1
|
Zhao H, Xiong T, Chu Y, Hao W, Zhao T, Sun X, Zhuang Y, Chen B, Zhao Y, Wang J, Chen Y, Dai J. Biomimetic Dual-Network Collagen Fibers with Porous and Mechanical Cues Reconstruct Neural Stem Cell Niche via AKT/YAP Mechanotransduction after Spinal Cord Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311456. [PMID: 38497893 DOI: 10.1002/smll.202311456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/21/2024] [Indexed: 03/19/2024]
Abstract
Tissue engineering scaffolds can mediate the maneuverability of neural stem cell (NSC) niche to influence NSC behavior, such as cell self-renewal, proliferation, and differentiation direction, showing the promising application in spinal cord injury (SCI) repair. Here, dual-network porous collagen fibers (PCFS) are developed as neurogenesis scaffolds by employing biomimetic plasma ammonia oxidase catalysis and conventional amidation cross-linking. Following optimizing the mechanical parameters of PCFS, the well-matched Young's modulus and physiological dynamic adaptability of PCFS (4.0 wt%) have been identified as a neurogenetic exciter after SCI. Remarkably, porous topographies and curving wall-like protrusions are generated on the surface of PCFS by simple and non-toxic CO2 bubble-water replacement. As expected, PCFS with porous and matched mechanical properties can considerably activate the cadherin receptor of NSCs and induce a series of serine-threonine kinase/yes-associated protein mechanotransduction signal pathways, encouraging cellular orientation, neuron differentiation, and adhesion. In SCI rats, implanted PCFS with matched mechanical properties further integrated into the injured spinal cords, inhibited the inflammatory progression and decreased glial and fibrous scar formation. Wall-like protrusions of PCFS drive multiple neuron subtypes formation and even functional neural circuits, suggesting a viable therapeutic strategy for nerve regeneration and functional recovery after SCI.
Collapse
Affiliation(s)
- Haitao Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
| | - Tiandi Xiong
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yun Chu
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Wangping Hao
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
| | - Tongtong Zhao
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Xinyue Sun
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Bing Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences, Beijing, 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences, Beijing, 100101, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, China
| | - Yanyan Chen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
2
|
Joshi IM, Mansouri M, Ahmed A, De Silva D, Simon RA, Esmaili P, Desa DE, Elias TM, Brown EB, Abhyankar VV. Microengineering 3D Collagen Matrices with Tumor-Mimetic Gradients in Fiber Alignment. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2308071. [PMID: 38706986 PMCID: PMC11067715 DOI: 10.1002/adfm.202308071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Indexed: 05/07/2024]
Abstract
Collagen fibers in the 3D tumor microenvironment (TME) exhibit complex alignment landscapes that are critical in directing cell migration through a process called contact guidance. Previous in vitro work studying this phenomenon has focused on quantifying cell responses in uniformly aligned environments. However, the TME also features short-range gradients in fiber alignment that result from cell-induced traction forces. Although the influence of graded biophysical taxis cues is well established, cell responses to physiological alignment gradients remain largely unexplored. In this work, fiber alignment gradients in biopsy samples are characterized and recreated using a new microfluidic biofabrication technique to achieve tunable sub-millimeter to millimeter scale gradients. This study represents the first successful engineering of continuous alignment gradients in soft, natural biomaterials. Migration experiments on graded alignment show that HUVECs exhibit increased directionality, persistence, and speed compared to uniform and unaligned fiber architectures. Similarly, patterned MDA-MB-231 aggregates exhibit biased migration toward increasing fiber alignment, suggesting a role for alignment gradients as a taxis cue. This user-friendly approach, requiring no specialized equipment, is anticipated to offer new insights into the biophysical cues that cells interpret as they traverse the extracellular matrix, with broad applicability in healthy and diseased tissue environments.
Collapse
Affiliation(s)
- Indranil M. Joshi
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | - Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | - Adeel Ahmed
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | - Dinindu De Silva
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | - Richard A. Simon
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | - Poorya Esmaili
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | - Danielle E. Desa
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
| | - Tresa M. Elias
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
| | - Edward B. Brown
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
| | - Vinay V. Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| |
Collapse
|
3
|
Crossley RM, Johnson S, Tsingos E, Bell Z, Berardi M, Botticelli M, Braat QJS, Metzcar J, Ruscone M, Yin Y, Shuttleworth R. Modeling the extracellular matrix in cell migration and morphogenesis: a guide for the curious biologist. Front Cell Dev Biol 2024; 12:1354132. [PMID: 38495620 PMCID: PMC10940354 DOI: 10.3389/fcell.2024.1354132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The extracellular matrix (ECM) is a highly complex structure through which biochemical and mechanical signals are transmitted. In processes of cell migration, the ECM also acts as a scaffold, providing structural support to cells as well as points of potential attachment. Although the ECM is a well-studied structure, its role in many biological processes remains difficult to investigate comprehensively due to its complexity and structural variation within an organism. In tandem with experiments, mathematical models are helpful in refining and testing hypotheses, generating predictions, and exploring conditions outside the scope of experiments. Such models can be combined and calibrated with in vivo and in vitro data to identify critical cell-ECM interactions that drive developmental and homeostatic processes, or the progression of diseases. In this review, we focus on mathematical and computational models of the ECM in processes such as cell migration including cancer metastasis, and in tissue structure and morphogenesis. By highlighting the predictive power of these models, we aim to help bridge the gap between experimental and computational approaches to studying the ECM and to provide guidance on selecting an appropriate model framework to complement corresponding experimental studies.
Collapse
Affiliation(s)
- Rebecca M. Crossley
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Samuel Johnson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Erika Tsingos
- Computational Developmental Biology Group, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Zoe Bell
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Optics11 life, Amsterdam, Netherlands
| | | | - Quirine J. S. Braat
- Department of Applied Physics and Science Education, Eindhoven University of Technology, Eindhoven, Netherlands
| | - John Metzcar
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
- Department of Informatics, Indiana University, Bloomington, IN, United States
| | | | - Yuan Yin
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
4
|
Stepanova D, Byrne HM, Maini PK, Alarcón T. Computational modeling of angiogenesis: The importance of cell rearrangements during vascular growth. WIREs Mech Dis 2024; 16:e1634. [PMID: 38084799 DOI: 10.1002/wsbm.1634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 03/16/2024]
Abstract
Angiogenesis is the process wherein endothelial cells (ECs) form sprouts that elongate from the pre-existing vasculature to create new vascular networks. In addition to its essential role in normal development, angiogenesis plays a vital role in pathologies such as cancer, diabetes and atherosclerosis. Mathematical and computational modeling has contributed to unraveling its complexity. Many existing theoretical models of angiogenic sprouting are based on the "snail-trail" hypothesis. This framework assumes that leading ECs positioned at sprout tips migrate toward low-oxygen regions while other ECs in the sprout passively follow the leaders' trails and proliferate to maintain sprout integrity. However, experimental results indicate that, contrary to the snail-trail assumption, ECs exchange positions within developing vessels, and the elongation of sprouts is primarily driven by directed migration of ECs. The functional role of cell rearrangements remains unclear. This review of the theoretical modeling of angiogenesis is the first to focus on the phenomenon of cell mixing during early sprouting. We start by describing the biological processes that occur during early angiogenesis, such as phenotype specification, cell rearrangements and cell interactions with the microenvironment. Next, we provide an overview of various theoretical approaches that have been employed to model angiogenesis, with particular emphasis on recent in silico models that account for the phenomenon of cell mixing. Finally, we discuss when cell mixing should be incorporated into theoretical models and what essential modeling components such models should include in order to investigate its functional role. This article is categorized under: Cardiovascular Diseases > Computational Models Cancer > Computational Models.
Collapse
Affiliation(s)
- Daria Stepanova
- Laboratorio Subterráneo de Canfranc, Canfranc-Estación, Huesca, Spain
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Philip K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Tomás Alarcón
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
- Centre de Recerca Matemàtica, Bellaterra, Barcelona, Spain
- Departament de Matemàtiques, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
5
|
Schindler D, Moldenhawer T, Beta C, Huisinga W, Holschneider M. Three-component contour dynamics model to simulate and analyze amoeboid cell motility in two dimensions. PLoS One 2024; 19:e0297511. [PMID: 38277351 PMCID: PMC10817190 DOI: 10.1371/journal.pone.0297511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 01/07/2024] [Indexed: 01/28/2024] Open
Abstract
Amoeboid cell motility is relevant in a wide variety of biomedical processes such as wound healing, cancer metastasis, and embryonic morphogenesis. It is characterized by pronounced changes of the cell shape associated with expansions and retractions of the cell membrane, which result in a crawling kind of locomotion. Despite existing computational models of amoeboid motion, the inference of expansion and retraction components of individual cells, the corresponding classification of cells, and the a priori specification of the parameter regime to achieve a specific motility behavior remain challenging open problems. We propose a novel model of the spatio-temporal evolution of two-dimensional cell contours comprising three biophysiologically motivated components: a stochastic term accounting for membrane protrusions and two deterministic terms accounting for membrane retractions by regularizing the shape and area of the contour. Mathematically, these correspond to the intensity of a self-exciting Poisson point process, the area-preserving curve-shortening flow, and an area adjustment flow. The model is used to generate contour data for a variety of qualitatively different, e.g., polarized and non-polarized, cell tracks that visually resemble experimental data very closely. In application to experimental cell tracks, we inferred the protrusion component and examined its correlation to common biomarkers: the F-actin density close to the membrane and its local motion. Due to the low model complexity, parameter estimation is fast, straightforward, and offers a simple way to classify contour dynamics based on two locomotion types: the amoeboid and a so-called fan-shaped type. For both types, we use cell tracks segmented from fluorescence imaging data of the model organism Dictyostelium discoideum. An implementation of the model is provided within the open-source software package AmoePy, a Python-based toolbox for analyzing and simulating amoeboid cell motility.
Collapse
Affiliation(s)
- Daniel Schindler
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
- CRC 1294 Data Assimilation, University of Potsdam, Potsdam, Germany
| | - Ted Moldenhawer
- Institute of Physics and Astronomy, University of Potsdam, Potsdam, Germany
- CRC 1294 Data Assimilation, University of Potsdam, Potsdam, Germany
| | - Carsten Beta
- Institute of Physics and Astronomy, University of Potsdam, Potsdam, Germany
- CRC 1294 Data Assimilation, University of Potsdam, Potsdam, Germany
| | - Wilhelm Huisinga
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
- CRC 1294 Data Assimilation, University of Potsdam, Potsdam, Germany
| | - Matthias Holschneider
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
- CRC 1294 Data Assimilation, University of Potsdam, Potsdam, Germany
| |
Collapse
|
6
|
Kim D, Youn J, Lee J, Kim H, Kim DS. Recent Progress in Fabrication of Electrospun Nanofiber Membranes for Developing Physiological In Vitro Organ/Tissue Models. Macromol Biosci 2023; 23:e2300244. [PMID: 37590903 DOI: 10.1002/mabi.202300244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/13/2023] [Indexed: 08/19/2023]
Abstract
Nanofiber membranes (NFMs), which have an extracellular matrix-mimicking structure and unique physical properties, have garnered great attention as biomimetic materials for developing physiologically relevant in vitro organ/tissue models. Recent progress in NFM fabrication techniques immensely contributes to the development of NFM-based cell culture platforms for constructing physiological organ/tissue models. However, despite the significance of the NFM fabrication technique, an in-depth discussion of the fabrication technique and its future aspect is insufficient. This review provides an overview of the current state-of-the-art of NFM fabrication techniques from electrospinning techniques to postprocessing techniques for the fabrication of various types of NFM-based cell culture platforms. Moreover, the advantages of the NFM-based culture platforms in the construction of organ/tissue models are discussed especially for tissue barrier models, spheroids/organoids, and biomimetic organ/tissue constructs. Finally, the review concludes with perspectives on challenges and future directions for fabrication and utilization of NFMs.
