101
|
Michael P, Yang N, Moore M, Santos M, Lam YT, Ward A, Hung JC, Tan R, Wise S. Synthetic Vascular Graft with Spatially Distinct Architecture for Rapid Biomimetic Cell Organisation in a Perfusion Bioreactor. Biomed Mater 2022; 17. [PMID: 35413704 DOI: 10.1088/1748-605x/ac66b2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 04/12/2022] [Indexed: 11/12/2022]
Abstract
Access to lab-grown fully functional blood vessels would provide an invaluable resource to vascular medicine. The complex architecture and cellular makeup of native vessels, however, makes this extremely challenging to reproduce in vitro. Bioreactor systems have helped advanced research in this area by replicating many of the physiological conditions necessary for full-scale tissue growth outside of the body. A key element underpinning these technologies are 3D vascular graft templates which serve as temporary scaffolds to direct cell growth into similar cellular architectures observed in native vessels. Grafts further engineered with appropriate physical cues to accommodate the multiple cell types that reside within native vessels may help improve the production efficiency and physiological accuracy of bioreactor-grown vessel substitutes. Here, we engineered two distinct scaffold architectures into an electrospun vascular graft aiming to encourage the spatial organisation of human vascular endothelial cells (hCAECs) in a continuous luminal monolayer, co-cultured with human fibroblasts (hFBs) populating the graft wall. Using an electrospun composite of polycaprolactone (PCL) and gelatin, we evaluated physical parameters including fibre thickness, fibre alignment, and porosity, that best mimicked the spatial composition and growth of hCAECs and hFBs in native vessels. Upon identifying the optimal scaffold architectures for each cell type, we constructed a custom-designed mandrel that combined these distinct architectures into a single vascular graft during a single electrospinning processing run. When connected to a perfusion bioreactor system, the dual architecture graft spatially oriented hCAECs and hFBs into the graft wall and lumen, respectively, directly from circulation. This biomimetic cell organisation was consistent with positive graft remodelling with significant collagen deposition in the graft wall. These findings demonstrate the influence of architectural cues to direct cell growth within vascular graft templates and the future potential of these approaches to more accurately and efficiency produce blood vessel substitutes in bioreactor systems.
Collapse
Affiliation(s)
- Praveesuda Michael
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Nianji Yang
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Matthew Moore
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Miguel Santos
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Yuen Ting Lam
- The University of Sydney Charles Perkins Centre, John Hopkins Drive, CAMPERDOWN, Sydney, 2006, AUSTRALIA
| | - Annabelle Ward
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Jui Chien Hung
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Richard Tan
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| | - Steven Wise
- The University of Sydney, John Hopkins Drive, CAMPERDOWN, Sydney, New South Wales, 2006, AUSTRALIA
| |
Collapse
|
102
|
Valdoz JC, Franks NA, Cribbs CG, Jacobs DJ, Dodson EL, Knight CJ, Poulson PD, Garfield SR, Johnson BC, Hemeyer BM, Sudo MT, Saunooke JA, Kartchner BC, Saxton A, Vallecillo-Zuniga ML, Santos M, Chamberlain B, Christensen KA, Nordin GP, Narayanan AS, Raghu G, Van Ry PM. Soluble ECM promotes organotypic formation in lung alveolar model. Biomaterials 2022; 283:121464. [DOI: 10.1016/j.biomaterials.2022.121464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/15/2022] [Accepted: 03/06/2022] [Indexed: 11/25/2022]
|
103
|
Zhu G, Lin Y, Ge T, Singh S, Liu H, Fan L, Wang S, Rhen J, Jiang D, Lyu Y, Yin Y, Li X, Benoit DSW, Li W, Xu Y, Pang J. A novel peptide inhibitor of Dll4-Notch1 signalling and its pro-angiogenic functions. Br J Pharmacol 2022; 179:1716-1731. [PMID: 34796471 PMCID: PMC9040338 DOI: 10.1111/bph.15743] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE The Dll4-Notch1 signalling pathway plays an important role in sprouting angiogenesis, vascular remodelling and arterial or venous specificity. Genetic or pharmacological inhibition of Dll4-Notch1 signalling leads to excessive sprouting angiogenesis. However, transcriptional inhibitors of Dll4-Notch1 signalling have not been described. EXPERIMENTAL APPROACH We designed a new peptide targeting Notch signalling, referred to as TAT-ANK, and assessed its effects on angiogenesis. In vitro, tube formation and fibrin gel bead assay were carried out, using human umbilical vein endothelial cells (HUVECs). In vivo, Matrigel plug angiogenesis assay, a developmental retinal model and tumour models in mice were used. The mechanisms underlying TAT-ANK activity were investigated by immunochemistry, western blotting, immunoprecipitation, RT-qPCR and luciferase reporter assays. KEY RESULTS The amino acid residues 179-191 in the G-protein-coupled receptor-kinase-interacting protein-1 (GIT1-ankyrin domain) are crucial for GIT1 binding to the Notch transcription repressor, RBP-J. We designed the peptide TAT-ANK, based on residues 179-191 in GIT1. TAT-ANK significantly inhibited Dll4 expression and Notch 1 activation in HUVECs by competing with activated Notch1 to bind to RBP-J. The analyses of biological functions showed that TAT-ANK promoted angiogenesis in vitro and in vivo by inhibiting Dll4-Notch1 signalling. CONCLUSIONS AND IMPLICATIONS We synthesized and investigated the biological actions of TAT-ANK peptide, a new inhibitor of Notch signalling. This peptide will be of significant interest to research on Dll4-Notch1 signalling and to clinicians carrying out clinical trials using Notch signalling inhibitors. Furthermore, our findings will have important conceptual and therapeutic implications for angiogenesis-related diseases.
Collapse
Affiliation(s)
- Guofu Zhu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Lin
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tandi Ge
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shekhar Singh
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Liu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Linlin Fan
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shumin Wang
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Jordan Rhen
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Dongyang Jiang
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuyan Lyu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiheng Yin
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiankai Li
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Danielle S. W. Benoit
- Departments of Biomedical Engineering and Chemical Engineering, Materials Science Program, and Centers for Musculoskeletal Research and Oral Biology, University of Rochester, Rochester, New York, USA
| | - Weiming Li
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yawei Xu
- Department of Cardiology, Pan-Vascular Research Institute of Tongji University, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinjiang Pang
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
104
|
Li R, Wang TY, Xu X, Emery OM, Yi M, Wu SP, DeMayo FJ. Spatial transcriptomic profiles of mouse uterine microenvironments at pregnancy day 7.5†. Biol Reprod 2022; 107:529-545. [PMID: 35357464 PMCID: PMC9382390 DOI: 10.1093/biolre/ioac061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 02/03/2022] [Accepted: 03/30/2022] [Indexed: 01/17/2023] Open
Abstract
Uterine dysfunctions lead to fertility disorders and pregnancy complications. Normal uterine functions at pregnancy depend on crosstalk among multiple cell types in uterine microenvironments. Here, we performed the spatial transcriptomics and single-cell RNA-seq assays to determine local gene expression profiles at the embryo implantation site of the mouse uterus on pregnancy day 7.5 (D7.5). The spatial transcriptomic annotation identified 11 domains of distinct gene signatures, including a mesometrial myometrium, an anti-mesometrial myometrium, a mesometrial decidua enriched with natural killer cells, a vascular sinus zone for maternal vessel remodeling, a fetal-maternal interface, a primary decidual zone, a transition decidual zone, a secondary decidual zone, undifferentiated stroma, uterine glands, and the embryo. The scRNA-Seq identified 12 types of cells in the D7.5 uterus including three types of stromal fibroblasts with differentiated and undifferentiated markers, one cluster of epithelium including luminal and glandular epithelium, mesothelium, endothelia, pericytes, myelomonocytic cell, natural killer cells, and lymphocyte B. These single-cell RNA signatures were then utilized to deconvolute the cell-type compositions of each individual uterine microenvironment. Functional annotation assays on spatial transcriptomic data revealed uterine microenvironments with distinguished metabolic preferences, immune responses, and various cellular behaviors that are regulated by region-specific endocrine and paracrine signals. Global interactome among regions is also projected based on the spatial transcriptomic data. This study provides high-resolution transcriptome profiles with locality information at the embryo implantation site to facilitate further investigations on molecular mechanisms for normal pregnancy progression.
Collapse
Affiliation(s)
- Rong Li
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Tian-yuan Wang
- Integrative Bioinformatics Supportive Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Xin Xu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Olivia M Emery
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - MyeongJin Yi
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Francesco J DeMayo
- Correspondence: Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, 111 T. W. Alexander Dr., Research Triangle Park, NC 27709, USA. Tel: +9842873987; E-mail:
| |
Collapse
|
105
|
Multi-Layered Human Blood Vessels-on-Chip Design Using Double Viscous Finger Patterning. Biomedicines 2022; 10:biomedicines10040797. [PMID: 35453546 PMCID: PMC9027030 DOI: 10.3390/biomedicines10040797] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 12/26/2022] Open
Abstract
Blood vessel-on-a-chip models aim at reproducing vascular functions. However, very few efficient methods have been designed to address the need for biological replicates in medium- to high-throughput screenings. Here, vessels-on-chip were designed in polydimethylsiloxane-glass chips using the viscous finger patterning technique which was adapted to create channels with various internal diameters inside a collagen solution and to simultaneously seed cells. This method was refined to create blood vessels composed of two concentric, distinct, and closely appositioned layers of human endothelial and perivascular cells arranged around a hollow lumen. These approaches allowed the formation of structurally correct blood vessels-on-chips which were constituted of either only endothelial cells or of both cell types in order to distinguish the vascular barrier reactivity to drugs in the presence or not of perivascular cells. The established vessels showed a tight vascular barrier, as assessed by immunostaining of the adherens junctions, and were reactive to the natural vasopermeant thrombin and to inflammatory cytokines. The presence of perivascular cells markedly increased the tightness of the vascular barrier and lowered its response to thrombin. The design allowed us to simultaneously challenge in real-time several tens of 3D-reconstituted, multicellular blood vessels in a standard multiwell plate format suitable for high-throughput drug screening.
Collapse
|
106
|
Contribution of Endothelial Laminin-Binding Integrins to Cellular Processes Associated with Angiogenesis. Cells 2022; 11:cells11050816. [PMID: 35269439 PMCID: PMC8909174 DOI: 10.3390/cells11050816] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/14/2022] [Accepted: 02/23/2022] [Indexed: 11/17/2022] Open
Abstract
Endothelial cells engage extracellular matrix and basement membrane components through integrin-mediated adhesion to promote angiogenesis. Angiogenesis involves the sprouting of endothelial cells from pre-existing vessels, their migration into surrounding tissue, the upregulation of angiogenesis-associated genes, and the formation of new endothelial tubes. To determine whether the endothelial laminin-binding integrins, α6β4, and α3β1 contribute to these processes, we employed RNAi technology in organotypic angiogenesis assays, as well in migration assays, in vitro. The endothelial depletion of either α6β4 or α3β1 inhibited endothelial sprouting, indicating that these integrins have non-redundant roles in this process. Interestingly, these phenotypes were accompanied by overlapping and distinct changes in the expression of angiogenesis-associated genes. Lastly, depletion of α6β4, but not α3β1, inhibited migration. Taken together, these results suggest that laminin-binding integrins regulate processes associated with angiogenesis by distinct and overlapping mechanisms.
