1
|
Donzanti MJ, Ferrick BJ, Mhatre O, Chernokal B, Renteria DC, Gleghorn JP. Stochastic to Deterministic: A Straightforward Approach to Create Serially Perfusable Multiscale Capillary Beds. ACS Biomater Sci Eng 2025; 11:239-248. [PMID: 39606830 DOI: 10.1021/acsbiomaterials.4c01247] [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: 11/29/2024]
Abstract
Generation of in vitro tissue models with serially perfused hierarchical vasculature would allow greater control of fluid perfusion throughout the network and enable direct mechanistic investigation of vasculogenesis, angiogenesis, and vascular remodeling. In this work, we have developed a method to produce a closed, serially perfused, multiscale vessel network fully embedded within an acellular hydrogel, where flow through the capillary bed is required prior to fluid exit. We confirmed that the acellular and cellular gel-gel interface was functionally annealed without preventing or biasing cell migration and endothelial self-assembly. Multiscale connectivity of the vessel network was validated via high-resolution microscopy techniques to confirm anastomosis between self-assembled and patterned vessels. Lastly, using a simple acrylic cassette and fluorescently labeled microspheres, the multiscale network was demonstrated to be perfusable. Directed flow from inlet to outlet mandated flow through the capillary bed. This method for producing closed, multiscale vascular networks was developed with the intention of straightforward fabrication and engineering techniques so as to be a low barrier to entry for researchers who wish to investigate mechanistic questions in vascular biology. This ease of use offers a facile extension of these methods for incorporation into organoid culture, organ-on-a-chip (OOC) models, and bioprinted tissues.
Collapse
Affiliation(s)
- Michael J Donzanti
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Bryan J Ferrick
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Omkar Mhatre
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Brea Chernokal
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Diana C Renteria
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware United States 19713
| |
Collapse
|
2
|
Mehdi SJ, Zhang H, Sun RW, Richter GT, Strub GM. Mural Cells Initiate Endothelial-to-Mesenchymal Transition in Adjacent Endothelial Cells in Extracranial AVMs. Cells 2024; 13:2122. [PMID: 39768212 PMCID: PMC11727354 DOI: 10.3390/cells13242122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Extracranial arteriovenous malformations (eAVMs) are complex vascular lesions characterized by anomalous arteriovenous connections, vascular instability, and disruptions in endothelial cell (EC)-to-mural cell (MC) interactions. This study sought to determine whether eAVM-MCs could induce endothelial-to-mesenchymal transition (EndMT), a process known to disrupt vascular integrity, in the eAVM microenvironment. eAVM and paired control tissues were analyzed using RT-PCR for EC (CD31, CD34, and CDH5) and EndMT-specific markers (SNAI1, SNAI2, ACTA2/α-SMA, N-cadherin/CDH2, VIM). Immunohistochemistry (IHC) was also performed to analyze MC- (PDGFR-β and α-SMA), EC (CD31, CD34, and CDH5), and EndMT-specific markers (CDH2 and SNAI1) in sequential paraffin-embedded sections of eAVM patient biopsies and in adjacent normal tissue biopsies from the same patients. Furthermore, eAVM-MCs and MCs from normal paired tissues (NMCs) were then isolated from fresh human surgical samples using flow cytometry and co-cultured with normal human umbilical vein vascular endothelial cells (HUVECs), followed by analysis of CD31 by immunofluorescence. RT-PCR analysis did not show a significant difference in the expression of EC markers between eAVM tissues and controls, whereas expression of EndMT-specific markers was upregulated in eAVM tissues compared to controls. IHC of eAVMs and paired control tissues demonstrated regions of significant perivascular eAVM-MC expansion (PDGFR-β+, and α-SMA+) surrounding dilated, morphologically abnormal vessels. These regions contained endothelium undergoing EndMT as evidenced by loss of CD31, CD34, and CDH5 expression and upregulation of the EndMT-associated genes CDH2 and SNAI1. Isolated eAVM-MCs induced loss of CD31 in HUVECs when grown in co-culture, while NMCs did not. This study suggests that the eAVM endothelium surrounded by expanded eAVM-MCs undergoes EndMT, potentially leading to the formation of dilated and fragile vessels, and implicates the eAVM-MCs in EndMT initiation and eAVM pathology.
Collapse
Affiliation(s)
- Syed J. Mehdi
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Haihong Zhang
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Ravi W. Sun
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Gresham T. Richter
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| | - Graham M. Strub
- Arkansas Children’s Research Institute (ACRI), Little Rock, AR 72202, USA; (S.J.M.)
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA
| |
Collapse
|
3
|
Yin J, Schellinx N, Maggi L, Gundel K, Wiesner C, Kotini MP, Lee M, Phng LK, Belting HG, Affolter M. Initiation of lumen formation from junctions via differential actomyosin contractility regulated by dynamic recruitment of Rasip1. Nat Commun 2024; 15:9714. [PMID: 39521779 PMCID: PMC11550478 DOI: 10.1038/s41467-024-54143-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
De novo lumen formation necessitates the precise segregation of junctional proteins from apical surfaces, yet the underlying mechanisms remain unclear. Using a zebrafish model, we develop a series of molecular reporters, photo-convertible and optogenetic tools to study the establishment of apical domains. Our study identifies Rasip1 as one of the earliest apical proteins recruited, which suppresses actomyosin contractility at junctional patches by inhibiting NMII, thereby allowing for the sustained outward flow of junctional complexes. Following the establishment of apical compartments, Rasip1 shuttles between junctions and the apical compartments in response to local high tension. Rasip1 confines Cdh5 to junctions by suppressing apical contractility. Conversely, the recruitment of Rasip1 to junctions is regulated by Heg1 and Krit1 to modulate contractility along junctions. Overall, de novo lumen formation and maintenance depend on the precise control of contractility within apical compartments and junctions, orchestrated by the dynamic recruitment of Rasip1.
Collapse
Affiliation(s)
- Jianmin Yin
- Department of Cell Biology, Biozentrum, University of Basel, Basel, Switzerland.
| | - Niels Schellinx
- Department of Cell Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Ludovico Maggi
- Department of Cell Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Kathrin Gundel
- Department of Cell Biology, Biozentrum, University of Basel, Basel, Switzerland
- Universitätsklinikum Bonn, Bonn, Germany
| | - Cora Wiesner
- Department of Cell Biology, Biozentrum, University of Basel, Basel, Switzerland
| | | | - Minkyoung Lee
- Department of Cell Biology, Biozentrum, University of Basel, Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Heinz-Georg Belting
- Department of Cell Biology, Biozentrum, University of Basel, Basel, Switzerland.
| | - Markus Affolter
- Department of Cell Biology, Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Sasovsky DJ, Angelina E, Leiva LC, Bal de Kier Joffé E, Lomonte B, Bustillo S. Comparative in vitro and in silico analysis of the ability of basic Asp49 phospholipase A 2 and Lys49-phospholipase A 2-like myotoxins from Bothrops diporus venom to inhibit the metastatic potential of murine mammary tumor cells and endothelial cell tubulogenesis: Asp49 vs Lys49 phospholipases A 2: Inhibition of metastasis and angiogenesis. Chem Biol Interact 2024; 402:111217. [PMID: 39197813 DOI: 10.1016/j.cbi.2024.111217] [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: 04/10/2024] [Revised: 05/08/2024] [Accepted: 08/26/2024] [Indexed: 09/01/2024]
Abstract
Snake venoms are a complex mixture of proteins and polypeptides that represent a valuable source of potential molecular tools for understanding physiological processes for the development of new drugs. In this study two major PLA2s, named PLA2-I (Asp49) and PLA2-II (Lys49), isolated from the venom of Bothrops diporus from Northeastern Argentina, have shown cytotoxic effects on LM3 murine mammary tumor cells, with PLA2-II-like exhibiting a stronger effect compared to PLA2-I. At sub-cytotoxic levels, both PLA2s inhibited adhesion, migration, and invasion of these adenocarcinoma cells. Moreover, these toxins hindered tubulogenesis in endothelial cells, implicating a potential role in inhibiting tumor angiogenesis. All these inhibitory effects were more pronounced for the catalytically-inactive toxin. Additionally, in silico studies strongly suggest that this PLA2-II-like myotoxin could effectively block fibronectin binding to the integrin receptor, offering a dual advantage over PLA2-I in interacting with the αVβ3 integrin. In conclusion, this study reports for the first time, integrating both in vitro and in silico approaches, a comparative analysis of the antimetastatic and antiangiogenic potential effects of two isoforms, an Asp49 PLA2-I and a Lys49 PLA2-II-like, both isolated from Bothrops diporus venom.
Collapse
Affiliation(s)
- Daniela J Sasovsky
- Grupo de Investigaciones Biológicas y Moleculares (GIByM) IQUIBA-NEA-CONICET, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Argentina
| | - Emilio Angelina
- Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Argentina; IQUIBA-NEA-CONICET, Argentina
| | - Laura C Leiva
- Grupo de Investigaciones Biológicas y Moleculares (GIByM) IQUIBA-NEA-CONICET, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Argentina
| | - Elisa Bal de Kier Joffé
- Universidad de Buenos Aires, Facultad de Medicina, Instituto de Oncología Angel H Roffo, Área Investigación, Argentina
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, 11501, Costa Rica
| | - Soledad Bustillo
- Grupo de Investigaciones Biológicas y Moleculares (GIByM) IQUIBA-NEA-CONICET, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Argentina.
| |
Collapse
|
5
|
Zhang Y, Liu J, de Souza Araujo I, Bahammam L, Munn L, Huang G. Neovascularization by DPSC-ECs in a Tube Model for Pulp Regeneration Study. J Dent Res 2024; 103:652-661. [PMID: 38716736 PMCID: PMC11122093 DOI: 10.1177/00220345241236392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2024] Open
Abstract
The process of neovascularization during cell-based pulp regeneration is difficult to study. Here we developed a tube model that simulates root canal space and allows direct visualization of the vascularization process in vitro. Endothelial-like cells (ECs) derived from guiding human dental pulp stem cells (DPSCs) into expressing endothelial cell markers CD144, vWF, VEGFR1, and VEGFR2 were used. Human microvascular endothelial cells (hMVECs) were used as a positive control. DPSC-ECs formed tubules on Matrigel similar to hMVECs. Cells were mixed in fibrinogen/thrombin or mouse blood and seeded into wells of 96-well plates or injected into a tapered plastic tube (14 mm in length and 1 or 2 mm diameter of the apex opening) with the larger end sealed with MTA to simulate root canal space. Cells/gels in wells or tubes were incubated for various times in vitro and observed under the microscope for morphological changes. Samples were then fixed and processed for histological analysis to determine vessel formation. Vessel-like networks were observed in culture from 1 to 3 d after cell seeding. Cells/gels in 96-well plates were maintained up to 25 d. Histologically, both hMVECs and DPSC-ECs in 96-well plates or tubes showed intracellular vacuole formation. Some cells showed merged large vacuoles indicating the lumenization. Tubular structures were also observed resembling blood vessels. Cells appeared healthy throughout the tube except some samples (1 mm apical diameter) in the coronal third. Histological analysis also showed pulp-like soft tissue throughout the tube samples with vascular-like structures. hMVECs formed larger vascular lumen size than DPSC-ECs while the latter tended to have more lumen and tubular structure counts. We conclude that DPSC-ECs can form vascular structures and sustained in the 3-dimensional fibrin gel system in vitro. The tube model appears to be a proper and simple system simulating the root canal space for vascular formation and pulp regeneration studies.
