1
|
Asadi Tokmedash M, Kim C, Chavda AP, Li A, Robins J, Min J. Engineering multifunctional surface topography to regulate multiple biological responses. Biomaterials 2025; 319:123136. [PMID: 39978049 PMCID: PMC11893264 DOI: 10.1016/j.biomaterials.2025.123136] [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: 09/27/2024] [Revised: 01/04/2025] [Accepted: 01/23/2025] [Indexed: 02/22/2025]
Abstract
Surface topography or curvature plays a crucial role in regulating cell behavior, influencing processes such as adhesion, proliferation, and gene expression. Recent advancements in nano- and micro-fabrication techniques have enabled the development of biomimetic systems that mimic native extracellular matrix (ECM) structures, providing new insights into cell-adhesion mechanisms, mechanotransduction, and cell-environment interactions. This review examines the diverse applications of engineered topographies across multiple domains, including antibacterial surfaces, immunomodulatory devices, tissue engineering scaffolds, and cancer therapies. It highlights how nanoscale features like nanopillars and nanospikes exhibit bactericidal properties, while many microscale patterns can direct stem cell differentiation and modulate immune cell responses. Furthermore, we discuss the interdisciplinary use of topography for combined applications, such as the simultaneous regulation of immune and tissue cells in 2D and 3D environments. Despite significant advances, key knowledge gaps remain, particularly regarding the effects of topographical cues on multicellular interactions and dynamic 3D contexts. This review summarizes current fabrication methods, explores specific and interdisciplinary applications, and proposes future research directions to enhance the design and utility of topographically patterned biomaterials in clinical and experimental settings.
Collapse
Affiliation(s)
| | - Changheon Kim
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ajay P Chavda
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Adrian Li
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jacob Robins
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jouha Min
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA; Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
2
|
van Sprang JF, Aarts JGM, Arts B, Brouns JEP, Komil MI, Bartels PAA, Dankers PYW. Supramolecular Additive Screening to Engineer Microfibrous Rafts for Expansion of Pluripotent Stem Cells in Dynamic Suspension. Adv Healthc Mater 2025; 14:e2404186. [PMID: 40059619 PMCID: PMC12023819 DOI: 10.1002/adhm.202404186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/02/2025] [Indexed: 04/26/2025]
Abstract
Human induced pluripotent stem cells (hiPSCs) hold the potential to generate any human tissue for transplantation in regenerative therapies. These complex cell therapies require billions of cells, which is challenging to acquire in planar adherent cultures. Transitioning hiPSCs to 3D suspension culture on microcarrier materials, often bead-shaped, improves the total surface area accessible to cells, thereby enabling culture scale-up. However, bead-shaped microcarriers do not have the optimal shape configuration, because it is the lowest surface-to-volume ratio of all geometrical shapes, and it also induces uncontrolled cell clumping. Application of synthetic, microfibrous rafts as a replacement for bead-shaped microcarriers potentially solves these issues. Here, microfibrous rafts are engineered by first screening a supramolecular biomaterial library composed of bisurea (BU)-peptide conjugate additives for its ability to induce hiPSC adhesion and maintenance of its pluripotent state, followed by electrospinning the screening-hit into raft-like structures. The resulting rafts contain cylinder-like microfibers, which have a higher surface-to-volume ratio compared to conventional bead-shaped microcarriers, and the flat configuration of the rafts prevents clumping.
Collapse
Affiliation(s)
- Johnick F. van Sprang
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Jasper G. M. Aarts
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Boris Arts
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Joyce E. P. Brouns
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Muhabbat I. Komil
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Paul A. A. Bartels
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| | - Patricia Y. W. Dankers
- Institute for Complex Molecular Systemsand Department of Biomedical EngineeringEindhoven University of TechnologyEindhoven5612AZThe Netherlands
| |
Collapse
|
3
|
Emonts C, Bauer B, Büchter C, Pufe T, Gries T, Tohidnezhad M. Effect of Collagen Coating and Fiber Profile on Tenocyte Growth on Braided Poly-ε-Caprolactone Scaffolds for Tendon and Ligament Regeneration. Int J Mol Sci 2025; 26:1735. [PMID: 40004198 PMCID: PMC11855873 DOI: 10.3390/ijms26041735] [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: 01/10/2025] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Using scaffolds is a promising alternative to current methods of treatment for ruptures of tendons and ligaments. However, scaffolds are subject to a wide range of challenges, including mechanical, degradation, process-related and biological requirements. Poly-ε-caprolactone (PCL) fibers have already shown potential for tendon tissue engineering (TTE) because of their degradation kinetics and excellent mechanical properties. The objective of this study was to enhance the PCL scaffold for TTE, specifically in regard to the filament morphology and collagen coating. PCL fibers were melt-spun as monofilaments with circular and snowflake-shaped cross-sections. Different scaffold densities were achieved by applying three different braiding angles in the braiding process. Morphological characterization was conducted including porosity and pore size distribution using µ-CT. The scaffolds were collagenized and cellularized with primary tenocytes for 7 days. Immunofluorescence staining showed a certain alignment of cell growing direction with fiber direction. In cell viability and cell proliferation assays, significant improvements in cell response were observed for the snowflake fiber and collagen coating groups, especially when combined. The data suggest that the utilization of non-circular fibers may facilitate enhanced cell guidance and surface area, while the application of a collagen coating could optimize the cellular environment for adhesion and proliferation.