Collapse
Affiliation(s)
- Dohui Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Jaeseung Youn
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Jisang Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Hyeonji Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
- Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk, 37673, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50, Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
7
|
Terragni F, Martinson WD, Carretero M, Maini PK, Bonilla LL. Soliton approximation in continuum models of leader-follower behavior. Phys Rev E 2023; 108:054407. [PMID: 38115402 DOI: 10.1103/physreve.108.054407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/17/2023] [Indexed: 12/21/2023]
Abstract
Complex biological processes involve collective behavior of entities (bacteria, cells, animals) over many length and time scales and can be described by discrete models that track individuals or by continuum models involving densities and fields. We consider hybrid stochastic agent-based models of branching morphogenesis and angiogenesis (new blood vessel creation from preexisting vasculature), which treat cells as individuals that are guided by underlying continuous chemical and/or mechanical fields. In these descriptions, leader (tip) cells emerge from existing branches and follower (stalk) cells build the new sprout in their wake. Vessel branching and fusion (anastomosis) occur as a result of tip and stalk cell dynamics. Coarse graining these hybrid models in appropriate limits produces continuum partial differential equations (PDEs) for endothelial cell densities that are more analytically tractable. While these models differ in nonlinearity, they produce similar equations at leading order when chemotaxis is dominant. We analyze this leading order system in a simple quasi-one-dimensional geometry and show that the numerical solution of the leading order PDE is well described by a soliton wave that evolves from vessel to source. This wave is an attractor for intermediate times until it arrives at the hypoxic region releasing the growth factor. The mathematical techniques used here thus identify common features of discrete and continuum approaches and provide insight into general biological mechanisms governing their collective dynamics.
Collapse
Affiliation(s)
- F Terragni
- Gregorio Millán Institute for Fluid Dynamics, Nanoscience and Industrial Mathematics, Universidad Carlos III de Madrid, 28911 Leganés, Spain
- Department of Mathematics, Universidad Carlos III de Madrid, 28911 Leganés, Spain
| | - W D Martinson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford OX2 6GG, United Kingdom
| | - M Carretero
- Gregorio Millán Institute for Fluid Dynamics, Nanoscience and Industrial Mathematics, Universidad Carlos III de Madrid, 28911 Leganés, Spain
- Department of Mathematics, Universidad Carlos III de Madrid, 28911 Leganés, Spain
| | - P K Maini
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford OX2 6GG, United Kingdom
| | - L L Bonilla
- Gregorio Millán Institute for Fluid Dynamics, Nanoscience and Industrial Mathematics, Universidad Carlos III de Madrid, 28911 Leganés, Spain
- Department of Mathematics, Universidad Carlos III de Madrid, 28911 Leganés, Spain
| |
Collapse
|
8
|
Wistner SC, Rashad L, Slaughter G. Advances in tissue engineering and biofabrication for in vitro skin modeling. BIOPRINTING (AMSTERDAM, NETHERLANDS) 2023; 35:e00306. [PMID: 38645432 PMCID: PMC11031264 DOI: 10.1016/j.bprint.2023.e00306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The global prevalence of skin disease and injury is continually increasing, yet conventional cell-based models used to study these conditions do not accurately reflect the complexity of human skin. The lack of inadequate in vitro modeling has resulted in reliance on animal-based models to test pharmaceuticals, biomedical devices, and industrial and environmental toxins to address clinical needs. These in vivo models are monetarily and morally expensive and are poor predictors of human tissue responses and clinical trial outcomes. The onset of three-dimensional (3D) culture techniques, such as cell-embedded and decellularized approaches, has offered accessible in vitro alternatives, using innovative scaffolds to improve cell-based models' structural and histological authenticity. However, these models lack adequate organizational control and complexity, resulting in variations between structures and the exclusion of physiologically relevant vascular and immunological features. Recently, biofabrication strategies, which combine biology, engineering, and manufacturing capabilities, have emerged as instrumental tools to recreate the heterogeneity of human skin precisely. Bioprinting uses computer-aided design (CAD) to yield robust and reproducible skin prototypes with unprecedented control over tissue design and assembly. As the interdisciplinary nature of biofabrication grows, we look to the promise of next-generation biofabrication technologies, such as organ-on-a-chip (OOAC) and 4D modeling, to simulate human tissue behaviors more reliably for research, pharmaceutical, and regenerative medicine purposes. This review aims to discuss the barriers to developing clinically relevant skin models, describe the evolution of skin-inspired in vitro structures, analyze the current approaches to biofabricating 3D human skin mimetics, and define the opportunities and challenges in biofabricating skin tissue for preclinical and clinical uses.
Collapse
Affiliation(s)
- Sarah C. Wistner
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Layla Rashad
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Gymama Slaughter
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, 23508, USA
| |
Collapse
|
9
|
Jorgensen AM, Gorkun A, Mahajan N, Willson K, Clouse C, Jeong CG, Varkey M, Wu M, Walker SJ, Molnar JA, Murphy SV, Lee SJ, Yoo JJ, Soker S, Atala A. Multicellular bioprinted skin facilitates human-like skin architecture in vivo. Sci Transl Med 2023; 15:eadf7547. [PMID: 37792956 DOI: 10.1126/scitranslmed.adf7547] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 09/15/2023] [Indexed: 10/06/2023]
Abstract
Bioprinting is a promising alternative method to generate skin substitutes because it can replicate the structural organization of the skin into biomimetic layers in vitro. In this study, six primary human skin cell types were used to bioprint a trilayer skin construct consisting of epidermis, dermis, and hypodermis. Transplantation of the bioprinted skin with human cells onto full-thickness wounds of nu/nu mice promoted rapid vascularization and formation of epidermal rete ridges analogous to the native human epidermis, with a normal-looking extracellular matrix. Cell-specific staining confirmed the integration of the implanted cells into the regenerated skin. Using a similar approach, a 5 centimeter-by-5 centimeter bioprinted autologous porcine skin graft was transplanted onto full-thickness wounds in a porcine excisional wound model. The bioprinted skin graft improved epithelialization, reduced skin contraction, and supported normal collagen organization with reduced fibrosis. Differential gene expression demonstrated pro-remodeling protease activity in wounds transplanted with bioprinted autologous skin grafts. These results demonstrate that bioprinted skin can support skin regeneration to allow for nonfibrotic wound healing and suggest that the skin bioprinting technology may be applicable for human clinical use.
Collapse
Affiliation(s)
- Adam M Jorgensen
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Anastasiya Gorkun
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Naresh Mahajan
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Kelsey Willson
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Cara Clouse
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Claire G Jeong
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Mathew Varkey
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Mingsong Wu
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Joseph A Molnar
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
- Department of Plastic and Reconstructive Surgery, Atrium Health Wake Forest Baptist Hospital, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| |
Collapse
|
10
|
LaBelle SA, Poulson AM, Maas SA, Rauff A, Ateshian GA, Weiss JA. Spatial Configurations of 3D Extracellular Matrix Collagen Density and Anisotropy Simultaneously Guide Angiogenesis. PLoS Comput Biol 2023; 19:e1011553. [PMID: 37871113 PMCID: PMC10621972 DOI: 10.1371/journal.pcbi.1011553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 11/02/2023] [Accepted: 09/29/2023] [Indexed: 10/25/2023] Open
Abstract
Extracellular matrix (ECM) collagen density and fibril anisotropy are thought to affect the development of new vasculatures during pathologic and homeostatic angiogenesis. Computational simulation is emerging as a tool to investigate the role of matrix structural configurations on cell guidance. However, prior computational models have only considered the orientation of collagen as a model input. Recent experimental evidence indicates that cell guidance is simultaneously influenced by the direction and intensity of alignment (i.e., degree of anisotropy) as well as the local collagen density. The objective of this study was to explore the role of ECM collagen anisotropy and density during sprouting angiogenesis through simulation in the AngioFE and FEBio modeling frameworks. AngioFE is a plugin for FEBio (Finite Elements for Biomechanics) that simulates cell-matrix interactions during sprouting angiogenesis. We extended AngioFE to represent ECM collagen as deformable 3D ellipsoidal fibril distributions (EFDs). The rate and direction of microvessel growth were modified to depend simultaneously on the ECM collagen anisotropy (orientation and degree of anisotropy) and density. The sensitivity of growing neovessels to these stimuli was adjusted so that AngioFE could reproduce the growth and guidance observed in experiments where microvessels were cultured in collagen gels of varying anisotropy and density. We then compared outcomes from simulations using EFDs to simulations that used AngioFE's prior vector field representation of collagen anisotropy. We found that EFD simulations were more accurate than vector field simulations in predicting experimentally observed microvessel guidance. Predictive simulations demonstrated the ability of anisotropy gradients to recruit microvessels across short and long distances relevant to wound healing. Further, simulations predicted that collagen alignment could enable microvessels to overcome dense tissue interfaces such as tumor-associated collagen structures (TACS) found in desmoplasia and tumor-stroma interfaces. This approach can be generalized to other mechanobiological relationships during cell guidance phenomena in computational settings.
Collapse
Affiliation(s)
- Steven A. LaBelle
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - A. Marsh Poulson
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Steve A. Maas
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Adam Rauff
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Gerard A. Ateshian
- Department of Mechanical Engineering, Columbia University, New York, New York, United States of America
| | - Jeffrey A. Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
11
|
Ristori T, Thuret R, Hooker E, Quicke P, Lanthier K, Ntumba K, Aspalter IM, Uroz M, Herbert SP, Chen CS, Larrivée B, Bentley K. Bmp9 regulates Notch signaling and the temporal dynamics of angiogenesis via Lunatic Fringe. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.25.557123. [PMID: 37808725 PMCID: PMC10557600 DOI: 10.1101/2023.09.25.557123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
In brief The mechanisms regulating the signaling pathways involved in angiogenesis are not fully known. Ristori et al. show that Lunatic Fringe (LFng) mediates the crosstalk between Bone Morphogenic Protein 9 (Bmp9) and Notch signaling, thereby regulating the endothelial cell behavior and temporal dynamics of their identity during sprouting angiogenesis. Highlights Bmp9 upregulates the expression of LFng in endothelial cells.LFng regulates the temporal dynamics of tip/stalk selection and rearrangement.LFng indicated to play a role in hereditary hemorrhagic telangiectasia.Bmp9 and LFng mediate the endothelial cell-pericyte crosstalk.Bone Morphogenic Protein 9 (Bmp9), whose signaling through Activin receptor-like kinase 1 (Alk1) is involved in several diseases, has been shown to independently activate Notch target genes in an additive fashion with canonical Notch signaling. Here, by integrating predictive computational modeling validated with experiments, we uncover that Bmp9 upregulates Lunatic Fringe (LFng) in endothelial cells (ECs), and thereby also regulates Notch activity in an inter-dependent, multiplicative fashion. Specifically, the Bmp9-upregulated LFng enhances Notch receptor activity creating a much stronger effect when Dll4 ligands are also present. During sprouting, this LFng regulation alters vessel branching by modulating the timing of EC phenotype selection and rearrangement. Our results further indicate that LFng can play a role in Bmp9-related diseases and in pericyte-driven vessel stabilization, since we find LFng contributes to Jag1 upregulation in Bmp9-stimulated ECs; thus, Bmp9-upregulated LFng results in not only enhanced EC Dll4-Notch1 activation, but also Jag1-Notch3 activation in pericytes.