Collapse
Affiliation(s)
- Y. Zhang
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - J. Liu
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - I.J. de Souza Araujo
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - L. Bahammam
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Faculty of Dentistry, King Abdulaziz University, Jeddah, Makkah, Kingdom of Saudi Arabia*
| | - L.L. Munn
- Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown, MA, USA
| | - G.T.J. Huang
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
6
|
Donzanti MJ, Mhatre O, Chernokal B, Renteria DC, Gleghorn JP. Stochastic to Deterministic: A straightforward approach to create serially perfusable multiscale capillary beds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592474. [PMID: 38766003 PMCID: PMC11100595 DOI: 10.1101/2024.05.03.592474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Generation of in vitro tissue models with serially perfused hierarchical vasculature would allow greater control of fluid perfusion throughout the network and enable direct mechanistic investigation of vasculogenesis, angiogenesis, and vascular remodeling. In this work, we have developed a method to produce a closed, serially perfused, multiscale vessel network embedded within an acellular hydrogel. We confirmed that the acellular and cellular gel-gel interface was functionally annealed without preventing or biasing cell migration and endothelial self-assembly. Multiscale connectivity of the vessel network was validated via high-resolution microscopy techniques to confirm anastomosis between self-assembled and patterned vessels. Lastly, using fluorescently labeled microspheres, the multiscale network was serially perfused to confirm patency and barrier function. Directional flow from inlet to outlet man-dated flow through the capillary bed. This method for producing closed, multiscale vascular networks was developed with the intention of straightforward fabrication and engineering techniques so as to be a low barrier to entry for researchers who wish to investigate mechanistic questions in vascular biology. This ease of use offers a facile extension of these methods for incorporation into organoid culture, organ-on-a-chip (OOC) models, and bioprinted tissues.
Collapse
|
7
|
Lin PK, Sun Z, Davis GE. Defining the Functional Influence of Endothelial Cell-Expressed Oncogenic Activating Mutations on Vascular Morphogenesis and Capillary Assembly. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:574-598. [PMID: 37838010 PMCID: PMC10988768 DOI: 10.1016/j.ajpath.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/02/2023] [Accepted: 08/15/2023] [Indexed: 10/16/2023]
Abstract
This study sought to define key molecules and signals controlling major steps in vascular morphogenesis, and how these signals regulate pericyte recruitment and pericyte-induced basement membrane deposition. The morphogenic impact of endothelial cell (EC) expression of activating mutants of Kirsten rat sarcoma virus (kRas), mitogen-activated protein kinase 1 (Mek1), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), Akt serine/threonine kinase 1 (Akt1), Ras homolog enriched in brain (Rheb) Janus kinase 2 (Jak2), or signal transducer and activator of transcription 3 (Stat3) expression versus controls was evaluated, along with EC signaling events, pharmacologic inhibitor assays, and siRNA suppression experiments. Primary stimulators of EC lumen formation included kRas, Akt1, and Mek1, whereas PIK3CA and Akt1 stimulated a specialized type of cystic lumen formation. In contrast, the key drivers of EC sprouting behavior were Jak2, Stat3, Mek1, PIK3CA, and mammalian target of rapamycin (mTor). These conclusions are further supported by pharmacologic inhibitor and siRNA suppression experiments. EC expression of active Akt1, kRas, and PIK3CA led to markedly dysregulated lumen formation coupled to strongly inhibited pericyte recruitment and basement membrane deposition. For example, activated Akt1 expression in ECs excessively stimulated lumen formation, decreased EC sprouting behavior, and showed minimal pericyte recruitment with reduced mRNA expression of platelet-derived growth factor-BB, platelet-derived growth factor-DD, and endothelin-1, critical EC-derived factors known to stimulate pericyte invasion. The study identified key signals controlling fundamental steps in capillary morphogenesis and maturation and provided mechanistic details on why EC activating mutations induced a capillary deficiency state with abnormal lumens, impaired pericyte recruitment, and basement deposition: predisposing stimuli for the development of vascular malformations.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
8
|
Akinsipe T, Mohamedelhassan R, Akinpelu A, Pondugula SR, Mistriotis P, Avila LA, Suryawanshi A. Cellular interactions in tumor microenvironment during breast cancer progression: new frontiers and implications for novel therapeutics. Front Immunol 2024; 15:1302587. [PMID: 38533507 PMCID: PMC10963559 DOI: 10.3389/fimmu.2024.1302587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 02/16/2024] [Indexed: 03/28/2024] Open
Abstract
The breast cancer tumor microenvironment (TME) is dynamic, with various immune and non-immune cells interacting to regulate tumor progression and anti-tumor immunity. It is now evident that the cells within the TME significantly contribute to breast cancer progression and resistance to various conventional and newly developed anti-tumor therapies. Both immune and non-immune cells in the TME play critical roles in tumor onset, uncontrolled proliferation, metastasis, immune evasion, and resistance to anti-tumor therapies. Consequently, molecular and cellular components of breast TME have emerged as promising therapeutic targets for developing novel treatments. The breast TME primarily comprises cancer cells, stromal cells, vasculature, and infiltrating immune cells. Currently, numerous clinical trials targeting specific TME components of breast cancer are underway. However, the complexity of the TME and its impact on the evasion of anti-tumor immunity necessitate further research to develop novel and improved breast cancer therapies. The multifaceted nature of breast TME cells arises from their phenotypic and functional plasticity, which endows them with both pro and anti-tumor roles during tumor progression. In this review, we discuss current understanding and recent advances in the pro and anti-tumoral functions of TME cells and their implications for developing safe and effective therapies to control breast cancer progress.
Collapse
Affiliation(s)
- Tosin Akinsipe
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Rania Mohamedelhassan
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Ayuba Akinpelu
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Satyanarayana R. Pondugula
- Department of Chemical Engineering, College of Engineering, Auburn University, Auburn, AL, United States
| | - Panagiotis Mistriotis
- Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - L. Adriana Avila
- Department of Biological Sciences, College of Science and Mathematics, Auburn University, Auburn, AL, United States
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
9
|
Schumacher JA, Wright ZA, Rufin Florat D, Anand SK, Dasyani M, Batta SPR, Laverde V, Ferrari K, Klimkaite L, Bredemeier NO, Gurung S, Koller GM, Aguera KN, Chadwick GP, Johnson RD, Davis GE, Sumanas S. SH2 domain protein E and ABL signaling regulate blood vessel size. PLoS Genet 2024; 20:e1010851. [PMID: 38190417 PMCID: PMC10798624 DOI: 10.1371/journal.pgen.1010851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/19/2024] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
Blood vessels in different vascular beds vary in size, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vessel size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow, eventually leading to the DA collapse. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA size in she mutants correlated with an increased endothelial expression of claudin 5a (cldn5a), which encodes a protein enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates vessel and lumen size during vascular tubulogenesis.
Collapse
Affiliation(s)
- Jennifer A. Schumacher
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Zoë A. Wright
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Diandra Rufin Florat
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surendra K. Anand
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Manish Dasyani
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surya Prakash Rao Batta
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Valentina Laverde
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Kaitlin Ferrari
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Laurita Klimkaite
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Nina O. Bredemeier
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Suman Gurung
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Gretchen M. Koller
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Kalia N. Aguera
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Griffin P. Chadwick
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Riley D. Johnson
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - George E. Davis
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Saulius Sumanas
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| |
Collapse
|
10
|
Lin PK, Koller GM, Davis GE. Elucidating the Morphogenic and Signaling Roles of Defined Growth Factors Controlling Human Endothelial Cell Lumen Formation Versus Sprouting Behavior. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:2203-2217. [PMID: 37689384 PMCID: PMC10699133 DOI: 10.1016/j.ajpath.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/25/2023] [Accepted: 08/18/2023] [Indexed: 09/11/2023]
Abstract
Five growth factors [ie, insulin, fibroblast growth factor-2 (FGF-2), stem cell factor, IL-3, and stromal-derived factor 1α] in combination are necessary for human endothelial cells (ECs) to undergo tube morphogenesis, a process requiring both lumen formation and sprouting behavior. This study investigated why these factors are required by subdividing the factors into 4 separate groups: insulin-only, insulin and FGF-2, no FGF-2 (all factors but without FGF-2), and all factors. The study found that the insulin-only condition failed to support EC morphogenesis or survival, the insulin and FGF-2 condition supported primarily EC lumen formation, and the no FGF-2 condition supported EC sprouting behavior. By comparison, the all-factors condition more strongly stimulated both EC lumen formation and sprouting behavior, and signaling analysis revealed prolonged stimulation of multiple promorphogenic signals coupled with inhibition of proregressive signals. Pharmacologic inhibition of Jak kinases more selectively blocked EC sprouting behavior, whereas inhibition of Raf, phosphatidylinositol 3-kinase, and Akt kinases showed selective blockade of lumen formation. Inhibition of Src family kinases and Notch led to increased sprouting coupled to decreased lumen formation, whereas inhibition of Pak, Mek, and mammalian target of rapamycin kinases blocked both sprouting and lumen formation. These findings reveal novel downstream biological and signaling activities of defined factors that are required for the assembly of human EC-lined capillary tube networks.
Collapse
Affiliation(s)
- Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Gretchen M Koller
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
11
|
White MJ, Singh T, Wang E, Smith Q, Kutys ML. 'Chip'-ing away at morphogenesis - application of organ-on-chip technologies to study tissue morphogenesis. J Cell Sci 2023; 136:jcs261130. [PMID: 37795818 PMCID: PMC10565497 DOI: 10.1242/jcs.261130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023] Open
Abstract
Emergent cell behaviors that drive tissue morphogenesis are the integrated product of instructions from gene regulatory networks, mechanics and signals from the local tissue microenvironment. How these discrete inputs intersect to coordinate diverse morphogenic events is a critical area of interest. Organ-on-chip technology has revolutionized the ability to construct and manipulate miniaturized human tissues with organotypic three-dimensional architectures in vitro. Applications of organ-on-chip platforms have increasingly transitioned from proof-of-concept tissue engineering to discovery biology, furthering our understanding of molecular and mechanical mechanisms that operate across biological scales to orchestrate tissue morphogenesis. Here, we provide the biological framework to harness organ-on-chip systems to study tissue morphogenesis, and we highlight recent examples where organ-on-chips and associated microphysiological systems have enabled new mechanistic insight in diverse morphogenic settings. We further highlight the use of organ-on-chip platforms as emerging test beds for cell and developmental biology.