Collapse
|
107
|
Karkanrood MV, Homayouni Tabrizi M, Ardalan T, Soltani M, Khadem F, Nosrat T, Moeini S. Pistacia atlantica fruit essential oil nanoemulsions (PAEO-NE), an effective antiangiogenic therapeutic and cell-dependent apoptosis inducer on A549 human lung cancer cells. INORG NANO-MET CHEM 2022. [DOI: 10.1080/24701556.2022.2034008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
| | | | - Touran Ardalan
- Department of Chemistry, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Mozhgan Soltani
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Toktam Nosrat
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Soheila Moeini
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
- Department of Biology, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| |
Collapse
|
108
|
Arab WT, Susapto HH, Alhattab D, Hauser CAE. Peptide nanogels as a scaffold for fabricating dermal grafts and 3D vascularized skin models. J Tissue Eng 2022; 13:20417314221111868. [PMID: 35923174 PMCID: PMC9340315 DOI: 10.1177/20417314221111868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Millions of people worldwide suffer from skin injuries, which create significant problems in their lives and are costly to cure. Tissue engineering is a promising approach that aims to fabricate functional organs using biocompatible scaffolds. We designed ultrashort tetrameric peptides with promising properties required for skin tissue engineering. Our work aimed to test the efficacy of these scaffolds for the fabrication of dermal grafts and 3D vascularized skin tissue models. We found that the direct contact of keratinocytes and fibroblasts enhanced the proliferation of the keratinocytes. Moreover, the expression levels of TGF-β1, b-FGF, IL-6, and IL-1α is correlated with the growth of the fibroblasts and keratinocytes in the co-culture. Furthermore, we successfully produced a 3D vascularized skin co-culture model using these peptide scaffolds. We believe that the described results represent an advancement in the fabrication of skin tissue equivalent, thereby providing the opportunity to rebuild missing, failing, or damaged parts. Graphical abstract
Collapse
Affiliation(s)
- Wafaa T Arab
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Hepi H Susapto
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computational Bioscience Research Center (CBRC), KAUST, Thuwal, Saudi Arabia
| | - Dana Alhattab
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computational Bioscience Research Center (CBRC), KAUST, Thuwal, Saudi Arabia
| | - Charlotte A E Hauser
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computational Bioscience Research Center (CBRC), KAUST, Thuwal, Saudi Arabia
| |
Collapse
|
109
|
Dorrier CE, Jones HE, Pintarić L, Siegenthaler JA, Daneman R. Emerging roles for CNS fibroblasts in health, injury and disease. Nat Rev Neurosci 2022; 23:23-34. [PMID: 34671105 PMCID: PMC8527980 DOI: 10.1038/s41583-021-00525-w] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2021] [Indexed: 02/06/2023]
Abstract
Recent transcriptomic, histological and functional studies have begun to shine light on the fibroblasts present in the meninges, choroid plexus and perivascular spaces of the brain and spinal cord. Although the origins and functions of CNS fibroblasts are still being described, it is clear that they represent a distinct cell population, or populations, that have likely been confused with other cell types on the basis of the expression of overlapping cellular markers. Recent work has revealed that fibroblasts play crucial roles in fibrotic scar formation in the CNS after injury and inflammation, which have also been attributed to other perivascular cell types such as pericytes and vascular smooth muscle cells. In this Review, we describe the current knowledge of the location and identity of CNS perivascular cell types, with a particular focus on CNS fibroblasts, including their origin, subtypes, roles in health and disease, and future areas for study.
Collapse
Affiliation(s)
- Cayce E Dorrier
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Hannah E Jones
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lucija Pintarić
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Julie A Siegenthaler
- Department of Pediatrics Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Cell Biology, Stem Cells and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Richard Daneman
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
110
|
Zhang Y, Wang H, Oliveira RHM, Zhao C, Popel AS. Systems biology of angiogenesis signaling: Computational models and omics. WIREs Mech Dis 2021; 14:e1550. [PMID: 34970866 PMCID: PMC9243197 DOI: 10.1002/wsbm.1550] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 01/10/2023]
Abstract
Angiogenesis is a highly regulated multiscale process that involves a plethora of cells, their cellular signal transduction, activation, proliferation, differentiation, as well as their intercellular communication. The coordinated execution and integration of such complex signaling programs is critical for physiological angiogenesis to take place in normal growth, development, exercise, and wound healing, while its dysregulation is critically linked to many major human diseases such as cancer, cardiovascular diseases, and ocular disorders; it is also crucial in regenerative medicine. Although huge efforts have been devoted to drug development for these diseases by investigation of angiogenesis‐targeted therapies, only a few therapeutics and targets have proved effective in humans due to the innate multiscale complexity and nonlinearity in the process of angiogenic signaling. As a promising approach that can help better address this challenge, systems biology modeling allows the integration of knowledge across studies and scales and provides a powerful means to mechanistically elucidate and connect the individual molecular and cellular signaling components that function in concert to regulate angiogenesis. In this review, we summarize and discuss how systems biology modeling studies, at the pathway‐, cell‐, tissue‐, and whole body‐levels, have advanced our understanding of signaling in angiogenesis and thereby delivered new translational insights for human diseases. This article is categorized under:Cardiovascular Diseases > Computational Models Cancer > Computational Models
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebeca Hannah M Oliveira
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
111
|
Dong J, Pacella M, Liu Y, Zhao L. Surface engineering and the application of laser-based processes to stents - A review of the latest development. Bioact Mater 2021; 10:159-184. [PMID: 34901537 PMCID: PMC8636930 DOI: 10.1016/j.bioactmat.2021.08.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/04/2021] [Accepted: 08/20/2021] [Indexed: 12/21/2022] Open
Abstract
Late in-stent thrombus and restenosis still represent two major challenges in stents’ design. Surface treatment of stent is attracting attention due to the increasing importance of stenting intervention for coronary artery diseases. Several surface engineering techniques have been utilised to improve the biological response in vivo on a wide range of biomedical devices. As a tailorable, precise, and ultra-fast process, laser surface engineering offers the potential to treat stent materials and fabricate various 3D textures, including grooves, pillars, nanowires, porous and freeform structures, while also modifying surface chemistry through nitridation, oxidation and coatings. Laser-based processes can reduce the biodegradable materials' degradation rate, offering many advantages to improve stents’ performance, such as increased endothelialisation rate, prohibition of SMC proliferation, reduced platelet adhesion and controlled corrosion and degradation. Nowadays, adequate research has been conducted on laser surface texturing and surface chemistry modification. Laser texturing on commercial stents has been also investigated and a promotion of performance of laser-textured stents has been proved. In this critical review, the influence of surface texture and surface chemistry on stents performance is firstly reviewed to understand the surface characteristics of stents required to facilitate cellular response. This is followed by the explicit illustration of laser surface engineering of stents and/or related materials. Laser induced periodic surface structure (LIPSS) on stent materials is then explored, and finally the application of laser surface modification techniques on latest generation of stent devices is highlighted to provide future trends and research direction on laser surface engineering of stents. Compared conventional surface engineering with laser-based methods for biomedical devices. Explained the influence of texture geometry and surface chemistry on stents biological response. Reviewed state of the art in laser surface engineering of stents for improved biological response. Reviewed state of the art in laser surface engineering to control degradation of bioresorbable stents. Highlighted novel laser surface engineering designs for improved stents'performance.
Collapse
Affiliation(s)
- J Dong
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - M Pacella
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - Y Liu
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK.,Centre for Biological Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| | - L Zhao
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, Leicestershire, LE11 3TU, UK
| |
Collapse
|
112
|
Ha SS, Kim JH, Savitri C, Choi D, Park K. Nano-Sized Extracellular Matrix Particles Lead to Therapeutic Improvement for Cutaneous Wound and Hindlimb Ischemia. Int J Mol Sci 2021; 22:ijms222413265. [PMID: 34948061 PMCID: PMC8705579 DOI: 10.3390/ijms222413265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 11/25/2022] Open
Abstract
Cell-derived matrix (CDM) has proven its therapeutic potential and been utilized as a promising resource in tissue regeneration. In this study, we prepared a human fibroblast-derived matrix (FDM) by decellularization of in vitro cultured cells and transformed the FDM into a nano-sized suspended formulation (sFDM) using ultrasonication. The sFDM was then homogeneously mixed with Pluronic F127 and hyaluronic acid (HA), to effectively administer sFDM into target sites. Both sFDM and sFDM containing hydrogel (PH/sFDM) were characterized via immunofluorescence, sol–gel transition, rheological analysis, and biochemical factors array. We found that PH/sFDM hydrogel has biocompatible, mechanically stable, injectable properties and can be easily administered into the external and internal target regions. sFDM itself holds diverse bioactive molecules. Interestingly, sFDM-containing serum-free media helped maintain the metabolic activity of endothelial cells significantly better than those in serum-free condition. PH/sFDM also promoted vascular endothelial growth factor (VEGF) secretion from monocytes in vitro. Moreover, when we evaluated therapeutic effects of PH/sFDM via the murine full-thickness skin wound model, regenerative potential of PH/sFDM was supported by epidermal thickness, significantly more neovessel formation, and enhanced mature collagen deposition. The hindlimb ischemia model also found some therapeutic improvements, as assessed by accelerated blood reperfusion and substantially diminished necrosis and fibrosis in the gastrocnemius and tibialis muscles. Together, based on sFDM holding a strong therapeutic potential, our engineered hydrogel (PH/sFDM) should be a promising candidate in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Sang Su Ha
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (S.S.H.); (C.S.)
| | - Jung-Hyun Kim
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Cininta Savitri
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (S.S.H.); (C.S.)
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
| | - Donghoon Choi
- Division of Cardiology, Department of Internal Medicine, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Korea;
- Correspondence: (D.C.); (K.P.)
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea; (S.S.H.); (C.S.)
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Korea
- Correspondence: (D.C.); (K.P.)
| |
Collapse
|
113
|
Song M, Finley SD. Mechanistic characterization of endothelial sprouting mediated by pro-angiogenic signaling. Microcirculation 2021; 29:e12744. [PMID: 34890488 PMCID: PMC9285777 DOI: 10.1111/micc.12744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 11/04/2021] [Accepted: 12/01/2021] [Indexed: 11/30/2022]
Abstract
Objective We aim to quantitatively characterize the crosstalk between VEGF‐ and FGF‐mediated angiogenic signaling and endothelial sprouting, to gain mechanistic insights and identify novel therapeutic strategies. Methods We constructed an experimentally validated hybrid agent‐based mathematical model that characterizes endothelial sprouting driven by FGF‐ and VEGF‐mediated signaling. We predicted the total sprout length, number of sprouts, and average length by the mono‐ and co‐stimulation of FGF and VEGF. Results The experimentally fitted and validated model predicts that FGF induces stronger angiogenic responses in the long‐term compared with VEGF stimulation. Also, FGF plays a dominant role in the combination effects in endothelial sprouting. Moreover, the model suggests that ERK and Akt pathways and cellular responses contribute differently to the sprouting process. Last, the model predicts that the strategies to modulate endothelial sprouting are context‐dependent, and our model can identify potential effective pro‐ and anti‐angiogenic targets under different conditions and study their efficacy. Conclusions The model provides detailed mechanistic insight into VEGF and FGF interactions in sprouting angiogenesis. More broadly, this model can be utilized to identify targets that influence angiogenic signaling leading to endothelial sprouting and to study the effects of pro‐ and anti‐angiogenic therapies.