Collapse
|
12
|
Matveeva DK, Ezdakova MI, Ratushnyy AY. Modification of the Properties of Extracellular Matrix of Senescent Mesenchymal Stem Cells. Bull Exp Biol Med 2023; 175:569-575. [PMID: 37770790 DOI: 10.1007/s10517-023-05905-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Indexed: 09/30/2023]
Abstract
Cell senescence leads to changes in the secretory activity of mesenchymal stem cells (MSC), including proteins of extracellular matrix (ECM). Here we studied the regulatory properties of ECM of senescent MSC in a model with endothelial cells (EC). EC were seeded onto a decellularized extracellular matrix of senescent MSC. Changes in cell morphology and a decrease in cell growth were observed. In addition, increased production of inflammatory chemokines MCP-1 and GROα and reduced synthesis of proangiogenic growth factor FGF-2 were revealed. Analysis of ECM showed quantitative and qualitative changes, including fibronectin layer morphology, total protein content, and concentration of deposited growth factors such as VEGF. Thus, our work demonstrates that senescence of MSC can lead to modification of the effects of their ECM on EC activity.
Collapse
Affiliation(s)
- D K Matveeva
- Institute for Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - M I Ezdakova
- Institute for Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - A Yu Ratushnyy
- Institute for Biomedical Problems, Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
13
|
Joshi IM, Mansouri M, Ahmed A, Simon RA, Bambizi PE, Desa DE, Elias TM, Brown EB, Abhyankar VV. Microengineering 3D Collagen Matrices with Tumor-Mimetic Gradients in Fiber Alignment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.09.548253. [PMID: 37502844 PMCID: PMC10369918 DOI: 10.1101/2023.07.09.548253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
In the tumor microenvironment (TME), collagen fibers facilitate tumor cell migration through the extracellular matrix. Previous studies have focused on studying the responses of cells on uniformly aligned or randomly aligned collagen fibers. However, the in vivo environment also features spatial gradients in alignment, which arise from the local reorganization of the matrix architecture due to cell-induced traction forces. Although there has been extensive research on how cells respond to graded biophysical cues, such as stiffness, porosity, and ligand density, the cellular responses to physiological fiber alignment gradients have been largely unexplored. This is due, in part, to a lack of robust experimental techniques to create controlled alignment gradients in natural materials. In this study, we image tumor biopsy samples and characterize the alignment gradients present in the TME. To replicate physiological gradients, we introduce a first-of-its-kind biofabrication technique that utilizes a microfluidic channel with constricting and expanding geometry to engineer 3D collagen hydrogels with tunable fiber alignment gradients that range from sub-millimeter to millimeter length scales. Our modular approach allows easy access to the microengineered gradient gels, and we demonstrate that HUVECs migrate in response to the fiber architecture. We provide preliminary evidence suggesting that MDA-MB-231 cell aggregates, patterned onto a specific location on the alignment gradient, exhibit preferential migration towards increasing alignment. This finding suggests that alignment gradients could serve as an additional taxis cue in the ECM. Importantly, our study represents the first successful engineering of continuous gradients of fiber alignment in soft, natural materials. We anticipate that our user-friendly platform, which needs no specialized equipment, will offer new experimental capabilities to study the impact of fiber-based contact guidance on directed cell migration.
Collapse
Affiliation(s)
- Indranil M. Joshi
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | - Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | - Adeel Ahmed
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | - Richard A. Simon
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| | | | - Danielle E. Desa
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
| | - Tresa M. Elias
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
| | - Edward B. Brown
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
| | - Vinay V. Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY
| |
Collapse
|
14
|
Everts PA, Lana JF, Onishi K, Buford D, Peng J, Mahmood A, Fonseca LF, van Zundert A, Podesta L. Angiogenesis and Tissue Repair Depend on Platelet Dosing and Bioformulation Strategies Following Orthobiological Platelet-Rich Plasma Procedures: A Narrative Review. Biomedicines 2023; 11:1922. [PMID: 37509560 PMCID: PMC10377284 DOI: 10.3390/biomedicines11071922] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Angiogenesis is the formation of new blood vessel from existing vessels and is a critical first step in tissue repair following chronic disturbances in healing and degenerative tissues. Chronic pathoanatomic tissues are characterized by a high number of inflammatory cells; an overexpression of inflammatory mediators; such as tumor necrosis factor-α (TNF-α) and interleukin-1 (IL-1); the presence of mast cells, T cells, reactive oxygen species, and matrix metalloproteinases; and a decreased angiogenic capacity. Multiple studies have demonstrated that autologous orthobiological cellular preparations (e.g., platelet-rich plasma (PRP)) improve tissue repair and regenerate tissues. There are many PRP devices on the market. Unfortunately, they differ greatly in platelet numbers, cellular composition, and bioformulation. PRP is a platelet concentrate consisting of a high concentration of platelets, with or without certain leukocytes, platelet-derived growth factors (PGFs), cytokines, molecules, and signaling cells. Several PRP products have immunomodulatory capacities that can influence resident cells in a diseased microenvironment, inducing tissue repair or regeneration. Generally, PRP is a blood-derived product, regardless of its platelet number and bioformulation, and the literature indicates both positive and negative patient treatment outcomes. Strangely, the literature does not designate specific PRP preparation qualifications that can potentially contribute to tissue repair. Moreover, the literature scarcely addresses the impact of platelets and leukocytes in PRP on (neo)angiogenesis, other than a general one-size-fits-all statement that "PRP has angiogenic capabilities". Here, we review the cellular composition of all PRP constituents, including leukocytes, and describe the importance of platelet dosing and bioformulation strategies in orthobiological applications to initiate angiogenic pathways that re-establish microvasculature networks, facilitating the supply of oxygen and nutrients to impaired tissues.
Collapse
Affiliation(s)
- Peter A Everts
- Research & Education Division, Gulf Coast Biologics, Fort Myers, FL 33916, USA
- OrthoRegen Group, Max-Planck University, Indaiatuba, São Paulo 13334-170, Brazil
| | - José Fábio Lana
- OrthoRegen Group, Max-Planck University, Indaiatuba, São Paulo 13334-170, Brazil
- Department of Orthopaedics, The Bone and Cartilage Institute, Indaiatuba, São Paulo 13334-170, Brazil
| | - Kentaro Onishi
- Department of PM&R and Orthopedic Surgery, University of Pittsburg Medical Center, Pittsburgh, PA 15213, USA
| | - Don Buford
- Texas Orthobiologics, Dallas, TX 75204, USA
| | - Jeffrey Peng
- Stanford Health Care-O'Connor Hospital Sports Medicine, Stanford University School of Medicine, San Jose, CA 95128, USA
| | - Ansar Mahmood
- Department of Trauma and Orthopaedic Surgery, University Hospitals, Birmingham B15 2GW, UK
| | - Lucas F Fonseca
- Department of Orthopaedics, The Federal University of São Paulo, São Paulo 04024-002, Brazil
| | - Andre van Zundert
- Department of Anaesthesia and Perioperative Medicine, Royal Brisbane and Women's Hospital, Brisbane and the University of Queensland, Brisbane 4072, Australia
| | - Luga Podesta
- Bluetail Medical Group & Podesta Orthopedic Sports Medicine, Naples, FL 34109, USA
| |
Collapse
|
15
|
Cai Y, Zhang F, Feng J, Wu B, Li H, Xiao S, Lu F, Wei Z, Deng C. Long-term follow-up and exploration of the mechanism of stromal vascular fraction gel in chronic wounds. Stem Cell Res Ther 2023; 14:163. [PMID: 37337292 DOI: 10.1186/s13287-023-03389-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 05/26/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Chronic refractory wounds easily relapse and seriously affect the patients' quality of life. Previous studies have shown that stromal vascular fraction gel (SVF-gel) significantly promotes the early healing of chronic wounds; however, the mechanisms of SVF-gel function per se remain unclear, and a long-term follow-up is lacking. This study aims to explore the mechanisms of SVF-gel promoting the healing of chronic wounds and follow up the long-term efficacy of SVF-gel. METHODS Autologous SVF-gel transplantation was performed in 20 patients with chronic wounds (from March 2016 to September 2019), and the size of the wound before and after SVF-gel transplantation was observed. The conditioned medium (CM) was harvested from SVF-gel under serum-free, serum-deprivation and 10% fetal bovine serum (FBS) microenvironment in vitro, respectively. The concentration of the growth factors in the two kinds of gel-CM was tested, and their effects on the proliferation and migration of human dermal fibroblasts (HDFs) were detected. RESULTS All patients had 100% wound closure eventually, and the average time to complete closure was 28.3 ± 9.7 days. The time of follow-up ranged from 2 to 6 years, and there was no wound recurrence. Interestingly, the concentrations of epidermal growth factor and transforming growth factor β1 of the CM were higher in serum-free and serum-deprivation condition than in 10% FBS microenvironment (p < 0.05). Correspondingly, the proliferation and migration ability of HDFs treated with gel-CM from serum-free condition were stronger than those treated with gel-CM from serum-deprivation (2% FBS) or 10% FBS microenvironment (p < 0.05). CONCLUSION These results indicate that it is safe, effective, and lasting in effect to treat chronic wounds with SVF-gel and mechanisms of action that include secreting various cytokines and promoting cell proliferation and migration ability. TRIAL REGISTRATION Chinese Clinical Trail Registry, ChiCTR2000034624. Registered 12 July 2020-Retrospectively registered, http://www.chictr.org.cn/showproj.aspx?proj=56058.
Collapse
Affiliation(s)
- Yuan Cai
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, 563000, Guizhou, China
| | - Fang Zhang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, 563000, Guizhou, China
| | - Jingwei Feng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bihua Wu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, 563000, Guizhou, China
| | - Hai Li
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, 563000, Guizhou, China
| | - Shune Xiao
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, 563000, Guizhou, China
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zairong Wei
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, 563000, Guizhou, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China.
| | - Chengliang Deng
- Department of Burns and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Zunyi, 563000, Guizhou, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
16
|
Everts PA, Panero AJ. Basic Science of Autologous Orthobiologics. Phys Med Rehabil Clin N Am 2023; 34:25-47. [DOI: 10.1016/j.pmr.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
17
|
Mohammadi M, Soltani M, Aghanajafi C, Kohandel M. Investigation of the evolution of tumor-induced microvascular network under the inhibitory effect of anti-angiogenic factor, angiostatin: A mathematical study. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:5448-5480. [PMID: 36896553 DOI: 10.3934/mbe.2023252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Anti-angiogenesis as a treatment strategy for normalizing the microvascular network of tumors is of great interest among researchers, especially in combination with chemotherapy or radiotherapy. According to the vital role that angiogenesis plays in tumor growth and in exposing the tumor to therapeutic agents, this work develops a mathematical framework to study the influence of angiostatin, a plasminogen fragment that shows the anti-angiogenic function, in the evolutionary behavior of tumor-induced angiogenesis. Angiostatin-induced microvascular network reformation is investigated in a two-dimensional space by considering two parent vessels around a circular tumor by a modified discrete angiogenesis model in different tumor sizes. The effects of imposing modifications on the existing model, i.e., the matrix-degrading enzyme effect, proliferation and death of endothelial cells, matrix density function, and a more realistic chemotactic function, are investigated in this study. Results show a decrease in microvascular density in response to the angiostatin. A functional relationship exists between angiostatin's ability to normalize the capillary network and tumor size or progression stage, such that capillary density decreases by 55%, 41%, 24%, and 13% in tumors with a non-dimensional radius of 0.4, 0.3, 0.2, and 0.1, respectively, after angiostatin administration.