Collapse
Affiliation(s)
- Matthew J. White
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Tania Singh
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
- UCSF-UC Berkeley Joint Program in Bioengineering, University of California San Francisco, San Francisco, CA 94143, USA
| | - Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA
| | - Matthew L. Kutys
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA 94143, USA
- UCSF-UC Berkeley Joint Program in Bioengineering, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
12
|
Lin PK, Davis GE. Extracellular Matrix Remodeling in Vascular Disease: Defining Its Regulators and Pathological Influence. Arterioscler Thromb Vasc Biol 2023; 43:1599-1616. [PMID: 37409533 PMCID: PMC10527588 DOI: 10.1161/atvbaha.123.318237] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/23/2023] [Indexed: 07/07/2023]
Abstract
Because of structural and cellular differences (ie, degrees of matrix abundance and cross-linking, mural cell density, and adventitia), large and medium-sized vessels, in comparison to capillaries, react in a unique manner to stimuli that induce vascular disease. A stereotypical vascular injury response is ECM (extracellular matrix) remodeling that occurs particularly in larger vessels in response to injurious stimuli, such as elevated angiotensin II, hyperlipidemia, hyperglycemia, genetic deficiencies, inflammatory cell infiltration, or exposure to proinflammatory mediators. Even with substantial and prolonged vascular damage, large- and medium-sized arteries, persist, but become modified by (1) changes in vascular wall cellularity; (2) modifications in the differentiation status of endothelial cells, vascular smooth muscle cells, or adventitial stem cells (each can become activated); (3) infiltration of the vascular wall by various leukocyte types; (4) increased exposure to critical growth factors and proinflammatory mediators; and (5) marked changes in the vascular ECM, that remodels from a homeostatic, prodifferentiation ECM environment to matrices that instead promote tissue reparative responses. This latter ECM presents previously hidden matricryptic sites that bind integrins to signal vascular cells and infiltrating leukocytes (in coordination with other mediators) to proliferate, invade, secrete ECM-degrading proteinases, and deposit injury-induced matrices (predisposing to vessel wall fibrosis). In contrast, in response to similar stimuli, capillaries can undergo regression responses (rarefaction). In summary, we have described the molecular events controlling ECM remodeling in major vascular diseases as well as the differential responses of arteries versus capillaries to key mediators inducing vascular injury.
Collapse
Affiliation(s)
- Prisca K. Lin
- 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
|
13
|
Sun Z, Lin PK, Yrigoin K, Kemp SS, Davis GE. Increased Matrix Metalloproteinase-1 Activation Enhances Disruption and Regression of k-RasV12-Expressing Arteriovenous Malformation-Like Vessels. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1319-1334. [PMID: 37328101 PMCID: PMC10477956 DOI: 10.1016/j.ajpath.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023]
Abstract
This study sought to identify potential mechanisms by which k-RasV12-expressing endothelial cell (EC) tubes demonstrate an increased propensity to regress compared with controls. Activated k-Ras mutations play a role in a variety of pathological conditions, including arteriovenous malformations, which are prone to bleed, causing serious hemorrhagic complications. ECs expressing active k-RasV12 demonstrate markedly excessive lumen formation with widened and shortened tubes accompanied by reduced pericyte recruitment and basement membrane deposition, leading to deficient capillary network assembly. The current study showed that active k-Ras-expressing ECs secreted greater amounts of MMP-1 proenzyme compared with control ECs, and readily converted it to increased active MMP-1 levels through the action of plasmin or plasma kallikrein (generated from their added zymogens). Active MMP-1 degraded three-dimensional collagen matrices, leading to more rapid and extensive regression of the active k-Ras-expressing EC tubes, in conjunction with matrix contraction, compared with control ECs. Under conditions where pericytes protect control EC tubes from plasminogen- and MMP-1-dependent tube regression, this failed to occur with k-RasV12 ECs, due to reduced pericyte interactions. In summary, k-RasV12-expressing EC vessels showed an increased propensity to regress in response to serine proteinases through accentuated levels of active MMP-1, a novel pathogenic mechanism that may underlie hemorrhagic events associated with arteriovenous malformation lesions.
Collapse
Affiliation(s)
- Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Ksenia Yrigoin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, Florida.
| |
Collapse
|
14
|
Schumacher JA, Wright ZA, Florat DR, Anand SK, Dasyani M, Klimkaite L, Bredemeier NO, Gurung S, Koller GM, Aguera KN, Chadwick GP, Johnson RD, Davis GE, Sumanas S. SH2 domain protein E (SHE) and ABL signaling regulate blood vessel size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547455. [PMID: 37461480 PMCID: PMC10349984 DOI: 10.1101/2023.07.03.547455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Blood vessels in different vascular beds vary in lumen diameter, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vascular lumen size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA lumen, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA lumen in she mutants correlated with an increased endothelial expression of claudin 5a and 5b (cldn5a / cldn5b), which encode proteins enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA lumen size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates lumen size during vascular tubulogenesis.
Collapse
Affiliation(s)
- Jennifer A. Schumacher
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, OH 45267, USA
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - Zoë A. Wright
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Diandra Rufin Florat
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Surendra K. Anand
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Manish Dasyani
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Laurita Klimkaite
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Nina O. Bredemeier
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Suman Gurung
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Gretchen M. Koller
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Kalia N. Aguera
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Griffin P. Chadwick
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - Riley D. Johnson
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - George E. Davis
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Saulius Sumanas
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, OH 45267, USA
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| |
Collapse
|
15
|
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: 6] [Impact Index Per Article: 3.0] [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
|
16
|
Chummun Phul I, Huët MAL, Bekah D, Bhaw-Luximon A. Polysucrose hydrogel loaded with natural molecules/extracts for multiphase-directed sustainable wound healing. RSC Med Chem 2023; 14:534-548. [PMID: 36970144 PMCID: PMC10034044 DOI: 10.1039/d2md00402j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Natural molecules/extracts have numerous beneficial effects on wound healing processes which are challenged by appropriate use and non-toxic dosage. Polysucrose-based (PSucMA) hydrogels have been synthesized with in situ loading of one or more natural molecules/extracts namely Manuka honey (MH), Eucalyptus honey (EH1, EH2), Ginkgo biloba (GK), thymol (THY) and metformin (MET). EH1 presented low amounts of hydroxymethylfurfural and methylglyoxal compared to MH indicating that EH1 was not temperature-abused. It also showed high diastase activity and conductivity. GK was added to PSucMA solution along with other additives including MH, EH1 and MET and crosslinked to form dual loaded hydrogels. The in vitro release profiles of EH1, MH, GK and THY from the hydrogels followed the exponential Korsmeyer-Peppas equation, with a release exponent value of less than 0.5 indicating a quasi-Fickian diffusion mechanism. The IC50 values of these natural products using L929 fibroblasts and RAW 264.7 macrophages indicated that EH1, MH and GK were cytocompatible at relatively high concentrations compared to MET, THY and curcumin used as a control. MH and EH1 induced high IL6 concentration compared to GK. In vitro studies were modelled to mimic the overlapping wound healing phases using human dermal fibroblasts (HDFs), macrophages and human umbilical endothelial cells (HUVECs) in dual culture. HDFs showed a highly interconnected cellular network on GK loaded scaffolds. EH1 loaded scaffolds were seen to induce formation of spheroids which increased in number and size in co-culture studies. The SEM images of HDF/HUVEC seeded GK, GKMH and GKEH1 loaded hydrogels indicated formation of vacuoles and lumen structures. These results indicated that a combination of GK and EH1 in the hydrogel scaffold would accelerate tissue regeneration by acting on the four overlapping phases of wound healing.
Collapse
Affiliation(s)
- Itisha Chummun Phul
- Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical & Biomaterials Research (CBBR), University of Mauritius 80837 Réduit Mauritius
| | - Marie Andrea Laetitia Huët
- Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical & Biomaterials Research (CBBR), University of Mauritius 80837 Réduit Mauritius
| | - Devesh Bekah
- Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical & Biomaterials Research (CBBR), University of Mauritius 80837 Réduit Mauritius
| | - Archana Bhaw-Luximon
- Biomaterials, Drug Delivery & Nanotechnology Unit, Centre for Biomedical & Biomaterials Research (CBBR), University of Mauritius 80837 Réduit Mauritius
| |
Collapse
|
17
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
18
|
Accelerated Angiogenesis of Human Umbilical Vein Endothelial Cells Under Negative Pressure Was Associated With the Regulation of Gene Expression Involved in the Proliferation and Migration. Ann Plast Surg 2022; 89:e51-e59. [DOI: 10.1097/sap.0000000000003298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Gonzalez-Avila G, Sommer B, Garcia-Hernandez AA, Ramos C, Delgado J, Vazquez L, Gonzalez RA, Sandoval C, Flores-Soto E. Matrix Metalloproteinases and Stress Hormones in Lung Cancer Progression. JOURNAL OF ONCOLOGY 2022; 2022:5349691. [PMID: 36213817 PMCID: PMC9536982 DOI: 10.1155/2022/5349691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022]
Abstract
Several matrix metalloproteinases (MMPs) and psychological stress are associated with poor cancer prognosis. The current work goal was to determine MMPs' and stress hormones' blood concentrations from lung adenocarcinoma (LAC) patients. Patients were divided into the following groups: tobacco smokers (TS), wood smoke-exposed (W), passive smokers (PS), TS exposed to wood smoke (TW), and patients with no recognizable risk factor (N). MMPs, tissue inhibitors of metalloproteinases (TIMPs), adrenaline, noradrenaline, and cortisol blood concentrations were measured by ELISA. Zymography and Western blot assays were performed to determine MMP-2 and MMP-9 active and latent forms. MMP-2, MMP-3, MMP-9, and TIMP-1 blood concentrations, and MMP-9 gelatinase activity were augmented, while MMP-12, MMP-14, and TIMP-2 were diminished in LAC patients. Cortisol was increased in LAC samples. Adrenaline concentrations were higher in W, TS, and TW, and noradrenaline was increased in W and N groups. Positive correlations were observed among cortisol and TIMP-1 (r s = 0.392) and TIMP-2 (r s = 0.409) in the W group and between noradrenaline and MMP-2 (r s = 0.391) in the N group. MMPs' blood concentration increments can be considered as lung cancer progression markers. Although stress hormones were also augmented, only weak correlations were observed between them and MMPs and TIMPs.