Collapse
Affiliation(s)
- Min Song
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Stacey D Finley
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.,Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA.,Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
114
|
Rijal G. Understanding the Role of Fibroblasts following a 3D Tumoroid Implantation for Breast Tumor Formation. Bioengineering (Basel) 2021; 8:bioengineering8110163. [PMID: 34821729 PMCID: PMC8615023 DOI: 10.3390/bioengineering8110163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 11/16/2022] Open
Abstract
An understanding of the participation and modulation of fibroblasts during tumor formation and growth is still unclear. Among many speculates, one might be the technical challenge to reveal the versatile function of fibroblasts in tissue complexity, and another is the dynamics in tissue physiology and cell activity. The histology of most solid tumors shows a predominant presence of fibroblasts, suggesting that tumor cells recruit fibroblasts for breast tumor growth. In this review paper, therefore, the migration, activation, differentiation, secretion, and signaling systems that are associated with fibroblasts and cancer-associated fibroblasts (CAFs) after implantation of a breast tumoroid, i.e., a lab-generated tumor tissue into an animal, are discussed.
Collapse
Affiliation(s)
- Girdhari Rijal
- Department of Medical Laboratory Sciences and Public Health, Tarleton State University, a Member of Texas A & M University System, Fort Worth, TX 76104, USA
| |
Collapse
|
115
|
Dikici S, Aldemir Dikici B, MacNeil S, Claeyssens F. Decellularised extracellular matrix decorated PCL PolyHIPE scaffolds for enhanced cellular activity, integration and angiogenesis. Biomater Sci 2021; 9:7297-7310. [PMID: 34617526 PMCID: PMC8547328 DOI: 10.1039/d1bm01262b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Wound healing involves a complex series of events where cell–cell and cell-extracellular matrix (ECM) interactions play a key role. Wounding can be simple, such as the loss of the epithelial integrity, or deeper and more complex, reaching to subcutaneous tissues, including blood vessels, muscles and nerves. Rapid neovascularisation of the wounded area is crucial for wound healing as it has a key role in supplying oxygen and nutrients during the highly demanding proliferative phase and transmigration of inflammatory cells to the wound area. One approach to circumvent delayed neovascularisation is the exogenous use of pro-angiogenic factors, which is expensive, highly dose-dependent, and the delivery of them requires a very well-controlled system to avoid leaky, highly permeable and haemorrhagic blood vessel formation. In this study, we decorated polycaprolactone (PCL)-based polymerised high internal phase emulsion (PolyHIPE) scaffolds with fibroblast-derived ECM to assess fibroblast, endothelial cell and keratinocyte activity in vitro and angiogenesis in ex ovo chick chorioallantoic membrane (CAM) assays. Our results showed that the inclusion of ECM in the scaffolds increased the metabolic activity of three types of cells that play a key role in wound healing and stimulated angiogenesis in ex ovo CAM assays over 7 days. Herein, we demonstrated that fibroblast-ECM functionalised PCL PolyHIPE scaffolds appear to have great potential to be used as an active wound dressing to promote angiogenesis and wound healing. Decellularisation of in vitro generated extracellular matrix (ECM) provides an effective way to stimulate angiogenesis and wound healing.![]()
Collapse
Affiliation(s)
- Serkan Dikici
- Department of Bioengineering, Izmir Institute of Technology, Izmir, 35430, Turkey. .,Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield, S3 7HQ, UK.
| | - Betül Aldemir Dikici
- Department of Bioengineering, Izmir Institute of Technology, Izmir, 35430, Turkey. .,Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield, S3 7HQ, UK.
| | - Sheila MacNeil
- Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield, S3 7HQ, UK.
| | - Frederik Claeyssens
- Department of Materials Science and Engineering, University of Sheffield, Kroto Research Institute, Sheffield, S3 7HQ, UK.
| |
Collapse
|
116
|
Kim S, Lee S, Lim J, Choi H, Kang H, Jeon NL, Son Y. Human bone marrow-derived mesenchymal stem cells play a role as a vascular pericyte in the reconstruction of human BBB on the angiogenesis microfluidic chip. Biomaterials 2021; 279:121210. [PMID: 34710793 DOI: 10.1016/j.biomaterials.2021.121210] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 10/07/2021] [Accepted: 10/20/2021] [Indexed: 12/16/2022]
Abstract
A blood-brain barrier (BBB) on a chip similar to the in vivo BBB is important for evaluating the efficacy of reparative cell therapeutics for ischemic stroke in vitro. In this study, we established human BBB-like microvasculature on an angiogenesis microfluidic chip and analyzed the role of human pericytes (hPCs) and human astrocytes (hACs) on the architecture of human brain microvascular endothelial cells (hBMEC)-derived microvasculature on a chip. We found that human bone marrow mesenchymal stem cells (hBM-MSCs) play a role as perivascular pericytes in tight BBB reformation with a better vessel-constrictive capacity than that of hPCs, providing evidence of reparative stem cells on BBB repair rather than a paracrine effect. We also demonstrated that pericytes play an important role in vessel constriction, and astrocytes may induce the maturation of a capillary network. Higher expression of VEGF, SDF-1α, PDGFRβ, N-cadherin, and α-SMA in hBM-MSCs than in hPCs and their subsequent downregulation with hBMEC co-culture suggest that hBM-MSCs may be better recruited and engaged in the BBB-microvasculature than hPCs. Collectively, the human BBB on a chip may be adopted as an alternative to evaluate in vitro cellular behavior and the engagement of cell therapeutics in BBB regeneration and may also be used for studying stroke.
Collapse
Affiliation(s)
- Sumin Kim
- Department of Genetic Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea
| | - Somin Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, South Korea
| | - Jungeun Lim
- Department of Mechanical Engineering, Seoul National University, Seoul, 08826, South Korea
| | - Hyeri Choi
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, South Korea
| | - Habin Kang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, South Korea
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, South Korea; Department of Mechanical Engineering, Seoul National University, Seoul, 08826, South Korea
| | - Youngsook Son
- Department of Genetic Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Yong in, 17104, South Korea; Kyung Hee Institute of Regenerative Medicine (KIRM), Medical Science Research Institute, Kyung Hee University Hospital, Seoul, 02447, South Korea.
| |
Collapse
|
117
|
Im J, Hyun J, Kim SW, Bhang SH. Enhancing the Angiogenic and Proliferative Capacity of Dermal Fibroblasts with Mulberry (Morus alba. L) Root Extract. Tissue Eng Regen Med 2021; 19:49-57. [PMID: 34674183 DOI: 10.1007/s13770-021-00404-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/14/2021] [Accepted: 09/23/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Enhancing blood flow and cell proliferation in the hair dermis is critical for treating hair loss. This study was designed to aid the development of alternative and effective solutions to overcome alopecia. Specifically, we examined the effects of Morus alba. L root extract (MARE, which has been used in traditional medicine as a stimulant for hair proliferation) on dermal fibroblasts and other cell types found in the epidermis. METHODS We first optimized the concentration of MARE that could be used to treat human dermal fibroblasts (HDFs) without causing cytotoxicity. After optimization, we focused on the effect of MARE on HDFs since these cells secrete paracrine factors related to cell proliferation and angiogenesis that affect hair growth. Conditioned medium (CM) derived from MARE-treated HDFs (MARE HDF-CM) was used to treat human umbilical vein endothelial cells (HUVECs) and hair follicle dermal papilla cells (HFDPCs). RESULTS Concentrations of MARE up to 20 wt% increased the expression of proliferative and anti-apoptotic genes in HDFs. MARE HDF-CM significantly improved the tubular structure formation and migration capacity of HUVECs. Additionally, MARE HDF-CM treatment upregulated the expression of hair growth-related genes in HFDPCs. CM collected from MARE-treated HDFs promoted the proliferation of HFDPCs and the secretion of angiogenic paracrine factors from these cells. CONCLUSION Since it can stimulate the secretion of pro-proliferative and pro-angiogenic paracrine factors from HDFs, MARE has therapeutic potential as a hair loss preventative.
Collapse
Affiliation(s)
- Jisoo Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Sung-Won Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea.
| |
Collapse
|
118
|
Walji N, Kheiri S, Young EWK. Angiogenic Sprouting Dynamics Mediated by Endothelial-Fibroblast Interactions in Microfluidic Systems. Adv Biol (Weinh) 2021; 5:e2101080. [PMID: 34655165 DOI: 10.1002/adbi.202101080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/18/2021] [Indexed: 11/09/2022]
Abstract
Angiogenesis, the development of new blood vessels from existing vasculature, is a key process in normal development and pathophysiology. In vitro models are necessary for investigating mechanisms of angiogenesis and developing antiangiogenic therapies. Microfluidic cell culture models of angiogenesis are favored for their ability to recapitulate 3D tissue structures and control spatiotemporal aspects of the microenvironments. To capture the angiogenesis process, microfluidic models often include endothelial cells and a fibroblast component. However, the influence of fibroblast organization on resulting angiogenic behavior remains unclear. Here a comparative study of angiogenic sprouting on a microfluidic chip induced by fibroblasts in 2D monolayer, 3D dispersed, and 3D spheroid culture formats, is conducted. Vessel morphology and sprout distribution for each configuration are measured, and these observations are correlated with measurements of secreted factors and numerical simulations of diffusion gradients. The results demonstrate that angiogenic sprouting varies in response to fibroblast organization with correlating variations in secretory profile and secreted factor gradients across the microfluidic device. This study is anticipated to shed light on how sprouting dynamics are mediated by fibroblast configuration such that the microfluidic cell culture design process includes the selection of a fibroblast component where the effects are known and leveraged.
Collapse
Affiliation(s)
- Noosheen Walji
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, M5S 3G8, Canada.,Institute of Biomedical Engineering, University of Toronto, 160 College St., Toronto, M5S 3E1, Canada
| | - Sina Kheiri
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, M5S 3G8, Canada
| | - Edmond W K Young
- Department of Mechanical and Industrial Engineering, University of Toronto, 5 King's College Road, Toronto, M5S 3G8, Canada.,Institute of Biomedical Engineering, University of Toronto, 160 College St., Toronto, M5S 3E1, Canada
| |
Collapse
|
119
|
Moreira HR, Marques AP. Vascularization in skin wound healing: where do we stand and where do we go? Curr Opin Biotechnol 2021; 73:253-262. [PMID: 34555561 DOI: 10.1016/j.copbio.2021.08.019] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/23/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022]
Abstract
Cutaneous healing is a highly complex process that, if altered due to, for example, impaired vascularization, results in chronic wounds or repaired neotissue of poor quality. Significant progress has been achieved in promoting neotissue vascularization during tissue repair/regeneration. In this review, we discuss the strategies that have been explored and how each one of them contributes to regulate vascularization in the context of cutaneous wound healing from two different perspectives - biomaterial-based and a cell-based approaches. Finally, we discuss the implications of these findings on the development of the 'next generation' approaches to target vascularization in wound healing highlighting the importance of going beyond its contribution to regulate vascularization and take into consideration the temporal features of the healing process and of different types of wounds.