Collapse
Affiliation(s)
- Mahya Mohammadi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19919-43344, Iran
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19919-43344, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- School of Optometry and Vision Science, Faculty of Science, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Advanced Bioengineering Initiative Center, Multidisciplinary International Complex, K. N. Toosi University of Technology, Tehran 19697-64499, Iran
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON N2L 3G1, Canada
| | - Cyrus Aghanajafi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran 19919-43344, Iran
| | - Mohammad Kohandel
- Department of Applied Mathematics, University of Waterloo, Waterloo, ON N2L 3G1, Canada
| |
Collapse
|
18
|
Ghasemi Nasab MS, Niroomand-Oscuii H, Bazmara H, Soltani M. Multi-scale model of lumen formation via inverse membrane blebbing mechanism during sprouting angiogenesis process. J Theor Biol 2023; 556:111312. [PMID: 36279960 DOI: 10.1016/j.jtbi.2022.111312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/04/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
Abstract
Cancer is one of the leading causes of mortality and morbidity among people worldwide. Cancer appears as solid tumors in many cases. Angiogenesis is the growth of blood vessels from the existing vasculature and is one of the imperative processes in tumor growth. Another vital phenomenon for formation and functionality of this vasculature network is lumen formation. The results of recent studies indicate the importance of blood pressure in this mechanism. Computational modeling can study these processes in different scales. Hence, wide varieties of these models have been proposed during recent years. In this research, a multi-scale model is developed for the angiogenesis process. In the extracellular scale, the growth factor concentration is calculated via the reaction diffusion equation. At the cellular scale, growth, migration, and the adhesion of endothelial cells are modeled by the Potts cellular model. At the intra-cellular scale by considering biochemical signals, a Boolean network model describes migration, division, or apoptosis of endothelial cells. A stochastic model developed for lumen formation via inverse membrane blebbing mechanism. A CFD simulation was also used to investigate the role of pulsated blood pressure in the inverse membrane blebbing mechanism. The lumen formation model shows stochastic behavior in blebs expansion and lumen expansion. Comparing the stochastic model's results with the CFD simulation also shows the vital role of pressure pulse and the topology of the blebs in bleb retraction.
Collapse
Affiliation(s)
| | | | | | - Majid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran; Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran; Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
19
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
20
|
Abdalrahman T, Checa S. On the role of mechanical signals on sprouting angiogenesis through computer modeling approaches. Biomech Model Mechanobiol 2022; 21:1623-1640. [PMID: 36394779 PMCID: PMC9700567 DOI: 10.1007/s10237-022-01648-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 10/08/2022] [Indexed: 11/19/2022]
Abstract
Sprouting angiogenesis, the formation of new vessels from preexisting vasculature, is an essential process in the regeneration of new tissues as well as in the development of some diseases like cancer. Although early studies identified chemical signaling as the main driver of this process, many recent studies have shown a strong role of mechanical signals in the formation of new capillaries. Different types of mechanical signals (e.g., external forces, cell traction forces, and blood flow-induced shear forces) have been shown to play distinct roles in the process; however, their interplay remains still largely unknown. During the last decades, mathematical and computational modeling approaches have been developed to investigate and better understand the mechanisms behind mechanically driven angiogenesis. In this manuscript, we review computational models of angiogenesis with a focus on models investigating the role of mechanics on the process. Our aim is not to provide a detailed review on model methodology but to describe what we have learnt from these models. We classify models according to the mechanical signals being investigated and describe how models have looked into their role on the angiogenic process. We show that a better understanding of the mechanobiology of the angiogenic process will require the development of computer models that incorporate the interactions between the multiple mechanical signals and their effect on cellular responses, since they all seem to play a key in sprout patterning. In the end, we describe some of the remaining challenges of computational modeling of angiogenesis and discuss potential avenues for future research.
Collapse
|
21
|
Jia G, Yang H, Wang K, Huang D, Chen W, Shan Y. The modeling study of the effect of morphological behaviors of extracellular matrix fibers on the dynamic interaction between tumor cells and antitumor immune response. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2022; 38:e3633. [PMID: 35703086 DOI: 10.1002/cnm.3633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 04/28/2022] [Accepted: 06/10/2022] [Indexed: 06/15/2023]
Abstract
Low response rate limits the effective application of immunotherapy, in which the interactions between tumor cells and immune cells play a significant role. The strength of regulation could be mediated by extracellular matrix (ECM) fibers, which is still insufficiently investigated. In the study, the cellular potts model was utilized to explore the role of morphological properties of ECM in tumor-immune interactions. It was observed that high-density random ECM fibers delayed the interaction between tumor cells and T cells. Moreover, the tumor-immune interactions were ECM morphology-specific. Radial ECM fibers exhibited weaker inhibitory role in the process of contact between tumor cells and T cells. This study provided the useful mechanism of tumor-immune interactions from the viewpoint of morphological effect of ECM fibers, facilitating improving the efficiency of immunotherapy.
Collapse
Affiliation(s)
- Guanjie Jia
- Department of Biomedical Engineering, Research Center for Nano-Biomaterials and Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Hao Yang
- Department of Biomedical Engineering, Research Center for Nano-Biomaterials and Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Kaiqun Wang
- Department of Biomedical Engineering, Research Center for Nano-Biomaterials and Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-Biomaterials and Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Weiyi Chen
- Department of Biomedical Engineering, Research Center for Nano-Biomaterials and Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Yanhu Shan
- School of Instrument and Electronics, North University of China, Taiyuan, China
| |
Collapse
|
22
|
Lana JFSD, Lana AVSD, da Fonseca LF, Coelho MA, Marques GG, Mosaner T, Ribeiro LL, Azzini GOM, Santos GS, Fonseca E, de Andrade MAP. Stromal Vascular Fraction for Knee Osteoarthritis - An Update. J Stem Cells Regen Med 2022; 18:11-20. [PMID: 36003656 DOI: 10.46582/jsrm.1801003] [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: 05/15/2021] [Accepted: 12/10/2021] [Indexed: 11/19/2022]
Abstract
Orthobiologics never cease to cause popularity within the medical science field, distinctly in regenerative medicine. Recently, adipose tissue has been an object of interest for many researchers and medical experts due to the fact that it represents a novel and potential cell source for tissue engineering and regenerative medicine purposes. Stromal vascular fraction (SVF), for instance, which is an adipose tissue-derivative, has generated optimistic results in many scenarios. Its biological potential can be harnessed and administered into injured tissues, particularly areas in which standard healing is disrupted. This is a typical feature of osteoarthritis (OA), a common degenerative joint disease which is outlined by persistent inflammation and destruction of surrounding tissues. SVF is known to carry a large amount of stem and progenitor cells, which are able to perform self-renewal, differentiation, and proliferation. Furthermore, they also secrete several cytokines and several growth factors, effectively sustaining immune modulatory effects and halting the escalated pro-inflammatory status of OA. Although SVF has shown interesting results throughout the medical community, additional research is still highly desirable in order to further elucidate its potential regarding musculoskeletal disorders, especially OA.
Collapse
Affiliation(s)
| | | | - Lucas Furtado da Fonseca
- Orthopaedic Department - Universidade Federal de São Paulo - Escola Paulista de Medicina, São Paulo - SP, Brazil
| | - Marcelo Amaral Coelho
- IOC - Instituto do Osso e da Cartilagem / The Bone and Cartilage Institute, Indaiatuba - SP, Brazil
| | | | - Tomas Mosaner
- IOC - Instituto do Osso e da Cartilagem / The Bone and Cartilage Institute, Indaiatuba - SP, Brazil
| | | | | | - Gabriel Silva Santos
- IOC - Instituto do Osso e da Cartilagem / The Bone and Cartilage Institute, Indaiatuba - SP, Brazil
| | - Eduardo Fonseca
- IOC - Instituto do Osso e da Cartilagem / The Bone and Cartilage Institute, Indaiatuba - SP, Brazil
| | | |
Collapse
|
23
|
Dong G, Wu H, Hu J, Teng L. Stromal Vascular Fraction Promotes Viability of Co-grafted Axial Skin Flaps in Rats Model. Aesthetic Plast Surg 2022; 46:1950-1963. [PMID: 35794244 DOI: 10.1007/s00266-022-02812-y] [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: 11/25/2021] [Accepted: 01/27/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Stromal vascular fraction (SVF) has been proved in promoting the vascularization of fascial flap through cell differentiation and paracrine effect and can be autologous transplanted without culture after isolation in vitro. We intend to establish a novel co-grafted flap model of rats to investigate the efficacy and mechanism of SVF on flaps and skinsin facilitating angiogenesis and immune regulation. METHOD 60 female Sprague Dawley rats were divided into the SVF group and the control group. A pedicled fascial flap combined with a free skin model was established, and 4×106 CM-DIl labeled SVF cells were transplanted into the fascia flap; the rats were executed on days 1, 2, 3, 7, 10 postoperatively (n = 6). Flow cytometry was carried out to determine the cell proportion and surface marker of SVFs. The therapeutic effects of SVF were evaluated via Doppler blood perfusion imager, flap survival rates, histology, immunohistochemistry and immunofluorescence. The bioinformatic mechanism analysis was achieved by high-throughput RNAseq of mRNA and LncRNA. RESULT Flow cytometry confirmed SVF contains heterogeneous cellular composition, especially hematopoietic cells. Doppler blood perfusion imager showed SVF significantly improved flap survival with higher blood perfusion and survival rates. Immunohistochemistry of CD31 displayed higher level of angiogenesis in SVF-treated group, and CM-DIL-labeled SVF cells could survive and participate in revascularization, and RNA sequencing results revealed SVF promoted wound healing by facilitating intercellular adhesion, cell migration and positive immune response. CONCLUSION SVF could reduce skin flap necrosis and activated neovascularization in rats by facilitating intercellular adhesion, cell migration and regulate positive immune response. LEVEL OF EVIDENCE N/A This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.
Collapse
Affiliation(s)
- Guoxuan Dong
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Badachu Road, Shijingshan District, No. 33, Beijing, 100144, China
| | - Huanhuan Wu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Badachu Road, Shijingshan District, No. 33, Beijing, 100144, China.
| | - JunLong Hu
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Badachu Road, Shijingshan District, No. 33, Beijing, 100144, China
| | - Li Teng
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Badachu Road, Shijingshan District, No. 33, Beijing, 100144, China.
| |
Collapse
|
24
|
Monteiro NO, Oliveira C, Silva TH, Martins A, Fangueiro JF, Reis RL, Neves NM. Biomimetic Surface Topography from the Rubus fruticosus Leaf as a Guidance of Angiogenesis in Tissue Engineering Applications. ACS Biomater Sci Eng 2022; 8:2943-2953. [PMID: 35706335 DOI: 10.1021/acsbiomaterials.2c00264] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The promotion of angiogenesis is a fundamental step for efficient organ/tissue reconstitution and replacement. Thus, several strategies to promote vascularization of scaffolds were studied to satisfy this unsolved clinical need. The interface between cells and substrates is a determinant for the success of tissue engineering (TE) strategies. Substrate's topography is reported to play a key role in influencing endothelial cell behavior, namely, on its proliferation, metabolic activity, morphology, migration, and secretion of cytokines and chemokines. Therefore, surface topography of the biomaterial-based grafts is a crucial property that is considered in the development of a new TE approach. Herein, we hypothesize that the surface of Rubus fruticosus leaf plays a crucial role in driving angiogenesis since its architecture resembles the vascular structures at a biologically relevant size scale. For this, we produced biomimetic polycaprolactone (PCL) membranes (BpMs) replicating the surface topography of a R. fruticosus leaf by replica molding and nanoimprint lithography. Our results showed an enhanced performance in terms of proliferation of the human endothelial cell line on top of the BpM. Moreover, an asymmetric cellular spatial distribution among the surface of the BpM was observed. These cells seem to have higher density for longer time periods in the region that replicates the leaf veins. Finally, we assess the angiogenic capacity through a chick chorioallantoic membrane assay, revealing that BpMs are more prone to support angiogenesis than flat PCL membranes. We strongly believe that this strategy can bring new insights into developing TE strategies with an enhanced performance in terms of the vascular integration between the host and the scaffolds implanted.