Collapse
Affiliation(s)
- Georgina Gonzalez-Avila
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, CP, Mexico
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, CP, Mexico
| | - A. Armando Garcia-Hernandez
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, CP, Mexico
| | - Carlos Ramos
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, CP, Mexico
| | - Javier Delgado
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, CP, Mexico
| | - Lilia Vazquez
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, CP, Mexico
| | - Rosa A. Gonzalez
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, CP, Mexico
| | - Cuauhtemoc Sandoval
- Laboratorio Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Calzada de Tlalpan 4502, Col. Sección XVI, Tlalpan, 14080 Ciudad de México, CP, Mexico
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán, 04510 Ciudad de México, CP, Mexico
| |
Collapse
|
20
|
Bae E, Huang P, Müller-Greven G, Hambardzumyan D, Sloan AE, Nowacki AS, Marko N, Carlin CR, Gladson CL. Integrin α3β1 promotes vessel formation of glioblastoma-associated endothelial cells through calcium-mediated macropinocytosis and lysosomal exocytosis. Nat Commun 2022; 13:4268. [PMID: 35879332 PMCID: PMC9314429 DOI: 10.1038/s41467-022-31981-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 07/12/2022] [Indexed: 12/13/2022] Open
Abstract
Therapeutic targeting of angiogenesis in glioblastoma has yielded mixed outcomes. Investigation of tumor-associated angiogenesis has focused on the factors that stimulate the sprouting, migration, and hyperproliferation of the endothelial cells. However, little is known regarding the processes underlying the formation of the tumor-associated vessels. To address this issue, we investigated vessel formation in CD31+ cells isolated from human glioblastoma tumors. The results indicate that overexpression of integrin α3β1 plays a central role in the promotion of tube formation in the tumor-associated endothelial cells in glioblastoma. Blocking α3β1 function reduced sprout and tube formation in the tumor-associated endothelial cells and vessel density in organotypic cultures of glioblastoma. The data further suggest a mechanistic model in which integrin α3β1-promoted calcium influx stimulates macropinocytosis and directed maturation of the macropinosomes in a manner that promotes lysosomal exocytosis during nascent lumen formation. Altogether, our data indicate that integrin α3β1 may be a therapeutic target on the glioblastoma vasculature.
Collapse
Affiliation(s)
- Eunnyung Bae
- Department of Cancer Biology, Cleveland, Clinic, Cleveland, OH, USA
| | - Ping Huang
- Department of Cancer Biology, Cleveland, Clinic, Cleveland, OH, USA
| | | | - Dolores Hambardzumyan
- Departments of Oncological Sciences and Neurosurgery, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Andrew Edward Sloan
- Department of Neurosurgery, Seidman Cancer Center, Cleveland, OH, USA
- University Hospital-Cleveland Medical Center and the Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Amy S Nowacki
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Nicholas Marko
- Department of Neurosurgery, LewisGale Medical Center, Salem, VA, USA
| | - Cathleen R Carlin
- University Hospital-Cleveland Medical Center and the Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Candece L Gladson
- Department of Cancer Biology, Cleveland, Clinic, Cleveland, OH, USA.
- University Hospital-Cleveland Medical Center and the Case Comprehensive Cancer Center, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.
- The Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
21
|
Zarubova J, Hasani-Sadrabadi MM, Ardehali R, Li S. Immunoengineering strategies to enhance vascularization and tissue regeneration. Adv Drug Deliv Rev 2022; 184:114233. [PMID: 35304171 PMCID: PMC10726003 DOI: 10.1016/j.addr.2022.114233] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022]
Abstract
Immune cells have emerged as powerful regulators of regenerative as well as pathological processes. The vast majority of regenerative immunoengineering efforts have focused on macrophages; however, growing evidence suggests that other cells of both the innate and adaptive immune system are as important for successful revascularization and tissue repair. Moreover, spatiotemporal regulation of immune cells and their signaling have a significant impact on the regeneration speed and the extent of functional recovery. In this review, we summarize the contribution of different types of immune cells to the healing process and discuss ways to manipulate and control immune cells in favor of vascularization and tissue regeneration. In addition to cell delivery and cell-free therapies using extracellular vesicles, we discuss in situ strategies and engineering approaches to attract specific types of immune cells and modulate their phenotypes. This field is making advances to uncover the extraordinary potential of immune cells and their secretome in the regulation of vascularization and tissue remodeling. Understanding the principles of immunoregulation will help us design advanced immunoengineering platforms to harness their power for tissue regeneration.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | | | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
22
|
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: 0.3] [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
|
23
|
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: 21] [Impact Index Per Article: 7.0] [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
|
24
|
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: 3.0] [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
|
25
|
Li Y, Fraser D, Mereness J, Van Hove A, Basu S, Newman M, Benoit DSW. Tissue Engineered Neurovascularization Strategies for Craniofacial Tissue Regeneration. ACS APPLIED BIO MATERIALS 2022; 5:20-39. [PMID: 35014834 PMCID: PMC9016342 DOI: 10.1021/acsabm.1c00979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Craniofacial tissue injuries, diseases, and defects, including those within bone, dental, and periodontal tissues and salivary glands, impact an estimated 1 billion patients globally. Craniofacial tissue dysfunction significantly reduces quality of life, and successful repair of damaged tissues remains a significant challenge. Blood vessels and nerves are colocalized within craniofacial tissues and act synergistically during tissue regeneration. Therefore, the success of craniofacial regenerative approaches is predicated on successful recruitment, regeneration, or integration of both vascularization and innervation. Tissue engineering strategies have been widely used to encourage vascularization and, more recently, to improve innervation through host tissue recruitment or prevascularization/innervation of engineered tissues. However, current scaffold designs and cell or growth factor delivery approaches often fail to synergistically coordinate both vascularization and innervation to orchestrate successful tissue regeneration. Additionally, tissue engineering approaches are typically investigated separately for vascularization and innervation. Since both tissues act in concert to improve craniofacial tissue regeneration outcomes, a revised approach for development of engineered materials is required. This review aims to provide an overview of neurovascularization in craniofacial tissues and strategies to target either process thus far. Finally, key design principles are described for engineering approaches that will support both vascularization and innervation for successful craniofacial tissue regeneration.
Collapse
Affiliation(s)
- Yiming Li
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - David Fraser
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jared Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Amy Van Hove
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Sayantani Basu
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Maureen Newman
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States.,Materials Science Program, University of Rochester, Rochester, New York 14627, United States.,Department of Chemical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Biomedical Genetics and Center for Oral Biology, University of Rochester Medical Center, Rochester, New York 14642, United States
| |
Collapse
|
26
|
Kemp SS, Lin PK, Sun Z, Castaño MA, Yrigoin K, Penn MR, Davis GE. Molecular basis for pericyte-induced capillary tube network assembly and maturation. Front Cell Dev Biol 2022; 10:943533. [PMID: 36072343 PMCID: PMC9441561 DOI: 10.3389/fcell.2022.943533] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Here we address the functional importance and role of pericytes in capillary tube network assembly, an essential process that is required for vascularized tissue development, maintenance, and health. Healthy capillaries may be directly capable of suppressing human disease. Considerable advances have occurred in our understanding of the molecular and signaling requirements controlling EC lumen and tube formation in 3D extracellular matrices. A combination of SCF, IL-3, SDF-1α, FGF-2 and insulin ("Factors") in conjunction with integrin- and MT1-MMP-induced signaling are required for EC sprouting behavior and tube formation under serum-free defined conditions. Pericyte recruitment to the abluminal EC tube surface results in elongated and narrow tube diameters and deposition of the vascular basement membrane. In contrast, EC tubes in the absence of pericytes continue to widen and shorten over time and fail to deposit basement membranes. Pericyte invasion, recruitment and proliferation in 3D matrices requires the presence of ECs. A detailed analysis identified that EC-derived PDGF-BB, PDGF-DD, ET-1, HB-EGF, and TGFβ1 are necessary for pericyte recruitment, proliferation, and basement membrane deposition. Blockade of these individual factors causes significant pericyte inhibition, but combined blockade profoundly interferes with these events, resulting in markedly widened EC tubes without basement membranes, like when pericytes are absent.
Collapse
Affiliation(s)
- Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Prisca K Lin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Zheying Sun
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Maria A Castaño
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Ksenia Yrigoin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - Marlena R Penn
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa, FL, United States
| |
Collapse
|
27
|
Kogan PS, Wirth F, Tomar A, Darr J, Teperino R, Lahm H, Dreßen M, Puluca N, Zhang Z, Neb I, Beck N, Luzius T, de la Osa de la Rosa L, Gärtner K, Hüls C, Zeidler R, Ramanujam D, Engelhardt S, Wenk C, Holdt LM, Mononen M, Sahara M, Cleuziou J, Hörer J, Lange R, Krane M, Doppler SA. Uncovering the molecular identity of cardiosphere-derived cells (CDCs) by single-cell RNA sequencing. Basic Res Cardiol 2022; 117:11. [PMID: 35258704 PMCID: PMC8902493 DOI: 10.1007/s00395-022-00913-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 01/31/2023]
Abstract
Cardiosphere-derived cells (CDCs) generated from human cardiac biopsies have been shown to have disease-modifying bioactivity in clinical trials. Paradoxically, CDCs' cellular origin in the heart remains elusive. We studied the molecular identity of CDCs using single-cell RNA sequencing (sc-RNAseq) in comparison to cardiac non-myocyte and non-hematopoietic cells (cardiac fibroblasts/CFs, smooth muscle cells/SMCs and endothelial cells/ECs). We identified CDCs as a distinct and mitochondria-rich cell type that shared biological similarities with non-myocyte cells but not with cardiac progenitor cells derived from human-induced pluripotent stem cells. CXCL6 emerged as a new specific marker for CDCs. By analysis of sc-RNAseq data from human right atrial biopsies in comparison with CDCs we uncovered transcriptomic similarities between CDCs and CFs. By direct comparison of infant and adult CDC sc-RNAseq data, infant CDCs revealed GO-terms associated with cardiac development. To analyze the beneficial effects of CDCs (pro-angiogenic, anti-fibrotic, anti-apoptotic), we performed functional in vitro assays with CDC-derived extracellular vesicles (EVs). CDC EVs augmented in vitro angiogenesis and did not stimulate scarring. They also reduced the expression of pro-apoptotic Bax in NRCMs. In conclusion, CDCs were disclosed as mitochondria-rich cells with unique properties but also with similarities to right atrial CFs. CDCs displayed highly proliferative, secretory and immunomodulatory properties, characteristics that can also be found in activated or inflammatory cell types. By special culture conditions, CDCs earn some bioactivities, including angiogenic potential, which might modify disease in certain disorders.