Collapse
Affiliation(s)
- Helena R Moreira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, Guimarães 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Avepark - Zona Industrial da Gandra, Guimarães 4805-017, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal.
| |
Collapse
|
120
|
Kempers L, van der Bijl I, van Stalborch AMD, Ponsioen B, Margadant C. Fast in vitro protocol for the visualization and quantitative high-throughput analysis of sprouting angiogenesis by confocal microscopy. STAR Protoc 2021; 2:100690. [PMID: 34557696 PMCID: PMC8445886 DOI: 10.1016/j.xpro.2021.100690] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
We describe an optimized, cost-effective, reproducible, and robust protocol to study sprouting angiogenesis in glass-bottom 96-well plates by confocal microscopy, ideal for screening of drug or shRNA libraries. Effective and stable knockdown of gene expression in primary endothelial cells is achieved by lentiviral transduction. Dynamic behavior of individual cells and fluorescent proteins is analyzed by time-lapse imaging, while competitive advantages in tip cell formation are assessed using mixtures of differentially labeled cell populations. Finally, we present a macro for high-throughput analysis. For complete information on the use and execution of this protocol, please refer to van der Bijl et al. (2020) and Kempers et al. (2021). High-throughput microscopy analysis of sprouting angiogenesis for drug/shRNA screening Analysis of dynamic cell and protein behavior during sprouting by time-lapse microscopy Mosaic assays to image competitive advantages in tip cell behavior Macro for fast automated quantitative analysis
Collapse
Affiliation(s)
- Lanette Kempers
- Sanquin Research and Landsteiner laboratory, Amsterdam University Medical Center, 1066 CX Amsterdam, The Netherlands
| | - Ivo van der Bijl
- Sanquin Research and Landsteiner laboratory, Amsterdam University Medical Center, 1066 CX Amsterdam, The Netherlands
| | - Anne-Marieke D van Stalborch
- Sanquin Research and Landsteiner laboratory, Amsterdam University Medical Center, 1066 CX Amsterdam, The Netherlands
| | - Bas Ponsioen
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Coert Margadant
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
121
|
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: 25] [Impact Index Per Article: 6.3] [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
|
122
|
A robust vasculogenic microfluidic model using human immortalized endothelial cells and Thy1 positive fibroblasts. Biomaterials 2021; 276:121032. [PMID: 34303155 PMCID: PMC9891349 DOI: 10.1016/j.biomaterials.2021.121032] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 02/03/2023]
Abstract
Human umbilical vein endothelial cells (HUVECs) and stromal cells, such as human lung fibroblasts (FBs), have been widely used to generate functional microvascular networks (μVNs) in vitro. However, primary cells derived from different donors have batch-to-batch variations and limited lifespans when cultured in vitro, which hampers the reproducibility of μVN formation. Here, we immortalize HUVECs and FBs by exogenously expressing human telomerase reverse transcriptase (hTERT) to obtain stable endothelial cell and FB sources for μVN formation in vitro. Interestingly, we find that immortalized HUVECs can only form functional μVNs with immortalized FBs from earlier passages but not from later passages. Mechanistically, we show that Thy1 expression decreases in FBs from later passages. Compared to Thy1 negative FBs, Thy1 positive FBs express higher IGFBP2, IGFBP7, and SPARC, which are important for angiogenesis and lumen formation during vasculogenesis in 3D. Moreover, Thy1 negative FBs physically block microvessel openings, reducing the perfusability of μVNs. Finally, by culturing immortalized FBs on gelatin-coated surfaces in serum-free medium, we are able to maintain the majority of Thy1 positive immortalized FBs to support perfusable μVN formation. Overall, we establish stable cell sources for μVN formation and characterize the functions of Thy1 positive and negative FBs in vasculogenesis in vitro.
Collapse
|
123
|
Bogunovic N, Meekel JP, Majolée J, Hekhuis M, Pyszkowski J, Jockenhövel S, Kruse M, Riesebos E, Micha D, Blankensteijn JD, Hordijk PL, Ghazanfari S, Yeung KK. Patient-Specific 3-Dimensional Model of Smooth Muscle Cell and Extracellular Matrix Dysfunction for the Study of Aortic Aneurysms. J Endovasc Ther 2021; 28:604-613. [PMID: 33902345 PMCID: PMC8276336 DOI: 10.1177/15266028211009272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Abdominal aortic aneurysms (AAAs) are associated with overall high mortality in case of rupture. Since the pathophysiology is unclear, no adequate pharmacological therapy exists. Smooth muscle cells (SMCs) dysfunction and extracellular matrix (ECM) degradation have been proposed as underlying causes. We investigated SMC spatial organization and SMC-ECM interactions in our novel 3-dimensional (3D) vascular model. We validated our model for future use by comparing it to existing 2-dimensional (2D) cell culture. Our model can be used for translational studies of SMC and their role in AAA pathophysiology. MATERIALS AND METHODS SMC isolated from the medial layer of were the aortic wall of controls and AAA patients seeded on electrospun poly-lactide-co-glycolide scaffolds and cultured for 5 weeks, after which endothelial cells (EC) are added. Cell morphology, orientation, mechanical properties and ECM production were quantified for validation and comparison between controls and patients. RESULTS We show that cultured SMC proliferate into multiple layers after 5 weeks in culture and produce ECM proteins, mimicking their behavior in the medial aortic layer. EC attach to multilayered SMC, mimicking layer interactions. The novel SMC model exhibits viscoelastic properties comparable to biological vessels; cytoskeletal organization increases during the 5 weeks in culture; increased cytoskeletal alignment and decreased ECM production indicate different organization of AAA patients' cells compared with control. CONCLUSION We present a valuable preclinical model of AAA constructed with patient specific cells with applications in both translational research and therapeutic developments. We observed SMC spatial reorganization in a time course of 5 weeks in our robust, patient-specific model of SMC-EC organization and ECM production.
Collapse
Affiliation(s)
- Natalija Bogunovic
- Amsterdam Cardiovascular Sciences, Department of Vascular Surgery, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Department of Physiology, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Department of Clinical Genetics, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Jorn P. Meekel
- Amsterdam Cardiovascular Sciences, Department of Vascular Surgery, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Department of Physiology, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Jisca Majolée
- Amsterdam Cardiovascular Sciences, Department of Physiology, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Marije Hekhuis
- Amsterdam Cardiovascular Sciences, Department of Clinical Genetics, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | | | - Stefan Jockenhövel
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Geleen, The Netherlands
- Department of Biohybrid & Medical Textiles (Biotex), RWTH Aachen University, Aachen, Germany
| | - Magnus Kruse
- Department of Biohybrid & Medical Textiles (Biotex), RWTH Aachen University, Aachen, Germany
- Institut für Textiltechnik der RWTH Aachen University, Aachen, Germany
| | - Elise Riesebos
- Amsterdam Cardiovascular Sciences, Department of Clinical Genetics, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Dimitra Micha
- Amsterdam Cardiovascular Sciences, Department of Clinical Genetics, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Jan D. Blankensteijn
- Amsterdam Cardiovascular Sciences, Department of Vascular Surgery, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Peter L. Hordijk
- Amsterdam Cardiovascular Sciences, Department of Physiology, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| | - Samaneh Ghazanfari
- Aachen-Maastricht Institute for Biobased Materials, Faculty of Science and Engineering, Maastricht University, Geleen, The Netherlands
- Department of Biohybrid & Medical Textiles (Biotex), RWTH Aachen University, Aachen, Germany
| | - Kak K. Yeung
- Amsterdam Cardiovascular Sciences, Department of Vascular Surgery, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Department of Physiology, Amsterdam University Medical Centers, Location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
124
|
Lagoutte P, Bettler E, Vadon-Le Goff S, Moali C. Procollagen C-proteinase enhancer-1 (PCPE-1), a potential biomarker and therapeutic target for fibrosis. Matrix Biol Plus 2021; 11:100062. [PMID: 34435180 PMCID: PMC8377038 DOI: 10.1016/j.mbplus.2021.100062] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
The correct balance between collagen synthesis and degradation is essential for almost every aspect of life, from development to healthy aging, reproduction and wound healing. When this balance is compromised by external or internal stress signals, it very often leads to disease as is the case in fibrotic conditions. Fibrosis occurs in the context of defective tissue repair and is characterized by the excessive, aberrant and debilitating deposition of fibril-forming collagens. Therefore, the numerous proteins involved in the biosynthesis of fibrillar collagens represent a potential and still underexploited source of therapeutic targets to prevent fibrosis. One such target is procollagen C-proteinase enhancer-1 (PCPE-1) which has the unique ability to accelerate procollagen maturation by BMP-1/tolloid-like proteinases (BTPs) and contributes to trigger collagen fibrillogenesis, without interfering with other BTP functions or the activities of other extracellular metalloproteinases. This role is achieved through a fine-tuned mechanism of action that is close to being elucidated and offers promising perspectives for drug design. Finally, the in vivo data accumulated in recent years also confirm that PCPE-1 overexpression is a general feature and early marker of fibrosis. In this review, we describe the results which presently support the driving role of PCPE-1 in fibrosis and discuss the questions that remain to be solved to validate its use as a biomarker or therapeutic target.
Collapse
Key Words
- ADAMTS, a disintegrin and metalloproteinase with thrombospondin motifs
- AS, aortic valve stenosis
- BMP, bone morphogenetic protein
- Biomarker
- CKD, chronic kidney disease
- CP, C-propeptide
- CUB, complement, Uegf, BMP-1
- CVD, cardiovascular disease
- Collagen
- DMD, Duchenne muscular dystrophy
- ECM, extracellular matrix
- EGF, epidermal growth factor
- ELISA, enzyme-linked immunosorbent assay
- Fibrillogenesis
- Fibrosis
- HDL, high-density lipoprotein
- HSC, hepatic stellate cell
- HTS, hypertrophic scar
- IPF, idiopathic pulmonary fibrosis
- LDL, low-density lipoprotein
- MI, myocardial infarction
- MMP, matrix metalloproteinase
- NASH, nonalcoholic steatohepatitis
- NTR, netrin
- OPMD, oculopharyngeal muscular dystrophy
- PABPN1, poly(A)-binding protein nuclear 1
- PCP, procollagen C-proteinase
- PCPE, procollagen C-proteinase enhancer
- PNP, procollagen N-proteinase
- Proteolysis
- SPC, subtilisin proprotein convertase
- TGF-β, transforming growth-factor β
- TIMP, tissue inhibitor of metalloproteinases
- TSPN, thrombospondin-like N-terminal
- Therapeutic target
- eGFR, estimated glomerular filtration rate
- mTLD, mammalian tolloid
- mTLL, mammalian tolloid-like
Collapse
Affiliation(s)
- Priscillia Lagoutte
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Emmanuel Bettler
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Sandrine Vadon-Le Goff
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Catherine Moali
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| |
Collapse
|
125
|
Goonoo N, Gimié F, Ait-Arsa I, Cordonin C, Andries J, Jhurry D, Bhaw-Luximon A. Piezoelectric core-shell PHBV/PDX blend scaffolds for reduced superficial wound contraction and scarless tissue regeneration. Biomater Sci 2021; 9:5259-5274. [PMID: 34164641 DOI: 10.1039/d1bm00379h] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The use of non-invasive scaffold materials which can mimic the innate piezoelectric properties of biological tissues is a promising strategy to promote native tissue regeneration. Piezoelectric and cell instructive electrospun core-shell PDX/PHBV mats have been engineered to promote native tissue and skin regeneration. In depth physicochemical characterisation, in vitro and in vivo studies of a rat model showed that the 20/80 PDX/PHBV composition possessed the right balance of physicochemical and piezoelectric properties leading to enhanced fibroblast stimulation, proliferation and migration, reduced fibroblast-mediated contraction and macrophage-induced inflammation, improved keratinocyte proliferation, proper balance between endothelial cell phenotypes, decreased in vivo fibrosis and accelerated in vivo scarless wound regeneration. Overall, this study highlights the importance of exploiting cell-material interactions to match tissue biological needs to sustain the wound healing cascade.