Collapse
Affiliation(s)
- Caroline Emonts
- Institut für Textiltechnik, RWTH Aachen University, 52074 Aachen, Germany; (B.B.)
| | - Benedict Bauer
- Institut für Textiltechnik, RWTH Aachen University, 52074 Aachen, Germany; (B.B.)
| | - Charlotte Büchter
- Institut für Textiltechnik, RWTH Aachen University, 52074 Aachen, Germany; (B.B.)
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, 52074 Aachen, Germany
| | - Thomas Gries
- Institut für Textiltechnik, RWTH Aachen University, 52074 Aachen, Germany; (B.B.)
| | - Mersedeh Tohidnezhad
- Department of Anatomy and Cell Biology, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
4
|
Dutta D, Nuntapramote T, Rehders M, Brix K, Brüggemann D. Topography-Mediated Induction of Epithelial Mesenchymal Transition via Alumina Textiles for Potential Wound Healing Applications. J Biomed Mater Res A 2025; 113:e37826. [PMID: 39529481 DOI: 10.1002/jbm.a.37826] [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: 05/17/2024] [Revised: 09/18/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Substrate topography is vital in determining cell growth and fate of cellular behavior. Although current in vitro studies of the underlying cellular signaling pathways mostly rely on their induction by specific growth factors or chemicals, the influence of substrate topography on specific changes in cells has been explored less often. This study explores the impact of substrate topography, specifically the tricot knit microfibrous structure of alumina textiles, on cell behavior, focusing on fibroblasts and keratinocytes for potential wound healing applications. The textiles, studied for the first time as in vitro substrates, demonstrated support for keratinocyte adhesion, leading to alterations in cell morphology and the expression of E-cadherin and fibronectin. These topography-induced changes resembled the epithelial-to-mesenchymal transition (EMT), crucial for wound healing, and were specific to keratinocytes and absent in identically treated fibroblasts. Biochemically induced EMT in keratinocytes cultured on flat alumina substrates mirrored the changes seen with alumina textiles alone, suggesting the tricot knit microfibrous topography could serve as an in vitro model system to induce EMT-like mechanisms. These results enhance our understanding of how substrate topography influences EMT-related processes in wound healing, paving the way for further evaluation of microfibrous alumina textiles as innovative wound dressings.