Collapse
Affiliation(s)
- Palgit-S. Kogan
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Felix Wirth
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Archana Tomar
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jonatan Darr
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Raffaele Teperino
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Harald Lahm
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Martina Dreßen
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Nazan Puluca
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Zhong Zhang
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Irina Neb
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Nicole Beck
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Tatjana Luzius
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Luis de la Osa de la Rosa
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Kathrin Gärtner
- Research Unit Gene Vectors, Helmholtz Center Munich German Research Center for Environmental Health, Munich, Germany
| | - Corinna Hüls
- Research Unit Gene Vectors, Helmholtz Center Munich German Research Center for Environmental Health, Munich, Germany
| | - Reinhard Zeidler
- Research Unit Gene Vectors, Helmholtz Center Munich German Research Center for Environmental Health, Munich, Germany ,Department of Otorhinolaryngology, Klinikum der Universität (KUM), Munich, Germany
| | - Deepak Ramanujam
- DZHK (German Center for Cardiovascular Research)-Partner Site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany ,Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, 80802 Munich, Germany
| | - Stefan Engelhardt
- DZHK (German Center for Cardiovascular Research)-Partner Site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany ,Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, 80802 Munich, Germany
| | - Catharina Wenk
- Institute of Laboratory Medicine, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Lesca M. Holdt
- Institute of Laboratory Medicine, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Mimmi Mononen
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden ,Department of Surgery, Yale University School of Medicine, CN06510 New Haven, CT USA
| | - Julie Cleuziou
- School of Medicine and Health, Department of Pediatric and Congenital Heart Surgery, Institute Insure, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
| | - Jürgen Hörer
- School of Medicine and Health, Department of Pediatric and Congenital Heart Surgery, Technical University of Munich, German Heart Center Munich, Lazarettstraße 36, 80636 Munich, Germany ,Division of Congenital and Pediatric Heart Surgery, University Hospital of Munich, Ludwig-Maximilians-Universität, Munich, Germany
| | - Rüdiger Lange
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany ,DZHK (German Center for Cardiovascular Research)-Partner Site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Markus Krane
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany ,DZHK (German Center for Cardiovascular Research)-Partner Site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany ,Division of Cardiac Surgery, Department of Surgery, Yale University School of Medicine, New Haven, CT USA
| | - Stefanie A. Doppler
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| |
Collapse
|
28
|
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: 0.8] [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
|
29
|
Kümper M, Hessenthaler S, Zamek J, Niland S, Pach E, Mauch C, Zigrino P. LOSS OF ENDOTHELIAL CELL MMP14 REDUCES MELANOMA GROWTH AND METASTASIS BY INCREASING TUMOR VESSEL STABILITY. J Invest Dermatol 2021; 142:1923-1933.e5. [DOI: 10.1016/j.jid.2021.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 12/03/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022]
|
30
|
Dynamic Expression of Membrane Type 1-Matrix Metalloproteinase (Mt1-mmp/Mmp14) in the Mouse Embryo. Cells 2021; 10:cells10092448. [PMID: 34572097 PMCID: PMC8465375 DOI: 10.3390/cells10092448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/03/2021] [Accepted: 09/15/2021] [Indexed: 01/13/2023] Open
Abstract
MT1-MMP/MMP14 belongs to a subgroup of the matrix metalloproteinases family that presents a transmembrane domain, with a cytosolic tail and the catalytic site exposed to the extracellular space. Deficient mice for this enzyme result in early postnatal death and display severe defects in skeletal, muscle and lung development. By using a transgenic line expressing the LacZ reporter under the control of the endogenous Mt1-mmp promoter, we reported a dynamic spatiotemporal expression pattern for Mt1-mmp from early embryonic to perinatal stages during cardiovascular development and brain formation. Thus, Mt1-mmp shows expression in the endocardium of the heart and the truncus arteriosus by E8.5, and is also strongly detected during vascular system development as well as in endothelial cells. In the brain, LacZ reporter expression was detected in the olfactory bulb, the rostral cerebral cortex and the caudal mesencephalic tectum. LacZ-positive cells were observed in neural progenitors of the spinal cord, neural crest cells and the intersomitic region. In the limb, Mt1-mmp expression was restricted to blood vessels, cartilage primordium and muscles. Detection of the enzyme was confirmed by Western blot and immunohistochemical analysis. We suggest novel functions for this metalloproteinase in angiogenesis, endocardial formation and vascularization during organogenesis. Moreover, Mt1-mmp expression revealed that the enzyme may contribute to heart, muscle and brain throughout development.
Collapse
|
31
|
Ewald ML, Chen YH, Lee AP, Hughes CCW. The vascular niche in next generation microphysiological systems. LAB ON A CHIP 2021; 21:3244-3262. [PMID: 34396383 PMCID: PMC8635227 DOI: 10.1039/d1lc00530h] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
In recent years, microphysiological system (MPS, also known as, organ-on-a-chip or tissue chip) platforms have emerged with great promise to improve the predictive capacity of preclinical modeling thereby reducing the high attrition rates when drugs move into trials. While their designs can vary quite significantly, in general MPS are bioengineered in vitro microenvironments that recapitulate key functional units of human organs, and that have broad applications in human physiology, pathophysiology, and clinical pharmacology. A critical next step in the evolution of MPS devices is the widespread incorporation of functional vasculature within tissues. The vasculature itself is a major organ that carries nutrients, immune cells, signaling molecules and therapeutics to all other organs. It also plays critical roles in inducing and maintaining tissue identity through expression of angiocrine factors, and in providing tissue-specific milieus (i.e., the vascular niche) that can support the survival and function of stem cells. Thus, organs are patterned, maintained and supported by the vasculature, which in turn receives signals that drive tissue specific gene expression. In this review, we will discuss published vascularized MPS platforms and present considerations for next-generation devices looking to incorporate this critical constituent. Finally, we will highlight the organ-patterning processes governed by the vasculature, and how the incorporation of a vascular niche within MPS platforms will establish a unique opportunity to study stem cell development.
Collapse
Affiliation(s)
- Makena L Ewald
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA.
| | | | | | | |
Collapse
|
32
|
Zhang Y, Li T, Zhao C, Li J, Huang R, Zhang Q, Li Y, Li X. An Integrated Smart Sensor Dressing for Real-Time Wound Microenvironment Monitoring and Promoting Angiogenesis and Wound Healing. Front Cell Dev Biol 2021; 9:701525. [PMID: 34422823 PMCID: PMC8378138 DOI: 10.3389/fcell.2021.701525] [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: 04/28/2021] [Accepted: 07/19/2021] [Indexed: 12/28/2022] Open
Abstract
Prolonged chronic wound healing not only places great stress on patients but also increase the health care burden. Fortunately, the emergence of tissue-engineered dressings has provided a potential solution for these patients. Recently, the relationship between the wound microenvironment and wound healing has been gradually clarified. Therefore, the state of wounds can be roughly ascertained by monitoring the microenvironment in real time. Here, we designed a three-layer integrated smart dressing, including a biomimetic nanofibre membrane, microenvironment sensor and β-cyclodextrin-containing gelatine methacryloyl (GelMA + β-cd) UV-crosslinked hydrogel. The hydrogel helped increase the expression of vascular endothelial growth factor (VEGF) through hypoxia-inducible factor-1α (HIF-1α) to promote neovascularization and wound healing. The microenvironment sensor, combined with the biological dressings, exhibited satisfactory measurement accuracy, stability, durability and biocompatibility. A BLE4.0 antenna was used to receive, display and upload wound microenvironment data in real time. Such integrated smart dressings can not only achieve biological functions but also monitor changes in the wound microenvironment in real time. These dressings can overcome the challenge of not knowing the state of the wound during the healing process and provide support for clinical work.
Collapse
Affiliation(s)
- Yuheng Zhang
- Department of Burn and Plastic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.,Air Force Hospital of Western Theater Command, Chengdu, China
| | - Tian Li
- Air Force Hospital of Western Theater Command, Chengdu, China.,School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Congying Zhao
- Department of Burn and Plastic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinqing Li
- Department of Burn and Plastic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Rong Huang
- Department of Burn and Plastic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Qianru Zhang
- School of Software Center for High Performance Computing, Northwestern Polytechnical University, Xi'an, China
| | - Yongqian Li
- Key Laboratory of Micro/Nano Systems for Aerospace, Ministry of Education, Northwestern Polytechnical University, Xi'an, China
| | - Xueyong Li
- Department of Burn and Plastic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
33
|
Lee M, Betz C, Yin J, Paatero I, Schellinx N, Carte AN, Wilson CW, Ye W, Affolter M, Belting HG. Control of dynamic cell behaviors during angiogenesis and anastomosis by Rasip1. Development 2021; 148:271819. [PMID: 34383884 PMCID: PMC8380458 DOI: 10.1242/dev.197509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/08/2021] [Indexed: 11/23/2022]
Abstract
Organ morphogenesis is driven by a wealth of tightly orchestrated cellular behaviors, which ensure proper organ assembly and function. Many of these cell activities involve cell-cell interactions and remodeling of the F-actin cytoskeleton. Here, we analyze the requirement for Rasip1 (Ras-interacting protein 1), an endothelial-specific regulator of junctional dynamics, during blood vessel formation. Phenotype analysis of rasip1 mutants in zebrafish embryos reveals distinct functions of Rasip1 during sprouting angiogenesis, anastomosis and lumen formation. During angiogenic sprouting, loss of Rasip1 causes cell pairing defects due to a destabilization of tricellular junctions, indicating that stable tricellular junctions are essential to maintain multicellular organization within the sprout. During anastomosis, Rasip1 is required to establish a stable apical membrane compartment; rasip1 mutants display ectopic, reticulated junctions and the apical compartment is frequently collapsed. Loss of Ccm1 and Heg1 function mimics the junctional defects of rasip1 mutants. Furthermore, downregulation of ccm1 and heg1 leads to a delocalization of Rasip1 at cell junctions, indicating that junctional tethering of Rasip1 is required for its function in junction formation and stabilization during sprouting angiogenesis. Summary:In vivo analysis of rasip1 mutants reveals multiple roles for Rasip1 during angiogenic sprouting, anastomosis and lumen formation, including stabilization of tricellular junctions to permit coordinated cell rearrangements and multicellular tube formation.
Collapse
Affiliation(s)
- Minkyoung Lee
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Charles Betz
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Jianmin Yin
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Ilkka Paatero
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Niels Schellinx
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Adam N Carte
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Christopher W Wilson
- Department of Molecular Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Weilan Ye
- Department of Molecular Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Markus Affolter
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Heinz-Georg Belting
- Department of Cell Biology, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| |
Collapse
|
34
|
Dey M, Ayan B, Yurieva M, Unutmaz D, Ozbolat IT. Studying Tumor Angiogenesis and Cancer Invasion in a Three-Dimensional Vascularized Breast Cancer Micro-Environment. Adv Biol (Weinh) 2021; 5:e2100090. [PMID: 33857356 PMCID: PMC8574137 DOI: 10.1002/adbi.202100090] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/28/2021] [Indexed: 12/19/2022]
Abstract
Metastatic breast cancer is one of the deadliest forms of malignancy, primarily driven by its characteristic micro-environment comprising cancer cells interacting with stromal components. These interactions induce genetic and metabolic alterations creating a conducive environment for tumor growth. In this study, a physiologically relevant 3D vascularized breast cancer micro-environment is developed comprising of metastatic MDA-MB-231 cells and human umbilical vein endothelial cells loaded in human dermal fibroblasts laden fibrin, representing the tumor stroma. The matrix, as well as stromal cell density, impacts the transcriptional profile of genes involved in tumor angiogenesis and cancer invasion, which are hallmarks of cancer. Cancer-specific canonical pathways and activated upstream regulators are also identified by the differential gene expression signatures of these composite cultures. Additionally, a tumor-associated vascular bed of capillaries is established exhibiting dilated vessel diameters, representative of in vivo tumor physiology. Further, employing aspiration-assisted bioprinting, cancer-endothelial crosstalk, in the form of collective angiogenesis of tumor spheroids bioprinted at close proximity, is identified. Overall, this bottom-up approach of tumor micro-environment fabrication provides an insight into the potential of in vitro tumor models and enables the identification of novel therapeutic targets as a preclinical drug screening platform.