Collapse
Affiliation(s)
- Nowsheen Goonoo
- Biomaterials, Drug Delivery and Nanotechnology Unit, Centre for Biomedical and Biomaterials Research, MSIRI Building, University of Mauritius, 80837 Réduit, Mauritius.
| | - Fanny Gimié
- Animalerie, Plateforme de recherche CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, Ile de La Réunion, France
| | - Imade Ait-Arsa
- Animalerie, Plateforme de recherche CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, Ile de La Réunion, France
| | - Colette Cordonin
- Animalerie, Plateforme de recherche CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, Ile de La Réunion, France
| | - Jessica Andries
- RIPA, Plateforme de recherche CYROI, 2 rue Maxime Rivière, 97490 Sainte Clotilde, Ile de La Réunion, France
| | - Dhanjay Jhurry
- Biomaterials, Drug Delivery and Nanotechnology Unit, Centre for Biomedical and Biomaterials Research, MSIRI Building, University of Mauritius, 80837 Réduit, Mauritius.
| | - Archana Bhaw-Luximon
- Biomaterials, Drug Delivery and Nanotechnology Unit, Centre for Biomedical and Biomaterials Research, MSIRI Building, University of Mauritius, 80837 Réduit, Mauritius.
| |
Collapse
|
126
|
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: 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: 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
|
127
|
King O, Sunyovszki I, Terracciano CM. Vascularisation of pluripotent stem cell-derived myocardium: biomechanical insights for physiological relevance in cardiac tissue engineering. Pflugers Arch 2021; 473:1117-1136. [PMID: 33855631 PMCID: PMC8245389 DOI: 10.1007/s00424-021-02557-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/22/2022]
Abstract
The myocardium is a diverse environment, requiring coordination between a variety of specialised cell types. Biochemical crosstalk between cardiomyocytes (CM) and microvascular endothelial cells (MVEC) is essential to maintain contractility and healthy tissue homeostasis. Yet, as myocytes beat, heterocellular communication occurs also through constantly fluctuating biomechanical stimuli, namely (1) compressive and tensile forces generated directly by the beating myocardium, and (2) pulsatile shear stress caused by intra-microvascular flow. Despite endothelial cells (EC) being highly mechanosensitive, the role of biomechanical stimuli from beating CM as a regulatory mode of myocardial-microvascular crosstalk is relatively unexplored. Given that cardiac biomechanics are dramatically altered during disease, and disruption of myocardial-microvascular communication is a known driver of pathological remodelling, understanding the biomechanical context necessary for healthy myocardial-microvascular interaction is of high importance. The current gap in understanding can largely be attributed to technical limitations associated with reproducing dynamic physiological biomechanics in multicellular in vitro platforms, coupled with limited in vitro viability of primary cardiac tissue. However, differentiation of CM from human pluripotent stem cells (hPSC) has provided an unlimited source of human myocytes suitable for designing in vitro models. This technology is now converging with the diverse field of tissue engineering, which utilises in vitro techniques designed to enhance physiological relevance, such as biomimetic extracellular matrix (ECM) as 3D scaffolds, microfluidic perfusion of vascularised networks, and complex multicellular architectures generated via 3D bioprinting. These strategies are now allowing researchers to design in vitro platforms which emulate the cell composition, architectures, and biomechanics specific to the myocardial-microvascular microenvironment. Inclusion of physiological multicellularity and biomechanics may also induce a more mature phenotype in stem cell-derived CM, further enhancing their value. This review aims to highlight the importance of biomechanical stimuli as determinants of CM-EC crosstalk in cardiac health and disease, and to explore emerging tissue engineering and hPSC technologies which can recapitulate physiological dynamics to enhance the value of in vitro cardiac experimentation.
Collapse
Affiliation(s)
- Oisín King
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK.
| | - Ilona Sunyovszki
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| | - Cesare M Terracciano
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
128
|
Ramirez-Calderon G, Susapto HH, Hauser CAE. Delivery of Endothelial Cell-Laden Microgel Elicits Angiogenesis in Self-Assembling Ultrashort Peptide Hydrogels In Vitro. ACS APPLIED MATERIALS & INTERFACES 2021; 13:29281-29292. [PMID: 34142544 DOI: 10.1021/acsami.1c03787] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Blood vessel generation is an essential process for tissue formation, regeneration, and repair. Notwithstanding, vascularized tissue fabrication in vitro remains a challenge, as current fabrication techniques and biomaterials lack translational potential in medicine. Naturally derived biomaterials harbor the risk of immunogenicity and pathogen transmission, while synthetic materials need functionalization or blending to improve their biocompatibility. In addition, the traditional top-down fabrication techniques do not recreate the native tissue microarchitecture. Self-assembling ultrashort peptides (SUPs) are promising chemically synthesized natural materials that self-assemble into three-dimensional nanofibrous hydrogels resembling the extracellular matrix (ECM). Here, we use a modular tissue-engineering approach, embedding SUP microgels loaded with human umbilical vein endothelial cells (HUVECs) into a 3D SUP hydrogel containing human dermal fibroblast neonatal (HDFn) cells to trigger angiogenesis. The SUPs IVFK and IVZK were used to fabricate microgels that gel in a water-in-oil emulsion using a microfluidic droplet generator chip. The fabricated SUP microgels are round structures that are 300-350 μm diameter in size and have ECM-like topography. In addition, they are stable enough to keep their original size and shape under cell culture conditions and long-term storage. When the SUP microgels were used as microcarriers for growing HUVECs and HDFn cells on the microgel surface, cell attachment, stretching, and proliferation could be demonstrated. Finally, we performed an angiogenesis assay in both SUP hydrogels using all SUP combinations between micro- and bulky hydrogels. Endothelial cells were able to migrate from the microgel to the surrounding area, showing angiogenesis features such as sprouting, branching, coalescence, and lumen formation. Although both SUP hydrogels support vascular network formation, IVFK outperformed IVZK in terms of vessel network extension and branching. Overall, these results demonstrated that cell-laden SUP microgels have great potential to be used as a microcarrier cell delivery system, encouraging us to study the angiogenesis process and to develop vascularized tissue-engineering therapies.
Collapse
|
129
|
Ferreira CF, Wong D, Binkley LH. "Crawling Attachment" during Periodontally Accelerated Osteogenic Orthodontics Procedure. Contemp Clin Dent 2021; 12:179-183. [PMID: 34220160 PMCID: PMC8237815 DOI: 10.4103/ccd.ccd_376_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/21/2020] [Indexed: 12/04/2022] Open
Abstract
The present clinical case reports an increased zone of keratinized gingiva that was generated following surgical excision of the gingiva during periodontally accelerated osteogenic orthodontics. The present case consists of angle Class I with mal-aligned teeth and impacted #11. The patient was evaluated up to 2 years and 1 month (2.1) showing a stable increased zone of keratinized tissue. Possible causes for this event are discussed in this case report. Additional long-term clinical studies are necessary to support these results.
Collapse
Affiliation(s)
- Cimara Fortes Ferreira
- Department of Periodontology, University of Tennessee Health Sciences College of Dentistry, Memphis, TN, USA
| | - David Wong
- Private Practice, Post-Graduate Program in Periodontics, University of Tennessee Health Sciences College of Dentistry, Memphis, TN, USA
| | - Lesley H Binkley
- Department of Periodontology, University of Tennessee Health Sciences College of Dentistry, Memphis, TN, USA
| |
Collapse
|
130
|
Zhu J, Wang Y, Zhong L, Pan F, Wang J. Advances in tissue engineering of vasculature through three-dimensional bioprinting. Dev Dyn 2021; 250:1717-1738. [PMID: 34115420 DOI: 10.1002/dvdy.385] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/07/2021] [Accepted: 06/03/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND A significant challenge facing tissue engineering is the fabrication of vasculature constructs which contains vascularized tissue constructs to recapitulate viable, complex and functional organs or tissues, and free-standing vascular structures potentially providing clinical applications in the future. Three-dimensional (3D) bioprinting has emerged as a promising technology, possessing a number of merits that other conventional biofabrication methods do not have. Over the last decade, 3D bioprinting has contributed a variety of techniques and strategies to generate both vascularized tissue constructs and free-standing vascular structures. RESULTS This review focuses on different strategies to print two kinds of vasculature constructs, namely vascularized tissue constructs and vessel-like tubular structures, highlighting the feasibility and shortcoming of the current methods for vasculature constructs fabrication. Generally, both direct printing and indirect printing can be employed in vascularized tissue engineering. Direct printing allows for structural fabrication with synchronous cell seeding, while indirect printing is more effective in generating complex architecture. During the fabrication process, 3D bioprinting techniques including extrusion bioprinting, inkjet bioprinting and light-assisted bioprinting should be selectively implemented to exert advantages and obtain the desirable tissue structure. Also, appropriate cells and biomaterials matter a lot to match various bioprinting techniques and thus achieve successful fabrication of specific vasculature constructs. CONCLUSION The 3D bioprinting has been developed to help provide various fabrication techniques, devoting to producing structurally stable, physiologically relevant, and biologically appealing constructs. However, although the optimization of biomaterials and innovation of printing strategies may improve the fabricated vessel-like structures, 3D bioprinting is still in the infant period and has a great gap between in vitro trials and in vivo applications. The article reviews the present achievement of 3D bioprinting in generating vasculature constructs and also provides perspectives on future directions of advanced vasculature constructs fabrication.
Collapse
Affiliation(s)
- Junjin Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuting Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linna Zhong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fangwei Pan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
131
|
Iturriaga-Goyon E, Buentello-Volante B, Magaña-Guerrero FS, Garfias Y. Future Perspectives of Therapeutic, Diagnostic and Prognostic Aptamers in Eye Pathological Angiogenesis. Cells 2021; 10:cells10061455. [PMID: 34200613 PMCID: PMC8227682 DOI: 10.3390/cells10061455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 12/23/2022] Open
Abstract
Aptamers are single-stranded DNA or RNA oligonucleotides that are currently used in clinical trials due to their selectivity and specificity to bind small molecules such as proteins, peptides, viral particles, vitamins, metal ions and even whole cells. Aptamers are highly specific to their targets, they are smaller than antibodies and fragment antibodies, they can be easily conjugated to multiple surfaces and ions and controllable post-production modifications can be performed. Aptamers have been therapeutically used for age-related macular degeneration, cancer, thrombosis and inflammatory diseases. The aim of this review is to highlight the therapeutic, diagnostic and prognostic possibilities associated with aptamers, focusing on eye pathological angiogenesis.
Collapse
Affiliation(s)
- Emilio Iturriaga-Goyon
- MD/PhD (PECEM) Program, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology, Conde de Valenciana, Chimalpopoca 14, Mexico City 06800, Mexico; (B.B.-V.); (F.S.M.-G.)
- Department of Biochemistry, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City 04510, Mexico
| | - Beatriz Buentello-Volante
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology, Conde de Valenciana, Chimalpopoca 14, Mexico City 06800, Mexico; (B.B.-V.); (F.S.M.-G.)
| | - Fátima Sofía Magaña-Guerrero
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology, Conde de Valenciana, Chimalpopoca 14, Mexico City 06800, Mexico; (B.B.-V.); (F.S.M.-G.)
| | - Yonathan Garfias
- Cell and Tissue Biology, Research Unit, Institute of Ophthalmology, Conde de Valenciana, Chimalpopoca 14, Mexico City 06800, Mexico; (B.B.-V.); (F.S.M.-G.)