Collapse
Affiliation(s)
- Nelson O Monteiro
- 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal.,ICVS/3B's─PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Catarina Oliveira
- 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal.,ICVS/3B's─PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Tiago H Silva
- 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal.,ICVS/3B's─PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Albino Martins
- 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal.,ICVS/3B's─PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana F Fangueiro
- 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal.,ICVS/3B's─PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal.,ICVS/3B's─PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno M Neves
- 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães 4805-017, Portugal.,ICVS/3B's─PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
25
|
He X, Lee B, Jiang Y. Extracellular matrix in cancer progression and therapy. MEDICAL REVIEW (2021) 2022; 2:125-139. [PMID: 37724245 PMCID: PMC10471113 DOI: 10.1515/mr-2021-0028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/31/2022] [Indexed: 09/20/2023]
Abstract
The tumor ecosystem with heterogeneous cellular compositions and the tumor microenvironment has increasingly become the focus of cancer research in recent years. The extracellular matrix (ECM), the major component of the tumor microenvironment, and its interactions with the tumor cells and stromal cells have also enjoyed tremendously increased attention. Like the other components of the tumor microenvironment, the ECM in solid tumors differs significantly from that in normal organs and tissues. We review recent studies of the complex roles the tumor ECM plays in cancer progression, from tumor initiation, growth to angiogenesis and invasion. We highlight that the biomolecular, biophysical, and mechanochemical interactions between the ECM and cells not only regulate the steps of cancer progression, but also affect the efficacy of systemic cancer treatment. We further discuss the strategies to target and modify the tumor ECM to improve cancer therapy.
Collapse
Affiliation(s)
- Xiuxiu He
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Byoungkoo Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
26
|
Jafari Nivlouei S, Soltani M, Shirani E, Salimpour MR, Travasso R, Carvalho J. A multiscale cell-based model of tumor growth for chemotherapy assessment and tumor-targeted therapy through a 3D computational approach. Cell Prolif 2022; 55:e13187. [PMID: 35132721 PMCID: PMC8891571 DOI: 10.1111/cpr.13187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/09/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES Computational modeling of biological systems is a powerful tool to clarify diverse processes contributing to cancer. The aim is to clarify the complex biochemical and mechanical interactions between cells, the relevance of intracellular signaling pathways in tumor progression and related events to the cancer treatments, which are largely ignored in previous studies. MATERIALS AND METHODS A three-dimensional multiscale cell-based model is developed, covering multiple time and spatial scales, including intracellular, cellular, and extracellular processes. The model generates a realistic representation of the processes involved from an implementation of the signaling transduction network. RESULTS Considering a benign tumor development, results are in good agreement with the experimental ones, which identify three different phases in tumor growth. Simulating tumor vascular growth, results predict a highly vascularized tumor morphology in a lobulated form, a consequence of cells' motile behavior. A novel systematic study of chemotherapy intervention, in combination with targeted therapy, is presented to address the capability of the model to evaluate typical clinical protocols. The model also performs a dose comparison study in order to optimize treatment efficacy and surveys the effect of chemotherapy initiation delays and different regimens. CONCLUSIONS Results not only provide detailed insights into tumor progression, but also support suggestions for clinical implementation. This is a major step toward the goal of predicting the effects of not only traditional chemotherapy but also tumor-targeted therapies.
Collapse
Affiliation(s)
- Sahar Jafari Nivlouei
- Department of Mechanical Engineering, Isfahan University of Technology, Isafahan, Iran.,Department of Physics, CFisUC, University of Coimbra, Coimbra, Portugal
| | - Madjid Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.,Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada.,Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada.,Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran.,Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Shirani
- Department of Mechanical Engineering, Isfahan University of Technology, Isafahan, Iran.,Department of Mechanical Engineering, Foolad Institute of Technology, Fooladshahr, Iran
| | | | - Rui Travasso
- Department of Physics, CFisUC, University of Coimbra, Coimbra, Portugal
| | - João Carvalho
- Department of Physics, CFisUC, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
27
|
Soltani M. Capillary network formation and structure in a modified discrete mathematical model of angiogenesis. Biomed Phys Eng Express 2021; 8. [PMID: 34883475 DOI: 10.1088/2057-1976/ac4175] [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: 06/08/2021] [Accepted: 12/09/2021] [Indexed: 01/01/2023]
Abstract
Angiogenesis, as part of cancer development, involves hierarchical complicated events and processes. Multiple studies have revealed the significance of the formation and structure of tumor-induced capillary networks. In this study, a discrete mathematical model of angiogenesis is studied and modified to capture the realistic physics of capillary network formation. Modifications are performed on the mathematical foundations of an existing discrete model of angiogenesis. The main modifications are the imposition of the matrix density effect, implementation of realistic boundary and initial conditions, and improvement of the method of governing equations based on physical observation. Results show that endothelial cells accelerate angiogenesis and capillary formation as they migrate toward the tumor and clearly exhibit the physical concept of haptotactic movement. On the other hand, consideration of blood flow-induced stress leads to a dynamic adaptive vascular network of capillaries which intelligibly reflects the brush border effect . The present modified model of capillary network formation is based on the physical rationale that defines a clear mathematical and physical interpretation of angiogenesis, which is likely to be used in cancer development modeling and anti-angiogenic therapies.
Collapse
Affiliation(s)
- M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.,Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Ontario, Canada.,Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Ontario, Canada.,Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Tehran Province, Iran
| |
Collapse
|
28
|
Wang C, Li S, Ademiloye AS, Nithiarasu P. Biomechanics of cells and subcellular components: A comprehensive review of computational models and applications. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2021; 37:e3520. [PMID: 34390323 DOI: 10.1002/cnm.3520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 08/10/2021] [Indexed: 06/13/2023]
Abstract
Cells are a fundamental structural, functional and biological unit for all living organisms. Up till now, considerable efforts have been made to study the responses of single cells and subcellular components to an external load, and understand the biophysics underlying cell rheology, mechanotransduction and cell functions using experimental and in silico approaches. In the last decade, computational simulation has become increasingly attractive due to its critical role in interpreting experimental data, analysing complex cellular/subcellular structures, facilitating diagnostic designs and therapeutic techniques, and developing biomimetic materials. Despite the significant progress, developing comprehensive and accurate models of living cells remains a grand challenge in the 21st century. To understand current state of the art, this review summarises and classifies the vast array of computational biomechanical models for cells. The article covers the cellular components at multi-spatial levels, that is, protein polymers, subcellular components, whole cells and the systems with scale beyond a cell. In addition to the comprehensive review of the topic, this article also provides new insights into the future prospects of developing integrated, active and high-fidelity cell models that are multiscale, multi-physics and multi-disciplinary in nature. This review will be beneficial for the researchers in modelling the biomechanics of subcellular components, cells and multiple cell systems and understanding the cell functions and biological processes from the perspective of cell mechanics.
Collapse
Affiliation(s)
- Chengyuan Wang
- Zienkiewicz Centre for Computational Engineering, Faculty of Science and Engineering, Swansea University, Bay Campus, Swansea, UK
| | - Si Li
- Zienkiewicz Centre for Computational Engineering, Faculty of Science and Engineering, Swansea University, Bay Campus, Swansea, UK
| | - Adesola S Ademiloye
- Zienkiewicz Centre for Computational Engineering, Faculty of Science and Engineering, Swansea University, Bay Campus, Swansea, UK
| | - Perumal Nithiarasu
- Zienkiewicz Centre for Computational Engineering, Faculty of Science and Engineering, Swansea University, Bay Campus, Swansea, UK
| |
Collapse
|
29
|
Ebrahim N, Badr OAM, Yousef MM, Hassouna A, Sabry D, Farid AS, Mostafa O, Saihati HAA, Seleem Y, Abd El Aziz E, Khalil AH, Nawar A, Shoulah AA, Aljasir M, Mohamed AZ, El-Sherbiny M, Elsherbiny NM, Eladl MA, Forsyth NR, Salim RF. Functional Recellularization of Acellular Rat Liver Scaffold by Induced Pluripotent Stem Cells: Molecular Evidence for Wnt/B-Catenin Upregulation. Cells 2021; 10:cells10112819. [PMID: 34831042 PMCID: PMC8616374 DOI: 10.3390/cells10112819] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Liver transplantation remains the only viable therapy for liver failure but has a severely restricted utility. Here, we aimed to decellularize rat livers to form acellular 3D bio-scaffolds suitable for seeding with induced pluripotent cells (iPSCs) as a tool to investigate the role of Wnt/β-catenin signaling in liver development and generation. METHODS Dissected rat livers were randomly divided into three groups: I (control); II (decellularized scaffolds) and III (recellularized scaffolds). Liver decellularization was established via an adapted perfusion procedure and assessed through the measurement of extracellular matrix (ECM) proteins and DNA content. Liver recellularization was assessed through histological examination and measurement of transcript levels of Wnt/β-catenin pathway, hepatogenesis, liver-specific microRNAs and growth factors essential for liver development. Adult rat liver decellularization was confirmed by the maintenance of ECM proteins and persistence of growth factors essential for liver regeneration. RESULTS iPSCs seeded rat decellularized livers displayed upregulated transcript expression of Wnt/β-catenin pathway-related, growth factors, and liver specification genes. Further, recellularized livers displayed restored liver-specific functions including albumin secretion and urea synthesis. CONCLUSION This establishes proof-of-principle for the generation of three-dimensional liver organ scaffolds as grafts and functional re-establishment.
Collapse
Affiliation(s)
- Nesrine Ebrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (N.E.); (M.M.Y.); (O.M.)
- Stem Cell Unit, Faculty of Medicine, Benha University, Banha 13511, Egypt
| | - Omnia A. M. Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Banha 13511, Egypt;
| | - Mohamed M. Yousef
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (N.E.); (M.M.Y.); (O.M.)
| | - Amira Hassouna
- School of Public Health and Interdisciplinary Studies, Faculty of Health and Environmental Sciences, AUT University, Auckland 1010, New Zealand;
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo 12613, Egypt;
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Bader University in Cairo, Cairo 11562, Egypt
| | - Ayman Samir Farid
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Banha 13511, Egypt;
| | - Ola Mostafa
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (N.E.); (M.M.Y.); (O.M.)
| | - Hajir A. Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Hafar Al Batin 39524, Saudi Arabia;
| | - Yasmin Seleem
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (Y.S.); (E.A.E.A.)
| | - Eman Abd El Aziz
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Banha 13511, Egypt; (Y.S.); (E.A.E.A.)
| | - Ahmed Hassan Khalil
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, Benha University, Banha 13511, Egypt;
| | - Ahmed Nawar
- Department of General Surgery, Faculty of Medicine, Benha University, Banha 13511, Egypt; (A.N.); (A.A.S.)
| | - Ahmed A. Shoulah
- Department of General Surgery, Faculty of Medicine, Benha University, Banha 13511, Egypt; (A.N.); (A.A.S.)
| | - Mohammad Aljasir
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia;
| | - Amira Zaki Mohamed
- Department of Microbiology, Faculty of Science, Tanta University, Tanta 31527, Egypt;
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 71666, Saudi Arabia;
- Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Nehal M. Elsherbiny
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 47512, Saudi Arabia
- Correspondence: or (N.M.E.); (M.A.E.); (R.F.S.)
| | - Mohamed Ahmed Eladl
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- Correspondence: or (N.M.E.); (M.A.E.); (R.F.S.)
| | - Nicholas Robert Forsyth
- Guy Hilton Research Laboratories, School of Pharmacy and Bioengineering, Faculty of Medicine and Health Sciences, Keele University, Newcastle ST5 5BG, UK;
| | - Rabab F. Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Banha 13511, Egypt
- Correspondence: or (N.M.E.); (M.A.E.); (R.F.S.)
| |
Collapse
|
30
|
Li X, You R, Zhang Q, Yan S, Luo Z, Qu J, Li M. Engineering vascularized dermal grafts by integrating a biomimetic scaffold and Wharton's jelly MSC-derived endothelial cells. J Mater Chem B 2021; 9:6466-6479. [PMID: 34364307 DOI: 10.1039/d1tb00857a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tissue engineering aims to generate functional tissue constructs with the necessary scaffold properties for cell colonization and the establishment of a vascular network. However, treatment of tissue defects using synthetic scaffolds remains a challenge mainly due to insufficient and slow vascularization. Our previous study developed a macroporous silk fibroin scaffold with a nanofibrous microstructure, and demonstrated that the nanofibrous structure can promote the viability of endothelial cells (ECs) and guide cell migration. Further studies are needed to clarify the effect of scaffold microstructures on cell-mediated vascularization. Here, we investigated the efficacy of EC-seeded nanofibrous scaffolds in improving vascularization in vivo. ECs derived from induced human Wharton's Jelly mesenchymal stem cells served as a potential source for cell transplantation. The cell-seeded scaffolds were implanted into dermal defects of SD rats, demonstrating that the multiscale hierarchical design significantly improved the capacity of transplanted cells to promote and accelerate neovascularization and dermal reconstruction via enhancing cell infiltration, collagen deposition and growth factor expression. Our findings provide new insight into the development of degradable macroporous composite materials with 3D microstructures as tissue engineering scaffolds with enhanced vascularization functions, and also provide new treatment options for cell transplantation.