Collapse
Affiliation(s)
- Madhuri Dey
- Department of Chemistry, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Bugra Ayan
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Marina Yurieva
- The Jackson Laboratory for Genomic Medicine and University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Derya Unutmaz
- The Jackson Laboratory for Genomic Medicine and University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
- Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA
- Materials Research Institute, Penn State University, University Park, PA 16802, USA
| |
Collapse
|
35
|
Herrera JL, Komatsu M. R-Ras Deficiency in Pericytes Causes Frequent Microphthalmia and Perturbs Retinal Vascular Development. J Vasc Res 2021; 58:252-266. [PMID: 33873190 PMCID: PMC8263468 DOI: 10.1159/000514555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The retinal vasculature is heavily invested by pericytes. Small GTPase R-Ras is highly expressed in endothelial cells and pericytes, suggesting importance of this Ras homolog for the regulation of the blood vessel wall. We investigated the specific contribution of pericyte-expressed R-Ras to the development of the retinal vasculature. METHODS The effect of R-Ras deficiency in pericytes was analyzed in pericyte-targeted conditional Rras knockout mice at birth and during the capillary plexus formation in the neonatal retina. RESULTS The offspring of these mice frequently exhibited unilateral microphthalmia. Analyses of the developing retinal vasculature in the eyes without microphthalmia revealed excessive endothelial cell proliferation, sprouting, and branching of the capillary plexus in these animals. These vessels were structurally defective with diminished pericyte coverage and basement membrane formation. Furthermore, these vessels showed reduced VE-cadherin staining and significantly elevated plasma leakage indicating the breakdown of the blood-retinal barrier. This defect was associated with considerable macrophage infiltration in the retina. CONCLUSIONS The normal retinal vascular development is dependent on R-Ras expression in pericytes, and the absence of it leads to unattenuated angiogenesis and significantly weakens the blood-retinal barrier. Our findings underscore the importance of R-Ras for pericyte function during the normal eye development.
Collapse
Affiliation(s)
- Jose Luis Herrera
- Cancer and Blood Disorders Institute, Institute for Fundamental Biomedical Research, and Department of Surgery, Johns Hopkins All Children's Hospital, and Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - Masanobu Komatsu
- Cancer and Blood Disorders Institute, Institute for Fundamental Biomedical Research, and Department of Surgery, Johns Hopkins All Children's Hospital, and Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| |
Collapse
|
36
|
Li Y, Hoffman MD, Benoit DSW. Matrix metalloproteinase (MMP)-degradable tissue engineered periosteum coordinates allograft healing via early stage recruitment and support of host neurovasculature. Biomaterials 2021; 268:120535. [PMID: 33271450 PMCID: PMC8110201 DOI: 10.1016/j.biomaterials.2020.120535] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/17/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022]
Abstract
Despite serving as the clinical "gold standard" treatment for critical size bone defects, decellularized allografts suffer from long-term failure rates of ~60% due to the absence of the periosteum. Stem and osteoprogenitor cells within the periosteum orchestrate autograft healing through host cell recruitment, which initiates the regenerative process. To emulate periosteum-mediated healing, tissue engineering approaches have been utilized with mixed outcomes. While vascularization has been widely established as critical for bone regeneration, innervation was recently identified to be spatiotemporally regulated together with vascularization and similarly indispensable to bone healing. Notwithstanding, there are no known approaches that have focused on periosteal matrix cues to coordinate host vessel and/or axon recruitment. Here, we investigated the influence of hydrogel degradation mechanism, i.e. hydrolytic or enzymatic (cell-dictated), on tissue engineered periosteum (TEP)-modified allograft healing, especially host vessel/nerve recruitment and integration. Matrix metalloproteinase (MMP)-degradable hydrogels supported endothelial cell migration from encapsulated spheroids whereas no migration was observed in hydrolytically degradable hydrogels in vitro, which correlated with increased neurovascularization in vivo. Specifically, ~2.45 and 1.84-fold, and ~3.48 and 2.58-fold greater vessel and nerve densities with high levels of vessel and nerve co-localization was observed using MMP degradable TEP (MMP-TEP) -modified allografts versus unmodified and hydrolytically degradable TEP (Hydro-TEP)-modified allografts, respectively, at 3 weeks post-surgery. MMP-TEP-modified allografts exhibited greater longitudinal graft-localized vascularization and endochondral ossification, along with 4-fold and 2-fold greater maximum torques versus unmodified and Hydro-TEP-modified allografts after 9 weeks, respectively, which was comparable to that of autografts. In summary, our results demonstrated that the MMP-TEP coordinated allograft healing via early stage recruitment and support of host neurovasculature.
Collapse
Affiliation(s)
- Yiming Li
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| | - Michael D Hoffman
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA; Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA; Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA; Materials Science Program, University of Rochester, Rochester, NY, USA; Department of Chemical Engineering, University of Rochester, Rochester, NY, USA; Department of Biomedical Genetics and Center for Oral Biology, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
37
|
Dhavalikar P, Robinson A, Lan Z, Jenkins D, Chwatko M, Salhadar K, Jose A, Kar R, Shoga E, Kannapiran A, Cosgriff-Hernandez E. Review of Integrin-Targeting Biomaterials in Tissue Engineering. Adv Healthc Mater 2020; 9:e2000795. [PMID: 32940020 PMCID: PMC7960574 DOI: 10.1002/adhm.202000795] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/27/2020] [Indexed: 12/12/2022]
Abstract
The ability to direct cell behavior has been central to the success of numerous therapeutics to regenerate tissue or facilitate device integration. Biomaterial scientists are challenged to understand and modulate the interactions of biomaterials with biological systems in order to achieve effective tissue repair. One key area of research investigates the use of extracellular matrix-derived ligands to target specific integrin interactions and induce cellular responses, such as increased cell migration, proliferation, and differentiation of mesenchymal stem cells. These integrin-targeting proteins and peptides have been implemented in a variety of different polymeric scaffolds and devices to enhance tissue regeneration and integration. This review first presents an overview of integrin-mediated cellular processes that have been identified in angiogenesis, wound healing, and bone regeneration. Then, research utilizing biomaterials are highlighted with integrin-targeting motifs as a means to direct these cellular processes to enhance tissue regeneration. In addition to providing improved materials for tissue repair and device integration, these innovative biomaterials provide new tools to probe the complex processes of tissue remodeling in order to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Prachi Dhavalikar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Robinson
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Dana Jenkins
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Malgorzata Chwatko
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Karim Salhadar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Anupriya Jose
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ronit Kar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Erik Shoga
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Aparajith Kannapiran
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | | |
Collapse
|
38
|
Sugihara K, Yamaguchi Y, Usui S, Nashimoto Y, Hanada S, Kiyokawa E, Uemura A, Yokokawa R, Nishiyama K, Miura T. A new perfusion culture method with a self-organized capillary network. PLoS One 2020; 15:e0240552. [PMID: 33112918 PMCID: PMC7592787 DOI: 10.1371/journal.pone.0240552] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/28/2020] [Indexed: 01/06/2023] Open
Abstract
A lack of perfusion has been one of the most significant obstacles for three-dimensional culture systems of organoids and embryonic tissues. Here, we developed a simple and reliable method to implement a perfusable capillary network in vitro. The method employed the self-organization of endothelial cells to generate a capillary network and a static pressure difference for culture medium circulation, which can be easily introduced to standard biological laboratories and enables long-term cultivation of vascular structures. Using this culture system, we perfused the lumen of the self-organized capillary network and observed a flow-induced vascular remodeling process, cell shape changes, and collective cell migration. We also observed an increase in cell proliferation around the self-organized vasculature induced by flow, indicating functional perfusion of the culture medium. We also reconstructed extravasation of tumor and inflammatory cells, and circulation inside spheroids including endothelial cells and human lung fibroblasts. In conclusion, this system is a promising tool to elucidate the mechanisms of various biological processes related to vascular flow.
Collapse
Affiliation(s)
- Kei Sugihara
- Department of Anatomy and Cell Biology, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yoshimi Yamaguchi
- Department of Anatomy and Cell Biology, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shiori Usui
- Department of Anatomy and Cell Biology, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yuji Nashimoto
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Miyagi, Japan
- Graduate School of Engineering, Tohoku University, Miyagi, Japan
- Department of Micro Engineering, Kyoto University, Kyoto, Japan
| | - Sanshiro Hanada
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Etsuko Kiyokawa
- Department of Oncologic Pathology, Kanazawa Medical University, Ishikawa, Japan
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Aichi, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto, Japan
| | - Koichi Nishiyama
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Takashi Miura
- Department of Anatomy and Cell Biology, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
- * E-mail:
| |
Collapse
|
39
|
Bowers SLK, Kemp SS, Aguera KN, Koller GM, Forgy JC, Davis GE. Defining an Upstream VEGF (Vascular Endothelial Growth Factor) Priming Signature for Downstream Factor-Induced Endothelial Cell-Pericyte Tube Network Coassembly. Arterioscler Thromb Vasc Biol 2020; 40:2891-2909. [PMID: 33086871 DOI: 10.1161/atvbaha.120.314517] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE In this work, we have sought to define growth factor requirements and the signaling basis for different stages of human vascular morphogenesis and maturation. Approach and Results: Using a serum-free model of endothelial cell (EC) tube morphogenesis in 3-dimensional collagen matrices that depends on a 5 growth factor combination, SCF (stem cell factor), IL (interleukin)-3, SDF (stromal-derived factor)-1α, FGF (fibroblast growth factor)-2, and insulin (factors), we demonstrate that VEGF (vascular endothelial growth factor) pretreatment of ECs for 8 hours (ie, VEGF priming) leads to marked increases in the EC response to the factors which includes; EC tip cells, EC tubulogenesis, pericyte recruitment and proliferation, and basement membrane deposition. VEGF priming requires VEGFR2, and the effect of VEGFR2 is selective to the priming response and does not affect factor-dependent tubulogenesis in the absence of priming. Key molecule and signaling requirements for VEGF priming include RhoA, Rock1 (Rho-kinase), PKCα (protein kinase C α), and PKD2 (protein kinase D2). siRNA suppression or pharmacological blockade of these molecules and signaling pathways interfere with the ability of VEGF to act as an upstream primer of downstream factor-dependent EC tube formation as well as pericyte recruitment. VEGF priming was also associated with the formation of actin stress fibers, activation of focal adhesion components, upregulation of the EC factor receptors, c-Kit, IL-3Rα, and CXCR4 (C-X-C chemokine receptor type 4), and upregulation of EC-derived PDGF (platelet-derived growth factor)-BB, PDGF-DD, and HB-EGF (heparin-binding epidermal growth factor) which collectively affect pericyte recruitment and proliferation. CONCLUSIONS Overall, this study defines a signaling signature for a separable upstream VEGF priming step, which can activate ECs to respond to downstream factors that are necessary to form branching tube networks with associated mural cells.