- Department of Biochemistry, Facultad de Medicina, Universidad Nacional Autónoma de México, Av. Universidad 3000, Mexico City 04510, Mexico
- Correspondence:
| |
Collapse
|
132
|
Imashiro C, Azuma T, Itai S, Kuribara T, Totani K, Onoe H, Takemura K. Travelling ultrasound promotes vasculogenesis of three-dimensional-monocultured human umbilical vein endothelial cells. Biotechnol Bioeng 2021; 118:3760-3769. [PMID: 34110012 PMCID: PMC8518538 DOI: 10.1002/bit.27852] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 05/05/2021] [Accepted: 06/07/2021] [Indexed: 12/31/2022]
Abstract
To generate three‐dimensional tissue in vitro, promoting vasculogenesis in cell aggregates is an important factor. Here, we found that ultrasound promoted vasculogenesis of human umbilical vein endothelial cells (HUVECs). Promotion of HUVEC network formation and lumen formation were observed using our method. In addition to morphological evaluations, protein expression was quantified by western blot assays. As a result, expression of proteins related to vasculogenesis and the response to mechanical stress on cells was enhanced by exposure to ultrasound. Although several previous studies have shown that ultrasound may promote vasculogenesis, the effect of ultrasound was unclear because of unregulated ultrasound, the complex culture environment, or two‐dimensional‐cultured HUVECs that cannot form a lumen structure. In this study, regulated ultrasound was propagated on three‐dimensional‐monocultured HUVECs, which clarified the effect of ultrasound on vasculogenesis. We believe this finding may be an innovation in the tissue engineering field.
Collapse
Affiliation(s)
- Chikahiro Imashiro
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo, Shinjuku-ku, Japan.,Department of Mechanical Engineering, Keio University, Yokohama, Kohoku-ku, Japan
| | - Tetsuya Azuma
- Department of Mechanical Engineering, Keio University, Yokohama, Kohoku-ku, Japan
| | - Shun Itai
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, Yokohama, Kohoku-ku, Japan
| | - Taiki Kuribara
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, Tokyo, Musashino-shi, Japan
| | - Kiichiro Totani
- Department of Materials and Life Science, Faculty of Science and Technology, Seikei University, Tokyo, Musashino-shi, Japan
| | - Hiroaki Onoe
- Department of Mechanical Engineering, Keio University, Yokohama, Kohoku-ku, Japan
| | - Kenjiro Takemura
- Department of Mechanical Engineering, Keio University, Yokohama, Kohoku-ku, Japan
| |
Collapse
|
133
|
Rogers MT, Gard AL, Gaibler R, Mulhern TJ, Strelnikov R, Azizgolshani H, Cain BP, Isenberg BC, Haroutunian NJ, Raustad NE, Keegan PM, Lech MP, Tomlinson L, Borenstein JT, Charest JL, Williams C. A high-throughput microfluidic bilayer co-culture platform to study endothelial-pericyte interactions. Sci Rep 2021; 11:12225. [PMID: 34108507 PMCID: PMC8190127 DOI: 10.1038/s41598-021-90833-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/17/2021] [Indexed: 01/27/2023] Open
Abstract
Microphysiological organ-on-chip models offer the potential to improve the prediction of drug safety and efficacy through recapitulation of human physiological responses. The importance of including multiple cell types within tissue models has been well documented. However, the study of cell interactions in vitro can be limited by complexity of the tissue model and throughput of current culture systems. Here, we describe the development of a co-culture microvascular model and relevant assays in a high-throughput thermoplastic organ-on-chip platform, PREDICT96. The system consists of 96 arrayed bilayer microfluidic devices containing retinal microvascular endothelial cells and pericytes cultured on opposing sides of a microporous membrane. Compatibility of the PREDICT96 platform with a variety of quantifiable and scalable assays, including macromolecular permeability, image-based screening, Luminex, and qPCR, is demonstrated. In addition, the bilayer design of the devices allows for channel- or cell type-specific readouts, such as cytokine profiles and gene expression. The microvascular model was responsive to perturbations including barrier disruption, inflammatory stimulation, and fluid shear stress, and our results corroborated the improved robustness of co-culture over endothelial mono-cultures. We anticipate the PREDICT96 platform and adapted assays will be suitable for other complex tissues, including applications to disease models and drug discovery.
Collapse
Affiliation(s)
- Miles T Rogers
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
- Raytheon BBN Technologies, Synthetic Biology, 10 Moulton St, Cambridge, MA, 02138, USA
| | - Ashley L Gard
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Robert Gaibler
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Thomas J Mulhern
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Rivka Strelnikov
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
- Microsoft Corporation, 1 Memorial Drive, Cambridge, MA, 02142, USA
| | - Hesham Azizgolshani
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Brian P Cain
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Brett C Isenberg
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Nerses J Haroutunian
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Nicole E Raustad
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
- Department of Biology, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Philip M Keegan
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
- Department of Biomedical Engineering, University of Wisconsin Madison, 1550 Engineering Dr, Madison, WI, 53706, USA
| | | | | | - Jeffrey T Borenstein
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA
| | - Joseph L Charest
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA.
| | - Corin Williams
- The Charles Stark Draper Laboratory Inc., 555 Technology Square, Cambridge, MA, 02139, USA.
| |
Collapse
|
134
|
Franchi-Mendes T, Lopes N, Brito C. Heterotypic Tumor Spheroids in Agitation-Based Cultures: A Scaffold-Free Cell Model That Sustains Long-Term Survival of Endothelial Cells. Front Bioeng Biotechnol 2021; 9:649949. [PMID: 34178955 PMCID: PMC8219978 DOI: 10.3389/fbioe.2021.649949] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 05/06/2021] [Indexed: 12/25/2022] Open
Abstract
Endothelial cells (ECs) are an important component of the tumor microenvironment, playing key roles in tumor development and progression that span from angiogenesis to immune regulation and drug resistance. Heterotypic tumor spheroids are one of the most widely used in vitro tumor microenvironment models, presenting improved recapitulation of tumor microenvironments compared to 2D cultures, in a simple and low-cost setup. Heterotypic tumor spheroid models incorporating endothelial cells have been proposed but present multiple limitations, such as the short culture duration typically obtained, the use of animal-derived matrices, and poor reproducibility; the diversity of culture conditions employed hinders comparison between studies and standardization of relevant culture parameters. Herein, we developed long-term cultures of triple heterotypic spheroids composed of the HCC1954 tumor cell line, human fibroblasts, and ECs. We explored culture parameters potentially relevant for EC maintenance, such as tumor cell line, seeding cell number, cell ratio, and agitation vs. static culture. In HCC1954-based spheroids, we observed maintenance of viable EC for up to 1 month of culture in agitation, with retention of the identity markers CD31 and von Willebrand factor. At the optimized tumor cell:fibroblast:EC ratio of 1:3:10, HCC1954-based spheroids had a higher EC area/total spheroid area at 1 month of culture than the other cell ratios tested. EC maintenance was tumor cell line-dependent, and in HCC1954-based spheroids it was also dependent on the presence of fibroblasts and agitation. Moreover, vascular endothelial growth factor (VEGF) supplementation was not required for maintenance of EC, as the factor was endogenously produced. ECs co-localized with fibroblasts, which accumulated preferentially in the core of the spheroids and secreted EC-relevant extracellular matrix proteins, such as collagen I and IV. This simple model setup does not rely on artificial or animal-derived scaffolds and can serve as a useful tool to explore the culture parameters influencing heterotypic spheroids, contributing to model standardization, as well as to explore molecular cross talk of ECs within the tumor microenvironment, and potentially its effects on drug response.
Collapse
Affiliation(s)
- Teresa Franchi-Mendes
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Nuno Lopes
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.,The Discoveries Centre for Regenerative and Precision Medicine, Oeiras, Portugal
| |
Collapse
|
135
|
Vajda J, Milojević M, Maver U, Vihar B. Microvascular Tissue Engineering-A Review. Biomedicines 2021; 9:589. [PMID: 34064101 PMCID: PMC8224375 DOI: 10.3390/biomedicines9060589] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/14/2021] [Accepted: 05/19/2021] [Indexed: 12/31/2022] Open
Abstract
Tissue engineering and regenerative medicine have come a long way in recent decades, but the lack of functioning vasculature is still a major obstacle preventing the development of thicker, physiologically relevant tissue constructs. A large part of this obstacle lies in the development of the vessels on a microscale-the microvasculature-that are crucial for oxygen and nutrient delivery. In this review, we present the state of the art in the field of microvascular tissue engineering and demonstrate the challenges for future research in various sections of the field. Finally, we illustrate the potential strategies for addressing some of those challenges.
Collapse
Affiliation(s)
- Jernej Vajda
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (J.V.); (M.M.)
| | - Marko Milojević
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (J.V.); (M.M.)
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Uroš Maver
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (J.V.); (M.M.)
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Boštjan Vihar
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (J.V.); (M.M.)
- IRNAS Ltd., Limbuška cesta 78b, 2000 Maribor, Slovenia
| |
Collapse
|
136
|
Role of Microenvironment in Non-Hodgkin Lymphoma: Understanding the Composition and Biology. ACTA ACUST UNITED AC 2021; 26:206-216. [PMID: 32496454 DOI: 10.1097/ppo.0000000000000446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lymphoma microenvironment is a dynamic and well-orchestrated network of various immune and stromal cells that is indispensable for tumor cell survival, growth, migration, immune escape, and drug resistance. Recent progress has enhanced our knowledge of the pivotal role of microenvironment in lymphomagenesis. Understanding the characteristics, functions, and contributions of various components of the tumor niche, along with its bidirectional interactions with tumor cells, is paramount. It offers the potential to identify new therapeutic targets with the ability to restore antitumor immune surveillance and eliminate the protumoral factors contributed by the tumor niche.
Collapse
|
137
|
You S, Chaney EJ, Tu H, Sun Y, Sinha S, Boppart SA. Label-Free Deep Profiling of the Tumor Microenvironment. Cancer Res 2021; 81:2534-2544. [PMID: 33741692 PMCID: PMC8137645 DOI: 10.1158/0008-5472.can-20-3124] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/12/2021] [Accepted: 03/18/2021] [Indexed: 11/16/2022]
Abstract
Label-free nonlinear microscopy enables nonperturbative visualization of structural and metabolic contrast within living cells in their native tissue microenvironment. Here a computational pipeline was developed to provide a quantitative view of the microenvironmental architecture within cancerous tissue from label-free nonlinear microscopy images. To enable single-cell and single-extracellular vesicle (EV) analysis, individual cells, including tumor cells and various types of stromal cells, and EVs were segmented by a multiclass pixelwise segmentation neural network and subsequently analyzed for their metabolic status and molecular structure in the context of the local cellular neighborhood. By comparing cancer tissue with normal tissue, extensive tissue reorganization and formation of a patterned cell-EV neighborhood was observed in the tumor microenvironment. The proposed analytic pipeline is expected to be useful in a wide range of biomedical tasks that benefit from single-cell, single-EV, and cell-to-EV analysis. SIGNIFICANCE: The proposed computational framework allows label-free microscopic analysis that quantifies the complexity and heterogeneity of the tumor microenvironment and opens possibilities for better characterization and utilization of the evolving cancer landscape.
Collapse
Affiliation(s)
- Sixian You
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Eric J Chaney
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Haohua Tu
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Yi Sun
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Saurabh Sinha
- Departement of Computer Science, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Stephen A Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois.