Collapse
Affiliation(s)
- Xiufang Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China.
| | | | | | | | | | | | | |
Collapse
|
31
|
Fazal F, Raghav S, Callanan A, Koutsos V, Radacsi N. Recent advancements in the bioprinting of vascular grafts. Biofabrication 2021; 13. [PMID: 34102613 DOI: 10.1088/1758-5090/ac0963] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023]
Abstract
Recent advancements in the bioinks and three-dimensional (3D) bioprinting methods used to fabricate vascular constructs are summarized herein. Critical biomechanical properties required to fabricate an ideal vascular graft are highlighted, as well as various testing methods have been outlined to evaluate the bio-fabricated grafts as per the Food and Drug Administration (FDA) and International Organization for Standardization (ISO) guidelines. Occlusive artery disease and cardiovascular disease are the major causes of death globally. These diseases are caused by the blockage in the arteries, which results in a decreased blood flow to the tissues of major organs in the body, such as the heart. Bypass surgery is often performed using a vascular graft to re-route the blood flow. Autologous grafts represent a gold standard for such bypass surgeries; however, these grafts may be unavailable due to the previous harvesting or possess a poor quality. Synthetic grafts serve well for medium to large-sized vessels, but they fail when used to replace small-diameter vessels, generally smaller than 6 mm. Various tissue engineering approaches have been used to address the urgent need for vascular graft that can withstand hemodynamic blood pressure and has the ability to grow and remodel. Among these approaches, 3D bioprinting offers an attractive solution to construct patient-specific vessel grafts with layered biomimetic structures.
Collapse
Affiliation(s)
- Faraz Fazal
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, EH9 3FB Edinburgh, United Kingdom.,Department of Mechanical Engineering, University of Engineering and Technology, Lahore, (New Campus) Pakistan
| | - Sakshika Raghav
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, EH9 3FB Edinburgh, United Kingdom
| | - Anthony Callanan
- School of Engineering, Institute for Bioengineering, The University of Edinburgh, The King's Buildings, EH9 3JL Edinburgh, United Kingdom
| | - Vasileios Koutsos
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, EH9 3FB Edinburgh, United Kingdom
| | - Norbert Radacsi
- School of Engineering, Institute for Materials and Processes, The University of Edinburgh, Robert Stevenson Road, EH9 3FB Edinburgh, United Kingdom
| |
Collapse
|
32
|
Jafari Nivlouei S, Soltani M, Carvalho J, Travasso R, Salimpour MR, Shirani E. Multiscale modeling of tumor growth and angiogenesis: Evaluation of tumor-targeted therapy. PLoS Comput Biol 2021; 17:e1009081. [PMID: 34161319 PMCID: PMC8259971 DOI: 10.1371/journal.pcbi.1009081] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/06/2021] [Accepted: 05/14/2021] [Indexed: 12/12/2022] Open
Abstract
The dynamics of tumor growth and associated events cover multiple time and spatial scales, generally including extracellular, cellular and intracellular modifications. The main goal of this study is to model the biological and physical behavior of tumor evolution in presence of normal healthy tissue, considering a variety of events involved in the process. These include hyper and hypoactivation of signaling pathways during tumor growth, vessels' growth, intratumoral vascularization and competition of cancer cells with healthy host tissue. The work addresses two distinctive phases in tumor development-the avascular and vascular phases-and in each stage two cases are considered-with and without normal healthy cells. The tumor growth rate increases considerably as closed vessel loops (anastomoses) form around the tumor cells resulting from tumor induced vascularization. When taking into account the host tissue around the tumor, the results show that competition between normal cells and cancer cells leads to the formation of a hypoxic tumor core within a relatively short period of time. Moreover, a dense intratumoral vascular network is formed throughout the entire lesion as a sign of a high malignancy grade, which is consistent with reported experimental data for several types of solid carcinomas. In comparison with other mathematical models of tumor development, in this work we introduce a multiscale simulation that models the cellular interactions and cell behavior as a consequence of the activation of oncogenes and deactivation of gene signaling pathways within each cell. Simulating a therapy that blocks relevant signaling pathways results in the prevention of further tumor growth and leads to an expressive decrease in its size (82% in the simulation).
Collapse
Affiliation(s)
- Sahar Jafari Nivlouei
- Department of Mechanical Engineering, Isfahan University of Technology, Isafahan, Iran
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal
| | - M. Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Ontario, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Ontario, Canada
- Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - João Carvalho
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Rui Travasso
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal
| | | | - Ebrahim Shirani
- Department of Mechanical Engineering, Isfahan University of Technology, Isafahan, Iran
- Department of Mechanical Engineering, Foolad Institute of Technology, Fooladshahr, Iran
| |
Collapse
|
33
|
Espina JA, Marchant CL, Barriga EH. Durotaxis: the mechanical control of directed cell migration. FEBS J 2021; 289:2736-2754. [PMID: 33811732 PMCID: PMC9292038 DOI: 10.1111/febs.15862] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/23/2021] [Accepted: 04/01/2021] [Indexed: 11/28/2022]
Abstract
Directed cell migration is essential for cells to efficiently migrate in physiological and pathological processes. While migrating in their native environment, cells interact with multiple types of cues, such as mechanical and chemical signals. The role of chemical guidance via chemotaxis has been studied in the past, the understanding of mechanical guidance of cell migration via durotaxis remained unclear until very recently. Nonetheless, durotaxis has become a topic of intensive research and several advances have been made in the study of mechanically guided cell migration across multiple fields. Thus, in this article we provide a state of the art about durotaxis by discussing in silico, in vitro and in vivo data. We also present insights on the general mechanisms by which cells sense, transduce and respond to environmental mechanics, to then contextualize these mechanisms in the process of durotaxis and explain how cells bias their migration in anisotropic substrates. Furthermore, we discuss what is known about durotaxis in vivo and we comment on how haptotaxis could arise from integrating durotaxis and chemotaxis in native environments.
Collapse
Affiliation(s)
- Jaime A Espina
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), Oeiras, Portugal
| | - Cristian L Marchant
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), Oeiras, Portugal
| | - Elias H Barriga
- Mechanisms of Morphogenesis Lab, Gulbenkian Institute of Science (IGC), Oeiras, Portugal
| |
Collapse
|
34
|
Robinson AJ, Pérez-Nava A, Ali SC, González-Campos JB, Holloway JL, Cosgriff-Hernandez EM. Comparative Analysis of Fiber Alignment Methods in Electrospinning. MATTER 2021; 4:821-844. [PMID: 35757372 PMCID: PMC9222234 DOI: 10.1016/j.matt.2020.12.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Fabrication of anisotropic materials is highly desirable in designing biomaterials and tissue engineered constructs. Electrospinning has been broadly adopted due to its versatility in producing non-woven fibrous meshes with tunable fiber diameters (from 10 nanometers to 10 microns), microarchitectures, and construct geometries. A myriad of approaches have been utilized to control fiber alignment of electrospun materials to achieve complex microarchitectures, improve mechanical properties, and provide topographical cellular cues. This review provides a comparative analysis of the techniques developed to generate fiber alignment in electrospun materials. A description of the underlying mechanisms that drive fiber alignment, setup variations for each technique, and the resulting impact on the aligned microarchitecture is provided. A critical analysis of the advantages and limitations of each approach is provided to guide researchers in method selection. Finally, future perspectives of advanced electrospinning methodologies are discussed in terms of developing a scalable method with precise control of microarchitecture.
Collapse
Affiliation(s)
- Andrew J. Robinson
- Department of Biomedical Engineering, University of Texas, Austin, Texas, 78712, United States
| | - Alejandra Pérez-Nava
- Biological and Chemical Research Institute, Universidad Michoacana de San Nicolás, de Hidalgo, Morelia, 58030, Mexico
| | - Shan C. Ali
- Department of Biomedical Engineering, University of Texas, Austin, Texas, 78712, United States
| | - J. Betzabe González-Campos
- Biological and Chemical Research Institute, Universidad Michoacana de San Nicolás, de Hidalgo, Morelia, 58030, Mexico
| | - Julianne L. Holloway
- Chemical Engineering, School for Engineering of Matter, Transport and Energy,Arizona State University, Tempe, 85287, Arizona, United States
| | | |
Collapse
|
35
|
Edgar LT, Franco CA, Gerhardt H, Bernabeu MO. On the preservation of vessel bifurcations during flow-mediated angiogenic remodelling. PLoS Comput Biol 2021; 17:e1007715. [PMID: 33539345 PMCID: PMC7909651 DOI: 10.1371/journal.pcbi.1007715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/26/2021] [Accepted: 11/26/2020] [Indexed: 11/18/2022] Open
Abstract
During developmental angiogenesis, endothelial cells respond to shear stress by migrating and remodelling the initially hyperbranched plexus, removing certain vessels whilst maintaining others. In this study, we argue that the key regulator of vessel preservation is cell decision behaviour at bifurcations. At flow-convergent bifurcations where migration paths diverge, cells must finely tune migration along both possible paths if the bifurcation is to persist. Experiments have demonstrated that disrupting the cells’ ability to sense shear or the junction forces transmitted between cells impacts the preservation of bifurcations during the remodelling process. However, how these migratory cues integrate during cell decision making remains poorly understood. Therefore, we present the first agent-based model of endothelial cell flow-mediated migration suitable for interrogating the mechanisms behind bifurcation stability. The model simulates flow in a bifurcated vessel network composed of agents representing endothelial cells arranged into a lumen which migrate against flow. Upon approaching a bifurcation where more than one migration path exists, agents refer to a stochastic bifurcation rule which models the decision cells make as a combination of flow-based and collective-based migratory cues. With this rule, cells favour branches with relatively larger shear stress or cell number. We found that cells must integrate both cues nearly equally to maximise bifurcation stability. In simulations with stable bifurcations, we found competitive oscillations between flow and collective cues, and simulations that lost the bifurcation were unable to maintain these oscillations. The competition between these two cues is haemodynamic in origin, and demonstrates that a natural defence against bifurcation loss during remodelling exists: as vessel lumens narrow due to cell efflux, resistance to flow and shear stress increases, attracting new cells to enter and rescue the vessel from regression. Our work provides theoretical insight into the role of junction force transmission has in stabilising vasculature during remodelling and as an emergent mechanism to avoid functional shunting. When new blood vessels are created, the endothelial cells that make up these vessels migrate and rearrange in response to blood flow to remodel and optimise the vessel network. An essential part of this process is maintaining the branched structure of the network; however, it is unclear what cues cells consider at regions where vessels branch (i.e., bifurcations). In this research, we present a computer model of cell migration to interrogate the process of preserving bifurcations during remodelling. In this model, cells at bifurcations are influenced by both flow and force transmitted from neighbouring cells. We found that both cues (flow-based and collective-based) must be considered equally in order to preserve branching in the vessel network. In simulations with stable bifurcations, we demonstrated that these cues oscillate: a strong signal in one was accompanied by a weak signal in the other. Furthermore, we found that these cues naturally compete with each other due to the coupling between blood flow and the size of the blood vessels, i.e. larger vessels with more cells produce less flow signals and vice versa. Our research provides insight into how forces transmitted between neighbouring cells stabilise and preserve branching during remodelling, as well as implicates the disruption of this force transmission as a potential mechanism when remodelling goes wrong as in the case of vascular malformation.