Collapse
Affiliation(s)
- Stephanie L K Bowers
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Scott S Kemp
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Kalia N Aguera
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Gretchen M Koller
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Joshua C Forgy
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - George E Davis
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
40
|
Kemp SS, Aguera KN, Cha B, Davis GE. Defining Endothelial Cell-Derived Factors That Promote Pericyte Recruitment and Capillary Network Assembly. Arterioscler Thromb Vasc Biol 2020; 40:2632-2648. [PMID: 32814441 DOI: 10.1161/atvbaha.120.314948] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE We sought to identify and investigate the functional role of the major endothelial cell (EC)-derived factors that control pericyte recruitment to EC tubes and pericyte-induced tube maturation during capillary network formation. Approach and Results: We identify PDGF (platelet-derived growth factor)-BB, PDGF-DD, ET (endothelin)-1, TGF (transforming growth factor)-β, and HB-EGF (heparin-binding epidermal growth factor), as the key individual and combined regulators of pericyte assembly around EC tubes. Using novel pericyte only assays, we demonstrate that PDGF-BB, PDGF-DD, and ET-1 are the primary direct drivers of pericyte invasion. Their addition to pericytes induces invasion as if ECs were present. In contrast, TGF-β and HB-EGF have minimal ability to directly stimulate pericyte invasion. In contrast, TGF-β1 can act as an upstream pericyte primer to stimulate invasion in response to PDGFs and ET-1. HB-EGF stimulates pericyte proliferation along with PDGFs and ET-1. Using EC-pericyte cocultures, individual, or combined blockade of these EC-derived factors, or their pericyte receptors, using neutralizing antibodies or chemical inhibitors, respectively, interferes with pericyte recruitment and proliferation. As individual factors, PDGF-BB and ET-1 have the strongest impact on these events. However, when the blocking reagents are combined to interfere with each of the above factors or their receptors, more dramatic and profound blockade of pericyte recruitment, proliferation, and pericyte-induced basement membrane deposition occurs. Under these conditions, ECs form tubes that become much wider and less elongated as if pericytes were absent. CONCLUSIONS Overall, these new studies define and characterize a functional role for key EC-derived factors controlling pericyte recruitment, proliferation, and pericyte-induced basement membrane deposition during capillary network assembly.
Collapse
Affiliation(s)
- Scott S Kemp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Kalia N Aguera
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - Byeong Cha
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| | - George E Davis
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
41
|
Sailem HZ, Al Haj Zen A. Morphological landscape of endothelial cell networks reveals a functional role of glutamate receptors in angiogenesis. Sci Rep 2020; 10:13829. [PMID: 32796870 PMCID: PMC7428010 DOI: 10.1038/s41598-020-70440-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
Angiogenesis plays a key role in several diseases including cancer, ischemic vascular disease, and Alzheimer's disease. Chemical genetic screening of endothelial tube formation provides a robust approach for identifying signalling components that impact microvascular network morphology as well as endothelial cell biology. However, the analysis of the resulting imaging datasets has been limited to a few phenotypic features such as the total tube length or the number of branching points. Here we developed a high content analysis framework for detailed quantification of various aspects of network morphology including network complexity, symmetry and topology. By applying our approach to a high content screen of 1,280 characterised drugs, we found that drugs that result in a similar phenotype share the same mechanism of action or common downstream signalling pathways. Our multiparametric analysis revealed that a group of glutamate receptor antagonists enhances branching and network connectivity. Using an integrative meta-analysis approach, we validated the link between these receptors and angiogenesis. We further found that the expression of these genes is associated with the prognosis of Alzheimer's patients. In conclusion, our work shows that detailed image analysis of complex endothelial phenotypes can reveal new insights into biological mechanisms modulating the morphogenesis of endothelial networks and identify potential therapeutics for angiogenesis-related diseases.
Collapse
Affiliation(s)
- Heba Z Sailem
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7LF, UK.
| | - Ayman Al Haj Zen
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Doha, Qatar.
- Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK.
| |
Collapse
|
42
|
Wright ME, Yu JK, Jain D, Maeda A, Yeh SCA, DaCosta RS, Lin CP, Santerre JP. Engineering functional microvessels in synthetic polyurethane random-pore scaffolds by harnessing perfusion flow. Biomaterials 2020; 256:120183. [PMID: 32622017 DOI: 10.1016/j.biomaterials.2020.120183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/24/2022]
Abstract
Recently reported biomaterial-based approaches toward prevascularizing tissue constructs rely on biologically or structurally complex scaffolds that are complicated to manufacture and sterilize, and challenging to customize for clinical applications. In the current work, a prevascularization method for soft tissue engineering that uses a non-patterned and non-biological scaffold is proposed. Human fibroblasts and HUVECs were seeded on an ionomeric polyurethane-based hydrogel and cultured for 14 days under medium perfusion. A flow rate of 0.05 mL/min resulted in a greater lumen density in the constructs relative to 0.005 and 0.5 mL/min, indicating the critical importance of flow magnitude in establishing microvessels. Constructs generated at 0.05 mL/min perfusion flow were implanted in a mouse subcutaneous model and intravital imaging was used to characterize host blood perfusion through the construct after 2 weeks. Engineered microvessels were functional (i.e. perfused with host blood and non-leaky) and neovascularization of the construct by host vessels was enhanced relative to non-prevascularized constructs. We report on the first strategy toward engineering functional microvessels in a tissue construct using non-bioactive, non-patterned synthetic polyurethane materials.
Collapse
Affiliation(s)
- Meghan Ee Wright
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Jonathan K Yu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Devika Jain
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Azusa Maeda
- Princess Margaret Cancer Centre and Techna Institute, University Health Network, Toronto, Canada
| | - Shu-Chi A Yeh
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ralph S DaCosta
- Princess Margaret Cancer Centre and Techna Institute, University Health Network, Toronto, Canada
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - J Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
43
|
Braile M, Cristinziano L, Marcella S, Varricchi G, Marone G, Modestino L, Ferrara AL, De Ciuceis A, Scala S, Galdiero MR, Loffredo S. LPS-mediated neutrophil VEGF-A release is modulated by cannabinoid receptor activation. J Leukoc Biol 2020; 109:621-631. [PMID: 32573828 DOI: 10.1002/jlb.3a0520-187r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/14/2020] [Accepted: 05/28/2020] [Indexed: 12/15/2022] Open
Abstract
Neutrophils (PMNs) are innate immune cells with primary roles in inflammation and in host defense against infections. Both inflammatory and tumor angiogenesis are modulated by a sequential, coordinated production of angiogenic factors such as vascular endothelial growth factors (VEGFs), angiopoietins, hepatocyte growth factor (HGF), and chemokines. These factors are produced by several immune cells, including PMNs. Activation of cannabinoid receptor type-1 (CB1 ) and -2 (CB2 ) has been suggested as a new strategy to modulate in vitro and in vivo angiogenesis. We sought to investigate whether activation of CB1 and CB2 by CB agonists modulate LPS-mediated angiogenic activity of human PMNs. Highly purified PMNs were isolated from buffy coats of healthy donors. Cells were stimulated with CB1 and CB2 agonists/antagonists alone and/or in combination with LPS. Angiogenic factors in cell-free supernatants were measured by ELISA. The modulation of activation markers of PMNs by CB agonists was evaluated by flow cytometry. Angiogenesis in vitro was measured as tube formation by optical microscopy. Endothelial cell permeability was assessed by an in vitro vascular permeability assay. LPS-activated PMNs released VEGF-A, CXCL8, and HGF. Preincubation of PMNs with low concentrations of CB1 and CB2 agonists inhibited VEGF-A release induced by LPS, but did not affect CXCL8 and HGF production. The effects of CB agonists on VEGF-A release induced by LPS were reversed by preincubation with CB antagonists. CB agonists modulated in vitro angiogenesis and endothelial permeability induced by supernatants of LPS-activated PMNs through the reduction of VEGF-A. Neutrophils play a central role in the control of bacterial infections and in the outcome of sepsis. The latter condition is associated with an increase in circulating levels of VEGF-A. We demonstrated that low concentrations of CB agonists inhibit VEGF-A release from LPS-activated PMNs. These results suggest that CB agonists might represent a novel therapeutic strategy in patients with sepsis.
Collapse
Affiliation(s)
- Mariantonia Braile
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Leonardo Cristinziano
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Simone Marcella
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,CNR Institute of Experimental Endocrinology and Oncology "G. Salvatore", Naples, Italy
| | - Giancarlo Marone
- Department of Public Health, University of Naples Federico II, Italy.,Azienda Ospedaliera Ospedali dei Colli-Monaldi Hospital Pharmacy, Naples, Italy
| | - Luca Modestino
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Agnese De Ciuceis
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Sara Scala
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,CNR Institute of Experimental Endocrinology and Oncology "G. Salvatore", Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,CNR Institute of Experimental Endocrinology and Oncology "G. Salvatore", Naples, Italy
| |
Collapse
|
44
|
Control of endothelial tubulogenesis by Rab and Ral GTPases, and apical targeting of caveolin-1-labeled vacuoles. PLoS One 2020; 15:e0235116. [PMID: 32569321 PMCID: PMC7307772 DOI: 10.1371/journal.pone.0235116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 06/08/2020] [Indexed: 12/26/2022] Open
Abstract
Here, we examine known GTPase regulators of vesicle trafficking events to assess whether they affect endothelial cell (EC) lumen and tube formation. We identify novel roles for the small GTPases Rab3A, Rab3B, Rab8A, Rab11A, Rab27A, RalA, RalB and caveolin-1 in co-regulating membrane trafficking events that control EC lumen and tube formation. siRNA suppression of individual GTPases such as Rab3A, Rab8A, and RalB markedly inhibit tubulogenesis, while greater blockade is observed with combinations of siRNAs such as Rab3A and Rab3B, Rab8A and Rab11A, and RalA and RalB. These combinations of siRNAs also disrupt very early events in lumen formation including the formation of intracellular vacuoles. In contrast, knockdown of the endocytosis regulator, Rab5A, fails to inhibit EC tube formation. Confocal microscopy and real-time videos reveal that caveolin-1 strongly labels intracellular vacuoles and localizes to the EC apical surface as they fuse to form the luminal membrane. In contrast, Cdc42 and Rab11A localize to a perinuclear, subapical region where intracellular vacuoles accumulate and fuse during lumen formation. Our new data demonstrates that EC tubulogenesis is coordinated by a series of small GTPases to control polarized membrane trafficking events to generate, deliver, and fuse caveolin-1-labeled vacuoles to create the apical membrane surface.