Collapse
Affiliation(s)
| | | | - Maren Rehders
- School of Science, Constructor University, Bremen, Germany
| | - Klaudia Brix
- School of Science, Constructor University, Bremen, Germany
| | - Dorothea Brüggemann
- Institute for Biophysics, University of Bremen, Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, Bremen, Germany
- Biophysics and Applied Biomaterials, University of Applied Sciences, Hochschule Bremen, Germany
| |
Collapse
|
5
|
Drewry MD, Shi D, Dailey MT, Rothermund K, Trbojevic S, Almarza AJ, Cui XT, Syed-Picard FN. Enhancing facial nerve regeneration with scaffold-free conduits engineered using dental pulp stem cells and their endogenous, aligned extracellular matrix. J Neural Eng 2024; 21:056015. [PMID: 39197480 PMCID: PMC11406051 DOI: 10.1088/1741-2552/ad749d] [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: 01/30/2024] [Revised: 07/02/2024] [Accepted: 08/28/2024] [Indexed: 09/01/2024]
Abstract
Objective. Engineered nerve conduits must simultaneously enhance axon regeneration and orient axon extension to effectively restore function of severely injured peripheral nerves. The dental pulp contains a population of stem/progenitor cells that endogenously express neurotrophic factors (NTFs), growth factors known to induce axon repair. We have previously generated scaffold-free dental pulp stem/progenitor cell (DPSC) sheets comprising an aligned extracellular matrix (ECM). Through the intrinsic NTF expression of DPSCs and the topography of the aligned ECM, these sheets both induce and guide axon regeneration. Here, the capacity of bioactive conduits generated using these aligned DPSC sheets to restore function in critical-sized nerve injuries in rodents was evaluated.Approach. Scaffold-free nerve conduits were formed by culturing DPSCs on a substrate with aligned microgrooves, inducing the cells to align and deposit an aligned ECM. The sheets were then detached from the substrate and assembled into scaffold-free cylindrical tissues.Main results. In vitroanalyses confirmed that scaffold-free DPSC conduits maintained an aligned ECM and had uniformly distributed NTF expression. Implanting the aligned DPSC conduits across critical-sized defects in the buccal branch of rat facial nerves resulted in the regeneration of a fascicular nerve-like structure and myelinated axon extension across the injury site. Furthermore, compound muscle action potential and stimulated whisker movement measurements revealed that the DPSC conduit treatment promoted similar functional recovery compared to the clinical standard of care, autografts. Significance. This study demonstrates that scaffold-free aligned DPSC conduits supply trophic and guidance cues, key design elements needed to successfully promote and orient axon regeneration. Consequently, these conduits restore function in nerve injuries to similar levels as autograft treatments. These conduits offer a novel bioactive approach to nerve repair capable of improving clinical outcomes and patient quality of life.
Collapse
Affiliation(s)
- Michelle D Drewry
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Delin Shi
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Matthew T Dailey
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Oral and Maxillofacial Surgery, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kristi Rothermund
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Sara Trbojevic
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Alejandro J Almarza
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Xinyan T Cui
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Fatima N Syed-Picard
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
6
|
Conner AA, Yao Y, Chan SW, Jain D, Wong SM, Yim EKF, Rizwan M. High-throughput analysis of topographical cues for the expansion of murine pluripotent stem cells. NANOTECHNOLOGY 2024; 35:455101. [PMID: 39084233 DOI: 10.1088/1361-6528/ad6994] [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: 04/05/2024] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
The expansion of pluripotent stem cells (PSCs)in vitroremains a critical barrier to their use in tissue engineering and regenerative medicine. Biochemical methods for PSC expansion are known to produce heterogeneous cell populations with varying states of pluripotency and are cost-intensive, hindering their clinical translation. Engineering biomaterials to physically control PSC fate offers an alternative approach. Surface or substrate topography is a promising design parameter for engineering biomaterials. Topographical cues have been shown to elicit profound effects on stem cell differentiation and proliferation. Previous reports have shown isotropic substrate topographies to be promising in expanding PSCs. However, the optimal feature to promote PSC proliferation and the pluripotent state has not yet been determined. In this work, the MultiARChitecture (MARC) plate is developed to conduct a high-throughput analysis of topographical cues in a 96-well plate format. The MARC plate is a reproducible and customizable platform for the analysis of multiple topographical patterns and features and is compatible with both microscopic assays and molecular biology techniques. The MARC plate is used to evaluate the expression of pluripotency markersOct4, Nanog, andSox2and the differentiation markerLmnAas well as the proliferation of murine embryonic stem (mES) cells. Our systematic analyses identified three topographical patterns that maintain pluripotency in mES cells after multiple passages: 1µm pillars (1µm spacing, square arrangement), 2µm wells (c-c (x, y) = 4, 4µm), and 5µm pillars (c-c (x, y) = 7.5, 7.5µm). This study represents a step towards developing a biomaterial platform for controlled murine PSC expansion.
