenesis.
Collapse
Affiliation(s)
- Marta S. Charpentier
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Panna Tandon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Claire E. Trincot
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Elitza K. Koutleva
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Frank L. Conlon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, UNC-CH, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
50
|
Davis GE, Norden PR, Bowers SLK. Molecular control of capillary morphogenesis and maturation by recognition and remodeling of the extracellular matrix: functional roles of endothelial cells and pericytes in health and disease. Connect Tissue Res 2015; 56:392-402. [PMID: 26305158 PMCID: PMC4765926 DOI: 10.3109/03008207.2015.1066781] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
This review addresses fundamental mechanisms underlying how capillaries form in three-dimensional extracellular matrices and how endothelial cells (ECs) and pericytes co-assemble to form capillary networks. In addition to playing a critical role in supplying oxygen and nutrients to tissues, recent work suggests that blood vessels supply important signals to facilitate tissue development. Here, we hypothesize that another major function of capillaries is to supply signals to suppress major disease mechanisms including inflammation, infection, thrombosis, hemorrhage, edema, ischemic injury, fibrosis, autoimmune disease and tumor growth/progression. Capillary dysfunction plays a key pathogenic role in many human diseases, and thus, this suppressing function may be attenuated and central toward the initiation and progression of disease. We describe how capillaries form through creation of EC-lined tube networks and vascular guidance tunnels in 3D extracellular matrices. Pericytes recruit to the abluminal EC tube surface within these tunnel spaces, and work together to assemble the vascular basement membrane matrix. These processes occur under serum-free conditions in 3D collagen or fibrin matrices and in response to five key growth factors which are stem cell factor, interleukin-3, stromal-derived factor-1α, fibroblast growth factor-2 and insulin. In addition, we identified a key role for EC-derived platelet-derived growth factor-BB and heparin-binding epidermal growth factor in pericyte recruitment and proliferation to promote EC-pericyte tube co-assembly and vascular basement membrane matrix deposition. A molecular understanding of capillary morphogenesis and maturation should lead to novel therapeutic strategies to repair capillary dysfunction in major human disease contexts including cancer and diabetes.
Collapse
Affiliation(s)
- George E Davis
- a Department of Medical Pharmacology and Physiology , Dalton Cardiovascular Research Center, University of Missouri School of Medicine , Columbia , MO , USA
| | - Pieter R Norden
- a Department of Medical Pharmacology and Physiology , Dalton Cardiovascular Research Center, University of Missouri School of Medicine , Columbia , MO , USA
| | - Stephanie L K Bowers
- a Department of Medical Pharmacology and Physiology , Dalton Cardiovascular Research Center, University of Missouri School of Medicine , Columbia , MO , USA
| |
Collapse
|