Collapse
Affiliation(s)
- Lowell T. Edgar
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
- * E-mail: (LTE); (MOB)
| | - Claudio A. Franco
- Instituto de Medicina Molecular—João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Miguel O. Bernabeu
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
- * E-mail: (LTE); (MOB)
| |
Collapse
|
36
|
Dhavalikar P, Robinson A, Lan Z, Jenkins D, Chwatko M, Salhadar K, Jose A, Kar R, Shoga E, Kannapiran A, Cosgriff-Hernandez E. Review of Integrin-Targeting Biomaterials in Tissue Engineering. Adv Healthc Mater 2020; 9:e2000795. [PMID: 32940020 PMCID: PMC7960574 DOI: 10.1002/adhm.202000795] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/27/2020] [Indexed: 12/12/2022]
Abstract
The ability to direct cell behavior has been central to the success of numerous therapeutics to regenerate tissue or facilitate device integration. Biomaterial scientists are challenged to understand and modulate the interactions of biomaterials with biological systems in order to achieve effective tissue repair. One key area of research investigates the use of extracellular matrix-derived ligands to target specific integrin interactions and induce cellular responses, such as increased cell migration, proliferation, and differentiation of mesenchymal stem cells. These integrin-targeting proteins and peptides have been implemented in a variety of different polymeric scaffolds and devices to enhance tissue regeneration and integration. This review first presents an overview of integrin-mediated cellular processes that have been identified in angiogenesis, wound healing, and bone regeneration. Then, research utilizing biomaterials are highlighted with integrin-targeting motifs as a means to direct these cellular processes to enhance tissue regeneration. In addition to providing improved materials for tissue repair and device integration, these innovative biomaterials provide new tools to probe the complex processes of tissue remodeling in order to enhance the rational design of biomaterial scaffolds and guide tissue regeneration strategies.
Collapse
Affiliation(s)
- Prachi Dhavalikar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrew Robinson
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Dana Jenkins
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Malgorzata Chwatko
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Karim Salhadar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Anupriya Jose
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ronit Kar
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Erik Shoga
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Aparajith Kannapiran
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | | |
Collapse
|
37
|
All Roads Lead to Directional Cell Migration. Trends Cell Biol 2020; 30:852-868. [PMID: 32873438 DOI: 10.1016/j.tcb.2020.08.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/30/2020] [Accepted: 08/04/2020] [Indexed: 01/17/2023]
Abstract
Directional cell migration normally relies on a variety of external signals, such as chemical, mechanical, or electrical, which instruct cells in which direction to move. Many of the major molecular and physical effects derived from these cues are now understood, leading to questions about whether directional cell migration is alike or distinct under these different signals, and how cells might be directed by multiple simultaneous cues, which would be expected in complex in vivo environments. In this review, we compare how different stimuli are spatially distributed, often as gradients, to direct cell movement and the mechanisms by which they steer cells. A comparison of the downstream effectors of directional cues suggests that different external signals regulate a common set of components: small GTPases and the actin cytoskeleton, which implies that the mechanisms downstream of different signals are likely to be closely related and underlies the idea that cell migration operates by a common set of physical principles, irrespective of the input.
Collapse
|
38
|
Hanumantharao SN, Que CA, Vogl BJ, Rao S. Engineered Three-Dimensional Scaffolds Modulating Fate of Breast Cancer Cells Using Stiffness and Morphology Related Cell Adhesion. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2020; 1:41-48. [PMID: 35402960 PMCID: PMC8979620 DOI: 10.1109/ojemb.2020.2965084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 11/25/2022] Open
Abstract
Goal: Artificially engineering the tumor microenvironment in vitro as a vital tool for understanding the mechanism of tumor progression. In this study, we developed three-dimensional cell scaffold systems with different topographical features and mechanical properties but similar surface chemistry. The cell behavior was modulated by the topography and mechanical properties of the scaffold. Methods: Adenocarcinoma (MCF7), triple-negative (MDA-MB-231) and premalignant (MCF10AneoT) breast cancer cells were seeded on the scaffold systems. The cell viability, cell-cell interaction and cell-matrix interactions were analyzed. The preferential growth and alignment of specific population of cells were demonstrated. Results: Among the different scaffolds, triple-negative breast cancer cells preferred honeycomb scaffolds while adenocarcinoma cells favored mesh scaffolds and premalignant cells preferred the aligned scaffolds. Conclusions: The 3D model system developed here can be used to support growth of only specific cell populations or for the growth of tumors. This model can be used for understanding the topographical and mechanical features affecting tumorigenesis, cancer cell growth and migration behavior of malignant and metastatic cancer cells.
Collapse
Affiliation(s)
| | | | | | - Smitha Rao
- Department of Biomedical EngineeringMichigan Technological UniversityHoughtonMI49931USA
| |
Collapse
|
39
|
Notch signaling and taxis mechanisms regulate early stage angiogenesis: A mathematical and computational model. PLoS Comput Biol 2020; 16:e1006919. [PMID: 31986145 PMCID: PMC7021322 DOI: 10.1371/journal.pcbi.1006919] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 02/14/2020] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
Abstract
During angiogenesis, new blood vessels sprout and grow from existing ones. This process plays a crucial role in organ development and repair, in wound healing and in numerous pathological processes such as cancer progression or diabetes. Here, we present a mathematical model of early stage angiogenesis that permits exploration of the relative importance of mechanical, chemical and cellular cues. Endothelial cells proliferate and move over an extracellular matrix by following external gradients of Vessel Endothelial Growth Factor, adhesion and stiffness, which are incorporated to a Cellular Potts model with a finite element description of elasticity. The dynamics of Notch signaling involving Delta-4 and Jagged-1 ligands determines tip cell selection and vessel branching. Through their production rates, competing Jagged-Notch and Delta-Notch dynamics determine the influence of lateral inhibition and lateral induction on the selection of cellular phenotypes, branching of blood vessels, anastomosis (fusion of blood vessels) and angiogenesis velocity. Anastomosis may be favored or impeded depending on the mechanical configuration of strain vectors in the ECM near tip cells. Numerical simulations demonstrate that increasing Jagged production results in pathological vasculatures with thinner and more abundant vessels, which can be compensated by augmenting the production of Delta ligands. Angiogenesis is the process by which new blood vessels grow from existing ones. This process plays a crucial role in organ development, in wound healing and in numerous pathological processes such as cancer growth or in diabetes. Angiogenesis is a complex, multi-step and well regulated process where biochemistry and physics are intertwined. The process entails signaling in vessel cells being driven by both chemical and mechanical mechanisms that result in vascular cell movement, deformation and proliferation. Mathematical models have the ability to bring together these mechanisms in order to explore their relative relevance in vessel growth. Here, we present a mathematical model of early stage angiogenesis that is able to explore the role of biochemical signaling and tissue mechanics. We use this model to unravel the regulating role of Jagged, Notch and Delta dynamics in vascular cells. These membrane proteins have an important part in determining the leading cell in each neo-vascular sprout. Numerical simulations demonstrate that increasing Jagged production results in pathological vasculatures with thinner and more abundant vessels, which can be compensated by augmenting the production of Delta ligands.
Collapse
|
40
|
Flegg JA, Menon SN, Byrne HM, McElwain DLS. A Current Perspective on Wound Healing and Tumour-Induced Angiogenesis. Bull Math Biol 2020; 82:23. [DOI: 10.1007/s11538-020-00696-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 01/02/2020] [Indexed: 12/19/2022]
|
41
|
Kristen M, Ainsworth MJ. Fiber Scaffold Patterning for Mending Hearts: 3D Organization Bringing the Next Step. Adv Healthc Mater 2020; 9:e1900775. [PMID: 31603288 PMCID: PMC7116178 DOI: 10.1002/adhm.201900775] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/14/2019] [Indexed: 12/14/2022]
Abstract
Heart failure (HF) is a leading cause of death worldwide. The most common conditions that lead to HF are coronary artery disease, myocardial infarction, valve disorders, high blood pressure, and cardiomyopathy. Due to the limited regenerative capacity of the heart, the only curative therapy currently available is heart transplantation. Therefore, there is a great need for the development of novel regenerative strategies to repair the injured myocardium, replace damaged valves, and treat occluded coronary arteries. Recent advances in manufacturing technologies have resulted in the precise fabrication of 3D fiber scaffolds with high architectural control that can support and guide new tissue growth, opening exciting new avenues for repair of the human heart. This review discusses the recent advancements in the novel research field of fiber patterning manufacturing technologies for cardiac tissue engineering (cTE) and to what extent these technologies could meet the requirements of the highly organized and structured cardiac tissues. Additionally, future directions of these novel fiber patterning technologies, designs, and applicability to advance cTE are presented.
Collapse
Affiliation(s)
- Marleen Kristen
- Regenerative Medicine Center, University Medical Center Utrecht,
Utrecht 3584 CT, The Netherlands; Department of Orthopedics, University Medical
Center Utrecht, Utrecht 3584 CX, The Netherlands
| | - Madison J. Ainsworth
- Regenerative Medicine Center, University Medical Center Utrecht,
Utrecht 3584 CT, The Netherlands; Department of Orthopedics, University Medical
Center Utrecht, Utrecht 3584 CX, The Netherlands
| |
Collapse
|
42
|
Stefanis AJ, Groh T, Arenbergerova M, Arenberger P, Bauer PO. Stromal Vascular Fraction and its Role in the Management of Alopecia: A Review. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2019; 12:35-44. [PMID: 32038756 PMCID: PMC6937163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Adipose cells organized in small clusters under the reticular dermis closely interact with hair follicular cells and regulate the hair cycle. Intradermal adipocyte progenitor cells are activated toward the end of the telogen phase to proliferate and differentiate into mature adipocytes. These cells, surrounding the hair follicles, secrete signaling molecules that control the progression of the hair cycle. Diseases associated with defects in adipocyte homeostasis, such as lipodystrophy and focal dermal hypoplasia, lead to alopecia. In this review, we discuss the potential influence of stromal vascular fraction from adipose tissue in the management of alopecia as well as its involvement in preclinical and clinical trials.
Collapse
Affiliation(s)
- Athanasios J Stefanis
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| | - Tomas Groh
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| | - Monika Arenbergerova
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| | - Petr Arenberger
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| | - Peter O Bauer
- Drs. Stefanis, Arenbergerova, and Arenberger are with the Department of Dermatology and Venereology, Faculty Hospital Kralovske Vinohrady and Third Medical Faculty of Charles University in Prague, the Czech Republic
- Drs. Groh and Bauer are with Bioinova, Ltd. in Prague, the Czech Republic
| |
Collapse
|
43
|
Reinhardt JW, Gooch KJ. An Agent-Based Discrete Collagen Fiber Network Model of Dynamic Traction Force-Induced Remodeling. J Biomech Eng 2019; 140:2654976. [PMID: 28975252 DOI: 10.1115/1.4037947] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Indexed: 01/17/2023]
Abstract
Microstructural properties of extracellular matrix (ECM) promote cell and tissue homeostasis as well as contribute to the formation and progression of disease. In order to understand how microstructural properties influence the mechanical properties and traction force-induced remodeling of ECM, we developed an agent-based model that incorporates repetitively applied traction force within a discrete fiber network. An important difference between our model and similar finite element models is that by implementing more biologically realistic dynamic traction, we can explore a greater range of matrix remodeling. Here, we validated our model by reproducing qualitative trends observed in three sets of experimental data reported by others: tensile and shear testing of cell-free collagen gels, collagen remodeling around a single isolated cell, and collagen remodeling between pairs of cells. In response to tensile and shear strain, simulated acellular networks with straight fibrils exhibited biphasic stress-strain curves indicative of strain-stiffening. When fibril curvature was introduced, stress-strain curves shifted to the right, delaying the onset of strain-stiffening. Our data support the notion that strain-stiffening might occur as individual fibrils successively align along the axis of strain and become engaged in tension. In simulations with a single, contractile cell, peak collagen displacement occurred closest to the cell and decreased with increasing distance. In simulations with two cells, compaction of collagen between cells appeared inversely related to the initial distance between cells. These results for cell-populated collagen networks match in vitro findings. A demonstrable benefit of modeling is that it allows for further analysis not feasible with experimentation. Within two-cell simulations, strain energy within the collagen network measured from the final state was relatively uniform around the outer surface of cells separated by 250 μm, but became increasingly nonuniform as the distance between cells decreased. For cells separated by 75 and 100 μm, strain energy peaked in the direction toward the other cell in the region in which fibrils become highly aligned and reached a minimum adjacent to this region, not on the opposite side of the cell as might be expected. This pattern of strain energy was partly attributable to the pattern of collagen compaction, but was still present when mapping strain energy divided by collagen density. Findings like these are of interest because fibril alignment, density, and strain energy may each contribute to contact guidance during tissue morphogenesis.