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
138
|
Effects of Ipriflavone-Loaded Mesoporous Nanospheres on the Differentiation of Endothelial Progenitor Cells and Their Modulation by Macrophages. NANOMATERIALS 2021; 11:nano11051102. [PMID: 33923311 PMCID: PMC8145259 DOI: 10.3390/nano11051102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022]
Abstract
Angiogenic biomaterials are designed to promote vascularization and tissue regeneration. Nanoparticles of bioactive materials loaded with drugs represent an interesting strategy to stimulate osteogenesis and angiogenesis and to inhibit bone resorption. In this work, porcine endothelial progenitor cells (EPCs), essential for blood vessel formation, were isolated and characterized to evaluate the in vitro effects of unloaded (NanoMBGs) and ipriflavone-loaded nanospheres (NanoMBG-IPs), which were designed to prevent osteoporosis. The expression of vascular endothelial growth factor receptor 2 (VEGFR2) was studied in EPCs under different culture conditions: (a) treatment with NanoMBGs or NanoMBG-IPs, (b) culture with media from basal, M1, and M2 macrophages previously treated with NanoMBGs or NanoMBG-IPs, (c) coculture with macrophages in the presence of NanoMBGs or NanoMBG-IPs, and (d) coculture with M2d angiogenic macrophages. The endocytic mechanisms for nanosphere incorporation by EPCs were identified using six different endocytosis inhibitors. The results evidence the great potential of these nanomaterials to enhance VEGFR2 expression and angiogenesis, after intracellular incorporation by EPCs through clathrin-dependent endocytosis, phagocytosis, and caveolae-mediated uptake. The treatment of EPCs with basal, M1, and M2 macrophage culture media and EPC/macrophage coculture studies also confirmed the angiogenic effect of these nanospheres on EPCs, even in the presence of phagocytic cells.
Collapse
|
139
|
Browne S, Gill EL, Schultheiss P, Goswami I, Healy KE. Stem cell-based vascularization of microphysiological systems. Stem Cell Reports 2021; 16:2058-2075. [PMID: 33836144 PMCID: PMC8452487 DOI: 10.1016/j.stemcr.2021.03.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
Microphysiological systems (MPSs) (i.e., tissue or organ chips) exploit microfluidics and 3D cell culture to mimic tissue and organ-level physiology. The advent of human induced pluripotent stem cell (hiPSC) technology has accelerated the use of MPSs to study human disease in a range of organ systems. However, in the reduction of system complexity, the intricacies of vasculature are an often-overlooked aspect of MPS design. The growing library of pluripotent stem cell-derived endothelial cell and perivascular cell protocols have great potential to improve the physiological relevance of vasculature within MPS, specifically for in vitro disease modeling. Three strategic categories of vascular MPS are outlined: self-assembled, interface focused, and 3D biofabricated. This review discusses key features and development of the native vasculature, linking that to how hiPSC-derived vascular cells have been generated, the state of the art in vascular MPSs, and opportunities arising from interdisciplinary thinking.
Collapse
Affiliation(s)
- Shane Browne
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Elisabeth L Gill
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Paula Schultheiss
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA
| | - Ishan Goswami
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA
| | - Kevin E Healy
- Department of Bioengineering and California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
140
|
Abstract
Recreating human organ-level function in vitro is a rapidly evolving field that integrates tissue engineering, stem cell biology, and microfluidic technology to produce 3D organoids. A critical component of all organs is the vasculature. Herein, we discuss general strategies to create vascularized organoids, including common source materials, and survey previous work using vascularized organoids to recreate specific organ functions and simulate tumor progression. Vascularization is not only an essential component of individual organ function but also responsible for coupling the fate of all organs and their functions. While some success in coupling two or more organs together on a single platform has been demonstrated, we argue that the future of vascularized organoid technology lies in creating organoid systems complete with tissue-specific microvasculature and in coupling multiple organs through a dynamic vascular network to create systems that can respond to changing physiological conditions.
Collapse
Affiliation(s)
- Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| | - Christopher C W Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, California 95616, USA;
| |
Collapse
|
141
|
Moreira HR, Raftery RM, da Silva LP, Cerqueira MT, Reis RL, Marques AP, O'Brien FJ. In vitro vascularization of tissue engineered constructs by non-viral delivery of pro-angiogenic genes. Biomater Sci 2021; 9:2067-2081. [PMID: 33475111 DOI: 10.1039/d0bm01560a] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vascularization is still one of the major challenges in tissue engineering. In the context of tissue regeneration, the formation of capillary-like structures is often triggered by the addition of growth factors which are associated with high cost, bolus release and short half-life. As an alternative to growth factors, we hypothesized that delivering genes-encoding angiogenic growth factors to cells in a scaffold microenvironment would lead to a controlled release of angiogenic proteins promoting vascularization, simultaneously offering structural support for new matrix deposition. Two non-viral vectors, chitosan (Ch) and polyethyleneimine (PEI), were tested to deliver plasmids encoding for vascular endothelial growth factor (pVEGF) and fibroblast growth factor-2 (pFGF2) to human dermal fibroblasts (hDFbs). hDFbs were successfully transfected with both Ch and PEI, without compromising the metabolic activity. Despite low transfection efficiency, superior VEGF and FGF-2 transgene expression was attained when pVEGF was delivered with PEI and when pFGF2 was delivered with Ch, impacting the formation of capillary-like structures by primary human dermal microvascular endothelial cells (hDMECs). Moreover, in a 3D microenvironment, when PEI-pVEGF and Ch-FGF2 were delivered to hDFbs, cells produced functional pro-angiogenic proteins which induced faster formation of capillary-like structures that were retained in vitro for longer time in a Matrigel assay. The dual combination of the plasmids resulted in a downregulation of the production of VEGF and an upregulation of FGF-2. The number of capillary-like segments obtained with this system was inferior to the delivery of plasmids individually but superior to what was observed with the non-transfected cells. This work confirmed that cell-laden scaffolds containing transfected cells offer a novel, selective and alternative approach to impact the vascularization during tissue regeneration. Moreover, this work provides a new platform for pathophysiology studies, models of disease, culture systems and drug screening.
Collapse
Affiliation(s)
- Helena R Moreira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Avepark, Barco, 4805-017 Guimarães, Portugal
| | | | | | | | | | | | | |
Collapse
|
142
|
Margolis EA, Cleveland DS, Kong YP, Beamish JA, Wang WY, Baker BM, Putnam AJ. Stromal cell identity modulates vascular morphogenesis in a microvasculature-on-a-chip platform. LAB ON A CHIP 2021; 21:1150-1163. [PMID: 33538719 PMCID: PMC7990720 DOI: 10.1039/d0lc01092h] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Supportive stromal cells of mesenchymal origins regulate vascular morphogenesis in developmental, pathological, and regenerative contexts, contributing to vessel formation, maturation, and long-term stability, in part via the secretion of bioactive molecules. In this work, we adapted a microfluidic lab-on-a-chip system that enables the formation and perfusion of microvascular capillary beds with connections to arteriole-scale endothelialized channels to explore how stromal cell (SC) identity influences endothelial cell (EC) morphogenesis. We compared and contrasted lung fibroblasts (LFs), dermal fibroblasts (DFs), and bone marrow-derived mesenchymal stem cells (MSCs) for their abilities to support endothelial morphogenesis and subsequent perfusion of microvascular networks formed in fibrin hydrogels within the microfluidic device. We demonstrated that while all 3 SC types supported EC morphogenesis, LFs in particular resulted in microvascular morphologies with the highest total network length, vessel diameter, and vessel interconnectivity across a range of SC-EC ratio and density conditions. Not only did LFs support robust vascular morphology, but also, they were the only SC type to support functional perfusion of the resultant capillary beds. Lastly, we identified heightened traction stress produced by LFs as a possible mechanism by which LFs enhance endothelial morphogenesis in 3D compared to other SC types examined. This study provides a unique comparison of three different SC types and their role in supporting the formation of microvasculature that could provide insights for the choice of cells for vascular cell-based therapies and the regulation of tissue-specific vasculature.
Collapse
Affiliation(s)
- Emily A Margolis
- Department of Biomedical Engineering, University of Michigan, 1101 Beal Ave., Ann Arbor, MI 48109, USA.
| | | | | | | | | | | | | |
Collapse
|
143
|
Diao JA, Wang JK, Chui WF, Mountain V, Gullapally SC, Srinivasan R, Mitchell RN, Glass B, Hoffman S, Rao SK, Maheshwari C, Lahiri A, Prakash A, McLoughlin R, Kerner JK, Resnick MB, Montalto MC, Khosla A, Wapinski IN, Beck AH, Elliott HL, Taylor-Weiner A. Human-interpretable image features derived from densely mapped cancer pathology slides predict diverse molecular phenotypes. Nat Commun 2021; 12:1613. [PMID: 33712588 PMCID: PMC7955068 DOI: 10.1038/s41467-021-21896-9] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/15/2021] [Indexed: 02/06/2023] Open
Abstract
Computational methods have made substantial progress in improving the accuracy and throughput of pathology workflows for diagnostic, prognostic, and genomic prediction. Still, lack of interpretability remains a significant barrier to clinical integration. We present an approach for predicting clinically-relevant molecular phenotypes from whole-slide histopathology images using human-interpretable image features (HIFs). Our method leverages >1.6 million annotations from board-certified pathologists across >5700 samples to train deep learning models for cell and tissue classification that can exhaustively map whole-slide images at two and four micron-resolution. Cell- and tissue-type model outputs are combined into 607 HIFs that quantify specific and biologically-relevant characteristics across five cancer types. We demonstrate that these HIFs correlate with well-known markers of the tumor microenvironment and can predict diverse molecular signatures (AUROC 0.601-0.864), including expression of four immune checkpoint proteins and homologous recombination deficiency, with performance comparable to 'black-box' methods. Our HIF-based approach provides a comprehensive, quantitative, and interpretable window into the composition and spatial architecture of the tumor microenvironment.
Collapse
Affiliation(s)
- James A Diao
- PathAI, Inc., Boston, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Jason K Wang
- PathAI, Inc., Boston, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | - Wan Fung Chui
- PathAI, Inc., Boston, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Richard N Mitchell
- Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | - Murray B Resnick
- PathAI, Inc., Boston, MA, USA
- Department of Pathology, Warren Alpert Medical School, Providence, RI, USA
| | | | | | | | | | | | | |
Collapse
|
144
|
Teixeira FC, Chaves S, Torres AL, Barrias CC, Bidarra SJ. Engineering a Vascularized 3D Hybrid System to Model Tumor-Stroma Interactions in Breast Cancer. Front Bioeng Biotechnol 2021; 9:647031. [PMID: 33791288 PMCID: PMC8006407 DOI: 10.3389/fbioe.2021.647031] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/16/2021] [Indexed: 01/23/2023] Open
Abstract
The stromal microenvironment of breast tumors, namely the vasculature, has a key role in tumor development and metastatic spread. Tumor angiogenesis is a coordinated process, requiring the cooperation of cancer cells, stromal cells, such as fibroblasts and endothelial cells, secreted factors and the extracellular matrix (ECM). In vitro models capable of capturing such complex environment are still scarce, but are pivotal to improve success rates in drug development and screening. To address this challenge, we developed a hybrid alginate-based 3D system, combining hydrogel-embedded mammary epithelial cells (parenchymal compartment) with a porous scaffold co-seeded with fibroblasts and endothelial cells (vascularized stromal compartment). For the stromal compartment, we used porous alginate scaffolds produced by freeze-drying with particle leaching, a simple, low-cost and non-toxic approach that provided storable ready-to-use scaffolds fitting the wells of standard 96-well plates. Co-seeded endothelial cells and fibroblasts were able to adhere to the surface, spread and organize into tubular-like structures. For the parenchymal compartment, a designed alginate gel precursor solution load with mammary epithelial cells was added to the pores of pre-vascularized scaffolds, forming a hydrogel in situ by ionic crosslinking. The 3D hybrid system supports epithelial morphogenesis in organoids/tumoroids and endothelial tubulogenesis, allowing heterotypic cell-cell and cell-ECM interactions, while presenting excellent experimental tractability for whole-mount confocal microscopy, histology and mild cell recovery for down-stream analysis. It thus provides a unique 3D in vitro platform to dissect epithelial-stromal interactions and tumor angiogenesis, which may assist in the development of selective and more effective anticancer therapies.