Collapse
|
45
|
Gonzalez-Avila G, Sommer B, García-Hernández AA, Ramos C. Matrix Metalloproteinases' Role in Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1245:97-131. [PMID: 32266655 DOI: 10.1007/978-3-030-40146-7_5] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer cells evolve in the tumor microenvironment (TME) by the acquisition of characteristics that allow them to initiate their passage through a series of events that constitute the metastatic cascade. For this purpose, tumor cells maintain a crosstalk with TME non-neoplastic cells transforming them into their allies. "Corrupted" cells such as cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), and tumor-associated neutrophils (TANs) as well as neoplastic cells express and secrete matrix metalloproteinases (MMPs). Moreover, TME metabolic conditions such as hypoxia and acidification induce MMPs' synthesis in both cancer and stromal cells. MMPs' participation in TME consists in promoting events, for example, epithelial-mesenchymal transition (EMT), apoptosis resistance, angiogenesis, and lymphangiogenesis. MMPs also facilitate tumor cell migration through the basement membrane (BM) and extracellular matrix (ECM). The aim of the present chapter is to discuss MMPs' contribution to the evolution of cancer cells, their cellular origin, and their influence in the main processes that take place in the TME.
Collapse
Affiliation(s)
- Georgina Gonzalez-Avila
- Laboratorio de Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico.
| | - Bettina Sommer
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | - A Armando García-Hernández
- Laboratorio de Oncología Biomédica, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| | - Carlos Ramos
- Laboratorio de Biología Celular, Departamento de Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico
| |
Collapse
|
46
|
Blatchley MR, Gerecht S. Reconstructing the Vascular Developmental Milieu In Vitro. Trends Cell Biol 2020; 30:15-31. [DOI: 10.1016/j.tcb.2019.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 10/14/2019] [Indexed: 12/25/2022]
|
47
|
Dihydroartemisinin inhibits endothelial cell tube formation by suppression of the STAT3 signaling pathway. Life Sci 2019; 242:117221. [PMID: 31881224 DOI: 10.1016/j.lfs.2019.117221] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 12/16/2019] [Accepted: 12/21/2019] [Indexed: 12/31/2022]
Abstract
AIMS Endothelial cell (EC) tube formation is crucial for tumor angiogenesis, which becomes a target for chemotherapy. The anti-malaria agent dihydroartemisinin (DHA) inhibited tumor growth and angiogenesis. The aim of this study was to investigate the effects of DHA on EC tube formation and the underlying mechanisms. MATERIALS AND METHODS Human umbilical vein endothelial cells (HUVECs) were cultured with different concentrations of DHA, and the tube formation was measured by in vitro angiogenesis assay. The protein levels of signal transducer and activator of transcription factor 3 (STAT3), phosphorylated STAT3 and fatty acid synthase (FASN) were detected by Western blotting. The gene expression of FASN was determined by real time-polymerase chain reaction (RT-PCR). The FASN siRNA and STAT3 (Y705D) vector were introduced into HUVECs by lipofectin transfection. KEY FINDINGS DHA treatment inhibited tube formation, and the phosphorylation of STAT3 on Y705 of HUVECs. The expression of FASN was down-regulated by DHA and STAT3 inhibitor. The inhibitory effect of DHA on FASN expression in HUVECs was eliminated by co-treatment with the STAT3 inhibitor. Over-expression of STAT3 (Y705D) relieved the inhibitory effect of DHA on tube-formation and FASN expression. Under hypoxia condition, expression of FASN was up-regulated but inhibited by DHA treatment in HUVECs through suppression of STAT3 phosphorylation. SIGNIFICANCE We demonstrate that DHA inhibits the protein level of FASN via attenuation of the Y705 phosphorylation of STAT3, and subsequently inhibits tube formation of HUVECs. Our results support the therapeutic potential of DHA on angiogenesis.
Collapse
|
48
|
Koller GM, Schafer C, Kemp SS, Aguera KN, Lin PK, Forgy JC, Griffin CT, Davis GE. Proinflammatory Mediators, IL (Interleukin)-1β, TNF (Tumor Necrosis Factor) α, and Thrombin Directly Induce Capillary Tube Regression. Arterioscler Thromb Vasc Biol 2019; 40:365-377. [PMID: 31852224 DOI: 10.1161/atvbaha.119.313536] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE In this work, we examine the molecular basis for capillary tube regression and identify key proregressive factors, signaling pathways, and pharmacological antagonists of this process. Approach and Results: We demonstrate that the proinflammatory mediators, IL (interleukin)-1β, TNF (tumor necrosis factor) α, and thrombin, singly and in combination, are potent regulators of capillary tube regression in vitro. These proregressive factors, when added to endothelial cell-pericyte cocultures, led to selective loss of endothelial cell-lined tube networks, with retention and proliferation of pericytes despite the marked destruction of adjacent capillary tubes. Moreover, treatment of macrophages with the TLR (toll-like receptor) agonists Pam3CSK4 and lipopolysaccharide generates conditioned media with marked proregressive activity, that is completely blocked by a combination of neutralizing antibodies directed to IL-1β and TNFα but not to other factors. The same combination of blocking antibodies, as well as the anti-inflammatory cytokine IL-10, interfere with macrophage-dependent hyaloid vasculature regression in mice suggesting that proinflammatory cytokine signaling regulates capillary regression in vivo. In addition, we identified a capillary regression signaling signature in endothelial cells downstream of these proregressive agents that is characterized by increased levels of ICAM-1 (intercellular adhesion molecule-1), phospho-p38, and phospho-MLC2 (myosin light chain-2) and decreased levels of phospho-Pak2, acetylated tubulin, phospho-cofilin, and pro-caspase3. Finally, we identified combinations of pharmacological agents (ie, FIST and FISTSB) that markedly rescue the proregressive activities of IL-1β, TNFα, and thrombin, individually and in combination. CONCLUSIONS Overall, these new studies demonstrate that the major proinflammatory mediators, IL-1β, TNFα, and thrombin, are key regulators of capillary tube regression-a critical pathological process regulating human disease.
Collapse
Affiliation(s)
- Gretchen M Koller
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa (G.M.K., S.S.K., K.N.A., P.K.L., J.C.F., G.E.D.)
| | - Christopher Schafer
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (C.S., C.T.G.), University of Oklahoma Health Sciences Center
| | - Scott S Kemp
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa (G.M.K., S.S.K., K.N.A., P.K.L., J.C.F., G.E.D.)
| | - Kalia N Aguera
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa (G.M.K., S.S.K., K.N.A., P.K.L., J.C.F., G.E.D.)
| | - Prisca K Lin
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa (G.M.K., S.S.K., K.N.A., P.K.L., J.C.F., G.E.D.)
| | - Joshua C Forgy
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa (G.M.K., S.S.K., K.N.A., P.K.L., J.C.F., G.E.D.)
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation (C.S., C.T.G.), University of Oklahoma Health Sciences Center.,Department of Cell Biology (C.T.G.), University of Oklahoma Health Sciences Center
| | - George E Davis
- From the Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida School of Medicine, Tampa (G.M.K., S.S.K., K.N.A., P.K.L., J.C.F., G.E.D.)
| |
Collapse
|
49
|
Gholobova D, Terrie L, Gerard M, Declercq H, Thorrez L. Vascularization of tissue-engineered skeletal muscle constructs. Biomaterials 2019; 235:119708. [PMID: 31999964 DOI: 10.1016/j.biomaterials.2019.119708] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 12/10/2019] [Accepted: 12/18/2019] [Indexed: 12/26/2022]
Abstract
Skeletal muscle tissue can be created in vitro by tissue engineering approaches, based on differentiation of muscle stem cells. Several approaches exist and generally result in three dimensional constructs composed of multinucleated myofibers to which we refer as myooids. Engineering methods date back to 3 decades ago and meanwhile a wide range of cell types and scaffold types have been evaluated. Nevertheless, in most approaches, myooids remain very small to allow for diffusion-mediated nutrient supply and waste product removal, typically less than 1 mm thick. One of the shortcomings of current in vitro skeletal muscle organoid development is the lack of a functional vascular structure, thus limiting the size of myooids. This is a challenge which is nowadays applicable to almost all organoid systems. Several approaches to obtain a vascular structure within myooids have been proposed. The purpose of this review is to give a concise overview of these approaches.
Collapse
Affiliation(s)
- D Gholobova
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium
| | - L Terrie
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium
| | - M Gerard
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium
| | - H Declercq
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium
| | - L Thorrez
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium.
| |
Collapse
|
50
|
Stephens CJ, Spector JA, Butcher JT. Biofabrication of thick vascularized neo-pedicle flaps for reconstructive surgery. Transl Res 2019; 211:84-122. [PMID: 31170376 PMCID: PMC6702068 DOI: 10.1016/j.trsl.2019.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 01/01/2023]
Abstract
Wound chronicity due to intrinsic and extrinsic factors perturbs adequate lesion closure and reestablishment of the protective skin barrier. Immediate and proper care of chronic wounds is necessary for a swift recovery and a reduction of patient vulnerability to infection. Advanced therapies supplemented with standard wound care procedures have been clinically implemented to restore aberrant tissue; however, these treatments are ineffective if local vasculature is too compromised to support minimally-invasive strategies. Autologous "flaps", which are tissues equipped with their own hierarchical vascular supply, can be harvested from one region of the patient and transplanted to the wound where it is reperfused upon microsurgical anastomosis to appropriate recipient vessels. Despite the success of autologous flap transfer, these procedures are extremely invasive, incur obligatory donor-site morbidity, and require sufficient donor-tissue availability, microsurgical expertise, and specialized equipment. 3D-bioprinting modalities, such as extrusion-based bioprinting, can be used to address the clinical constraints of autologous flap transfer, primarily addressing donor-site morbidity and tissue availability. This advancement in regenerative medicine allows the biofabrication of heterogeneous tissue structures with high shape fidelity and spatial resolution to generate biomimetic constructs with the anatomically-precise geometries of native tissue to ensure tissue-specific function. Yet, meaningful progress toward this clinical application has been limited by the lack of vascularization required to meet the nutrient and oxygen demands of clinically relevant tissue volumes. Thus, various criteria for the fabrication of functional tissues with hierarchical, patent vasculature must be considered when implementing 3D-bioprinting technologies for deep, chronic wounds.
Collapse
Affiliation(s)
- Chelsea J Stephens
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jason A Spector
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York; Division of Plastic Surgery, Weill Cornell Medical College, New York, New York
| | - Jonathan T Butcher
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.
| |
Collapse
|