Collapse
Affiliation(s)
- James W Reinhardt
- Department of Biomedical Engineering, The Ohio State University, 270 Bevis Hall, 1080 Carmack Road, Columbus, OH 43210 e-mail:
| | - Keith J Gooch
- Department of Biomedical Engineering, The Ohio State University, 270 Bevis Hall, 1080 Carmack Road, Columbus, OH 43210.,Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, 473 W. 12th Avenue, Columbus, OH 43210 e-mail:
| |
Collapse
|
44
|
Azimzade Y, Saberi AA, Sahimi M. Regulation of migration of chemotactic tumor cells by the spatial distribution of collagen fiber orientation. Phys Rev E 2019; 99:062414. [PMID: 31330715 DOI: 10.1103/physreve.99.062414] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Indexed: 02/03/2023]
Abstract
Collagen fibers, an important component of the extracellular matrix (ECM), can both inhibit and promote cellular migration. In vitro studies have revealed that the fibers' orientations are crucial to cellular invasion, while in vivo investigations have led to the development of tumor-associated collagen signatures (TACS) as an important prognostic factor. Studying biophysical regulation of cell invasion and the effect of the fibers' orientation not only deepens our understanding of the phenomenon, but also helps classify the TACSs precisely, which is currently lacking. We present a stochastic model for random or chemotactic migration of cells in fibrous ECM, and study the role of the various factors in it. The model provides a framework for quantitative classification of the TACSs, and reproduces quantitatively recent experimental data for cell motility. It also indicates that the spatial distribution of the fibers' orientations and extended correlations between them, hitherto ignored, as well as dynamics of cellular motion all contribute to regulation of the cells' invasion length, which represents a measure of metastatic risk. Although the fibers' orientations trivially affect randomly moving cells, their effect on chemotactic cells is completely nontrivial and unexplored, which we study in this paper.
Collapse
Affiliation(s)
- Youness Azimzade
- Department of Physics, The University of Tehran, Tehran 14395-547, Iran
| | - Abbas Ali Saberi
- Department of Physics, The University of Tehran, Tehran 14395-547, Iran
| | - Muhammad Sahimi
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089-1211, USA
| |
Collapse
|
45
|
Crosby CO, Valliappan D, Shu D, Kumar S, Tu C, Deng W, Parekh SH, Zoldan J. Quantifying the Vasculogenic Potential of Induced Pluripotent Stem Cell-Derived Endothelial Progenitors in Collagen Hydrogels. Tissue Eng Part A 2019; 25:746-758. [PMID: 30618333 PMCID: PMC6535961 DOI: 10.1089/ten.tea.2018.0274] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/01/2019] [Indexed: 11/12/2022] Open
Abstract
IMPACT STATEMENT Our work reinforces the role of extracellular matrix (ECM) density and matrix metalloprotease activity on the formation of microvasculature from induced pluripotent stem cell (iPSC)-derived vascular cells. The cell-matrix interactions discussed in this study underscore the importance of understanding the role of mechanoregulation and matrix degradation on vasculogenesis and can potentially drive the development of ECM-mimicking angiogenic biomaterials. Furthermore, our work has broader implications concerning the response of iPSC-derived cells to the mechanics of engineered microenvironments. An understanding of these interactions will be critical to creating physiologically relevant transplantable tissue replacements.
Collapse
Affiliation(s)
- Cody O. Crosby
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Deepti Valliappan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - David Shu
- Department of Chemical Engineering, The University of Texas at Austin, Austin, Texas
| | - Sachin Kumar
- Department of Molecular Spectroscopy, Max Planck Institute for Polymer Research, Mainz, Germany
| | - Chengyi Tu
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Wei Deng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Sapun H. Parekh
- Department of Molecular Spectroscopy, Max Planck Institute for Polymer Research, Mainz, Germany
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
46
|
Crosby CO, Zoldan J. Mimicking the physical cues of the ECM in angiogenic biomaterials. Regen Biomater 2019; 6:61-73. [PMID: 30967961 PMCID: PMC6447000 DOI: 10.1093/rb/rbz003] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/02/2018] [Accepted: 12/29/2018] [Indexed: 12/12/2022] Open
Abstract
A functional microvascular system is imperative to build and maintain healthy tissue. Impaired microvasculature results in ischemia, thereby limiting the tissue's intrinsic regeneration capacity. Therefore, the ability to regenerate microvascular networks is key to the development of effective cardiovascular therapies. To stimulate the formation of new microvasculature, researchers have focused on fabricating materials that mimic the angiogenic properties of the native extracellular matrix (ECM). Here, we will review biomaterials that seek to imitate the physical cues that are natively provided by the ECM to encourage the formation of microvasculature in engineered constructs and ischemic tissue in the body.
Collapse
Affiliation(s)
- Cody O Crosby
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
47
|
Chen L, Wang ZC, Ma JJ, Sun WJ, Wang SW, Gu ZC, Yang X. Autologous nanofat transplantation accelerates foot wound healing in diabetic rats. Regen Med 2019; 14:231-241. [PMID: 30810491 DOI: 10.2217/rme-2018-0169] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aim: This study explored the effects of local transplantation of autologous nanofat in the treatment of rats with diabetic foot wounds. Materials & methods: Nanofat was transplanted into the left foot wound of diabetic rats. Phosphate-buffered saline injection in the right served as control. We measured wound size, the extent of epithelization, microvessel density and the expression levels of cytokines at six different time-points postoperation. Results: Compared with the control feet, nanofat-treated feet had significantly smaller wound areas at 7 and 9 days after grafting and showed better re-epithelialization, a greater number of microvessels and higher levels of angiogenic factor expression. Conclusion: This research shows that autologous nanofat transplantation can promote diabetic foot wound healing in rats.
Collapse
Affiliation(s)
- Li Chen
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Zheng-Cai Wang
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Jing-Jing Ma
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Wen-Jia Sun
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Shao-Wen Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Zi-Chun Gu
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| | - Xuan Yang
- Department of Plastic & Reconstructive Surgery, Sir Run Run Shaw Hospital, Medical College, Zhejiang University, Hang Zhou 310016, PR China
| |
Collapse
|
48
|
Salavati H, Soltani M. The impact of endothelial cells proliferation in a multiscale realistic reproduction of angiogenesis. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2018.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
49
|
Merino-Casallo F, Gomez-Benito MJ, Juste-Lanas Y, Martinez-Cantin R, Garcia-Aznar JM. Integration of in vitro and in silico Models Using Bayesian Optimization With an Application to Stochastic Modeling of Mesenchymal 3D Cell Migration. Front Physiol 2018; 9:1246. [PMID: 30271351 PMCID: PMC6142046 DOI: 10.3389/fphys.2018.01246] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 08/17/2018] [Indexed: 11/13/2022] Open
Abstract
Cellular migration plays a crucial role in many aspects of life and development. In this paper, we propose a computational model of 3D migration that is solved by means of the tau-leaping algorithm and whose parameters have been calibrated using Bayesian optimization. Our main focus is two-fold: to optimize the numerical performance of the mechano-chemical model as well as to automate the calibration process of in silico models using Bayesian optimization. The presented mechano-chemical model allows us to simulate the stochastic behavior of our chemically reacting system in combination with mechanical constraints due to the surrounding collagen-based matrix. This numerical model has been used to simulate fibroblast migration. Moreover, we have performed in vitro analysis of migrating fibroblasts embedded in 3D collagen-based fibrous matrices (2 mg/ml). These in vitro experiments have been performed with the main objective of calibrating our model. Nine model parameters have been calibrated testing 300 different parametrizations using a completely automatic approach. Two competing evaluation metrics based on the Bhattacharyya coefficient have been defined in order to fit the model parameters. These metrics evaluate how accurately the in silico model is replicating in vitro measurements regarding the two main variables quantified in the experimental data (number of protrusions and the length of the longest protrusion). The selection of an optimal parametrization is based on the balance between the defined evaluation metrics. Results show how the calibrated model is able to predict the main features observed in the in vitro experiments.
Collapse
Affiliation(s)
- Francisco Merino-Casallo
- Multiscale in Mechanical and Biological Engineering, Department of Mechanical Engineering, Aragón Institute of Engineering Research, Universidad de Zaragoza, Zaragoza, Spain
| | - Maria J Gomez-Benito
- Multiscale in Mechanical and Biological Engineering, Department of Mechanical Engineering, Aragón Institute of Engineering Research, Universidad de Zaragoza, Zaragoza, Spain
| | - Yago Juste-Lanas
- Multiscale in Mechanical and Biological Engineering, Department of Mechanical Engineering, Aragón Institute of Engineering Research, Universidad de Zaragoza, Zaragoza, Spain
| | - Ruben Martinez-Cantin
- Centro Universitario de la Defensa, Zaragoza, Spain.,SigOpt, Inc., San Francisco, CA, United States
| | - Jose M Garcia-Aznar
- Multiscale in Mechanical and Biological Engineering, Department of Mechanical Engineering, Aragón Institute of Engineering Research, Universidad de Zaragoza, Zaragoza, Spain
| |
Collapse
|
50
|
Deng C, Wang L, Feng J, Lu F. Treatment of human chronic wounds with autologous extracellular matrix/stromal vascular fraction gel: A STROBE-compliant study. Medicine (Baltimore) 2018; 97:e11667. [PMID: 30095623 PMCID: PMC6133447 DOI: 10.1097/md.0000000000011667] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stem cell therapy is considered as the most promising treatment for chronic wounds. Extracellular matrix/stromal vascular fraction gel (ECM/SVF gel), an adipose-derived stem cell-based cytotherapy, has shown healing potential in experimental wounds in animal models. However, the effects of ECM/SVF gel on human chronic wounds have not been investigated. The aim of the present study is to investigate the therapeutic effect of ECM/SVF gel on human chronic wounds.Autologous ECM/SVF gel was prepared and used to treat patients with chronic wounds in clinics, with negative pressure wound therapy as the positive control. Wound healing rate per week and histological changes were performed.The average wound healing rate per week in the ECM/SVF gel group was 34.55 ± 11.18% compared with 10.16 ± 2.67% in the negative pressure wound therapy group (P < .001). Histological analysis with hematoxylin and eosin, Masson's trichrome staining, and CD31 immunohistochemistry showed less lymphocyte infiltration, more collagen accumulation, and more newly formed vessels in the ECM/SVF gel group treated skins compared to the control.ECM/SVF gel is an effective therapeutic option for chronic wound healing in clinics.
Collapse
Affiliation(s)
- Chengliang Deng
- Department of Plastic Surgery, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Liangyue Wang
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Jingwei Feng
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Feng Lu
- Department of Plastic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| |
Collapse
|