Collapse
Affiliation(s)
- Filipa C Teixeira
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Sara Chaves
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Ana Luísa Torres
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Cristina C Barrias
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sílvia J Bidarra
- i3S - Instituto de Inovação e Investigação em Saúde, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
145
|
Yurina NV, Ageeva TA, Goryachkin AM, Varaksin NA, Ryabicheva TG, Ostanin AA, Chernykh ER, Romashchenko AV, Proskurina AS, Bogachev S, Purtov AV. Effects of Recombinant Angiogenin on Collagen Fiber Formation and Angiogenesis in the Dermis of Wistar Rats. Clin Cosmet Investig Dermatol 2021; 14:187-196. [PMID: 33679135 PMCID: PMC7926187 DOI: 10.2147/ccid.s294825] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022]
Abstract
Purpose The purpose of this study was to assess the capability of recombinant angiogenin isolated from Pichia pastoris yeasts to stimulate regenerative processes in the dermis of experimental animals. Patients and Methods Wistar rats were administered with recombinant angiogenin intracutaneously. Morphological examination of the skin and the assessment of the proliferative activity of the epidermal cells were carried out. Additionally, cytokine production by human whole blood cells exposed to angiogenin was analyzed ex vivo. Results Administration of angiogenin stimulates collagen fiber formation and angiogenesis. This stimulation is tightly associated with an increase in the number of fibroblasts, an increased numerical density of dermal blood vessels and an increased density of collagen fibers; also, it activates the proliferation of basal cells. Angiogenin induces the production of MCP, IL-8, IL-6, IL-1β, TNF-α, IL-10, TGF-β, and VEGF by blood cells. Conclusion The results obtained indicate a broad spectrum of actions of recombinant angiogenin during regenerative processes in the basal layer of the dermis.
Collapse
Affiliation(s)
- Natalia V Yurina
- Autonomous Non-Profit Organization "Regional Center for High Medical Technologies", Novosibirsk, Russia
| | | | | | | | | | - Alexandr A Ostanin
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Elena R Chernykh
- Research Institute of Fundamental and Clinical Immunology, Novosibirsk, Russia
| | - Alexander V Romashchenko
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Anastasia S Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | |
Collapse
|
146
|
Pulkkinen HH, Kiema M, Lappalainen JP, Toropainen A, Beter M, Tirronen A, Holappa L, Niskanen H, Kaikkonen MU, Ylä-Herttuala S, Laakkonen JP. BMP6/TAZ-Hippo signaling modulates angiogenesis and endothelial cell response to VEGF. Angiogenesis 2021; 24:129-144. [PMID: 33021694 PMCID: PMC7921060 DOI: 10.1007/s10456-020-09748-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022]
Abstract
The BMP/TGFβ-Smad, Notch and VEGF signaling guides formation of endothelial tip and stalk cells. However, the crosstalk of bone morphogenetic proteins (BMPs) and vascular endothelial growth factor receptor 2 (VEGFR2) signaling has remained largely unknown. We demonstrate that BMP family members regulate VEGFR2 and Notch signaling, and act via TAZ-Hippo signaling pathway. BMPs were found to be regulated after VEGF gene transfer in C57/Bl6 mice and in a porcine myocardial ischemia model. BMPs 2/4/6 were identified as endothelium-specific targets of VEGF. BMP2 modulated VEGF-mediated endothelial sprouting via Delta like Canonical Notch Ligand 4 (DLL4). BMP6 modulated VEGF signaling by regulating VEGFR2 expression and acted via Hippo signaling effector TAZ, known to regulate cell survival/proliferation, and to be dysregulated in cancer. In a matrigel plug assay in nude mice BMP6 was further demonstrated to induce angiogenesis. BMP6 is the first member of BMP family found to directly regulate both Hippo signaling and neovessel formation. It may thus serve as a target in pro/anti-angiogenic therapies.
Collapse
Affiliation(s)
- H H Pulkkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - M Kiema
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - J P Lappalainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Chemistry, University of Eastern Finland and Eastern Finland Laboratory Centre, Kuopio, Finland
| | - A Toropainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - M Beter
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - A Tirronen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - L Holappa
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - H Niskanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - M U Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - S Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Science Service Center, Kuopio University Hospital, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Johanna P Laakkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
147
|
Linares J, Marín-Jiménez JA, Badia-Ramentol J, Calon A. Determinants and Functions of CAFs Secretome During Cancer Progression and Therapy. Front Cell Dev Biol 2021; 8:621070. [PMID: 33553157 PMCID: PMC7862334 DOI: 10.3389/fcell.2020.621070] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple lines of evidence are indicating that cancer development and malignant progression are not exclusively epithelial cancer cell-autonomous processes but may also depend on crosstalk with the surrounding tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) are abundantly represented in the TME and are continuously interacting with cancer cells. CAFs are regulating key mechanisms during progression to metastasis and response to treatment by enhancing cancer cells survival and aggressiveness. The latest advances in CAFs biology are pointing to CAFs-secreted factors as druggable targets and companion tools for cancer diagnosis and prognosis. Especially, extensive research conducted in the recent years has underscored the potential of several cytokines as actionable biomarkers that are currently evaluated in the clinical setting. In this review, we explore the current understanding of CAFs secretome determinants and functions to discuss their clinical implication in oncology.
Collapse
Affiliation(s)
- Jenniffer Linares
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Juan A. Marín-Jiménez
- Department of Medical Oncology, Catalan Institute of Oncology (ICO) - L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jordi Badia-Ramentol
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Alexandre Calon
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| |
Collapse
|
148
|
Lee S, Kang H, Park D, Yu J, Koh SK, Cho D, Kim D, Kang K, Jeon NL. Modeling 3D Human Tumor Lymphatic Vessel Network Using High‐Throughput Platform. Adv Biol (Weinh) 2021. [DOI: 10.1002/adbi.202000195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Somin Lee
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Habin Kang
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Dohyun Park
- Department of Mechanical Engineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - James Yu
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Seung Kwon Koh
- Department of Health Sciences and Technology SAIHST Sungkyunkwan University 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
| | - Duck Cho
- Department of Health Sciences and Technology SAIHST Sungkyunkwan University 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
- Department of Laboratory Medicine and Genetics Samsung Medical Center Sungkyunkwan University School of Medicine 115, Irwon‐ro, Gangnam‐gu Seoul 06355 Republic of Korea
| | - Da‐Hyun Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science College of Veterinary Medicine Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Kyung‐Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science College of Veterinary Medicine Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program for Bioengineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Department of Mechanical Engineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Institute of Advanced Machinery and Design Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
- Institute of BioEngineering Seoul National University 1, Gwanak‐ro, Gwanak‐gu Seoul 08826 Republic of Korea
| |
Collapse
|
149
|
Yue T, Zhao D, Phan DTT, Wang X, Park JJ, Biviji Z, Hughes CCW, Lee AP. A modular microfluidic system based on a multilayered configuration to generate large-scale perfusable microvascular networks. MICROSYSTEMS & NANOENGINEERING 2021; 7:4. [PMID: 33456784 PMCID: PMC7787972 DOI: 10.1038/s41378-020-00229-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/19/2020] [Accepted: 11/17/2020] [Indexed: 05/03/2023]
Abstract
The vascular network of the circulatory system plays a vital role in maintaining homeostasis in the human body. In this paper, a novel modular microfluidic system with a vertical two-layered configuration is developed to generate large-scale perfused microvascular networks in vitro. The two-layer polydimethylsiloxane (PDMS) configuration allows the tissue chambers and medium channels not only to be designed and fabricated independently but also to be aligned and bonded accordingly. This method can produce a modular microfluidic system that has high flexibility and scalability to design an integrated platform with multiple perfused vascularized tissues with high densities. The medium channel was designed with a rhombic shape and fabricated to be semiclosed to form a capillary burst valve in the vertical direction, serving as the interface between the medium channels and tissue chambers. Angiogenesis and anastomosis at the vertical interface were successfully achieved by using different combinations of tissue chambers and medium channels. Various large-scale microvascular networks were generated and quantified in terms of vessel length and density. Minimal leakage of the perfused 70-kDa FITC-dextran confirmed the lumenization of the microvascular networks and the formation of tight vertical interconnections between the microvascular networks and medium channels in different structural layers. This platform enables the culturing of interconnected, large-scale perfused vascularized tissue networks with high density and scalability for a wide range of multiorgan-on-a-chip applications, including basic biological studies and drug screening.
Collapse
Affiliation(s)
- Tao Yue
- Department of Biomedical Engineering, University of California, Irvine, CA USA
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai, China
| | - Da Zhao
- Department of Biomedical Engineering, University of California, Irvine, CA USA
| | - Duc T. T. Phan
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA USA
| | - Xiaolin Wang
- Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, China
- National Key Laboratory of Science and Technology for Micro/Nano Fabrication, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory for Thin Film and Micro Fabrication of the Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Joshua Jonghyun Park
- Department of Electrical Engineering and Computer Science, University of California, Irvine, CA USA
| | - Zayn Biviji
- Department of Applied Mathematics - Biology, Brown University, Providence, RI USA
| | - Christopher C. W. Hughes
- Department of Biomedical Engineering, University of California, Irvine, CA USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA USA
| | - Abraham P. Lee
- Department of Biomedical Engineering, University of California, Irvine, CA USA
- Department of Mechanical and Aerospace Engineering, University of California, Irvine, CA USA
| |
Collapse
|
150
|
Pauty J, Nakano S, Usuba R, Nakajima T, Johmura Y, Omori S, Sakamoto N, Kikuchi A, Nakanishi M, Matsunaga YT. A 3D tissue model-on-a-chip for studying the effects of human senescent fibroblasts on blood vessels. Biomater Sci 2021; 9:199-211. [DOI: 10.1039/d0bm01297a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Senescent cells modify their environment and cause tissue aging that leads to organ dysfunction. Developing strategies for healthy aging rises a need for in vitro models that enables to study senescence and senotherapeutics at a tissue level.
Collapse
Affiliation(s)
- Joris Pauty
- Institute of Industrial Science
- The University of Tokyo
- Tokyo 153-8505
- Japan
| | - Shizuka Nakano
- Institute of Industrial Science
- The University of Tokyo
- Tokyo 153-8505
- Japan
- Department of Materials Science and Technology
| | - Ryo Usuba
- Institute of Industrial Science
- The University of Tokyo
- Tokyo 153-8505
- Japan
| | - Tadaaki Nakajima
- Institute of Industrial Science
- The University of Tokyo
- Tokyo 153-8505
- Japan
| | - Yoshikazu Johmura
- Division of Cancer Cell Biology
- Department of Cancer Biology
- Institute of Medical Science
- The University of Tokyo
- Tokyo 108-8639
| | - Satotaka Omori
- Division of Cancer Cell Biology
- Department of Cancer Biology
- Institute of Medical Science
- The University of Tokyo
- Tokyo 108-8639
| | - Naoya Sakamoto
- Graduate School of Systems Design
- Tokyo Metropolitan University
- Tokyo
- 192-0397
- Japan
| | - Akihiko Kikuchi
- Department of Materials Science and Technology
- Faculty of Industrial Science and Technology
- Tokyo University of Science
- Tokyo 125-8585
- Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology
- Department of Cancer Biology
- Institute of Medical Science
- The University of Tokyo
- Tokyo 108-8639
| | | |
Collapse
|