Collapse
Affiliation(s)
- Abigail A Conner
- Department of Chemical Engineering, University of Waterloo, Waterloo, Canada
| | - Yuan Yao
- Department of Chemical Engineering, University of Waterloo, Waterloo, Canada
| | - Sarah W Chan
- Department of Chemical Engineering, University of Waterloo, Waterloo, Canada
| | - Deepak Jain
- Department of Chemical Engineering, University of Waterloo, Waterloo, Canada
| | - Suzanne M Wong
- Department of Chemical Engineering, University of Waterloo, Waterloo, Canada
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, Canada
- Center for Biotechnology and Bioengineering, University of Waterloo, Waterloo, Canada
- Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Canada
| | - Muhammad Rizwan
- Department of Chemical Engineering, University of Waterloo, Waterloo, Canada
- Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| |
Collapse
|
7
|
Tai Y, Goodrich R, Maldonado M, Ortiz J, Martinez J, Ico G, Ko A, Shih HP, Nam J. Nanofiber-microwell cell culture system for spatially patterned differentiation of pluripotent stem cells in 3D. Mater Today Bio 2024; 26:101109. [PMID: 38883422 PMCID: PMC11180340 DOI: 10.1016/j.mtbio.2024.101109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/04/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024] Open
Abstract
The intricate interplay between biochemical and physical cues dictates pluripotent stem cell (PSC) differentiation to form various tissues. While biochemical modulation has been extensively studied, the role of biophysical microenvironments in early lineage commitment remains elusive. Here, we introduce a novel 3D cell culture system combining electrospun nanofibers with microfabricated polydimethylsiloxane (PDMS) patterns. This system enables the controlled formation of semispherical human induced pluripotent stem cell (hiPSC) colonies, facilitating the investigation of local mechanical stem cell niches on mechano-responsive signaling and lineage specification. Our system unveiled spatially organized RhoA activity coupled with actin-myosin cable formation, suggesting mechano-dependent hiPSC behaviors. Nodal network analysis of RNA-seq data revealed RhoA downstream regulation of YAP signaling, DNA histone modifications, and patterned germ layer specification. Notably, altering colony morphology through controlled PDMS microwell shaping effectively modulated the spatial distribution of mechano-sensitive mediators and subsequent differentiation. This study provides a cell culture platform to decipher the role of biophysical cues in early embryogenesis, offering valuable insights for material design in tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Youyi Tai
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Robyn Goodrich
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Maricela Maldonado
- Department of Biomedical Engineering, California State University Long Beach, CA, 90840, USA
| | - Jessica Ortiz
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Jeniree Martinez
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Gerardo Ico
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Angel Ko
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| | - Hung Ping Shih
- Department of Translational Research and Cellular Therapeutics, City of Hope, CA, 91010, USA
| | - Jin Nam
- Department of Bioengineering, University of California, Riverside, CA, 92521, USA
| |
Collapse
|
8
|
Wang L, Shi Y, Qiu Z, Dang J, Sun L, Qu X, He J, Fan H. Bioactive 3D Electrohydrodynamic Printed Lattice Architectures Augment Tenogenesis of Tendon Stem/Progenitor Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:18574-18590. [PMID: 38567837 DOI: 10.1021/acsami.4c01372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Tendon defect repair remains a tough clinical procedure that hinders functional motion in patients. Electrohydrodynamic (EHD) three-dimensional (3D) printing, as a novel strategy, can controllably fabricate biomimetic micro/nanoscale architecture, but the hydrophobic and bioinert nature of polymers might be adverse to cell-material interplay. In this work, 3D EHD printed polycaprolactone (PCL) was immobilized on basic fibroblast growth factor (bFGF) using polydopamine (PDA), and the proliferation and tenogenic differentiation of tendon stem/progenitor cells (TSPCs) in vitro was researched. A subcutaneous model was established to evaluate the effects of tenogenesis and immunomodulation. We then investigated the in situ implantation and immunomodulation effects in an Achilles tendon defect model. After immobilization of bFGF, the scaffolds profoundly facilitated proliferation and tenogenic differentiation; however, PDA had only a proliferative effect. Intriguingly, the bFGF immobilized on EHD printed PCL indicated a synergistic effect on the highest expression of tenogenic gene and protein markers at 14 days, and the tenogenesis may be induced by activating the transforming growth factor-β (TGF-β) signal pathway in vitro. The subcutaneous engraftment study confirmed a tendon-like structure, similar to that of the native tendon, as well as an M2 macrophage polarization effect. Additionally, the bioactive scaffold exhibited superior efficacy in new collagen formation and repair of Achilles tendon defects. Our study revealed that the topographic cues alone were insufficient to trigger tenogenic differentiation, requiring appropriate chemical signals, and that appropriate immunomodulation was conducive to tenogenesis. The tenogenesis of TSPCs on the bioactive scaffold may be correlated with the TGF-β signal pathway and M2 macrophage polarization.
Collapse
Affiliation(s)
- Lei Wang
- Department of Orthopedic Surgery, Xijing Hospital, the Air Force Military Medical University, Xi'an 710032, China
| | - Yubo Shi
- Department of Orthopedic Surgery, Xijing Hospital, the Air Force Military Medical University, Xi'an 710032, China
| | - Zhennan Qiu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jingyi Dang
- Department of Orthopedic Surgery, Xijing Hospital, the Air Force Military Medical University, Xi'an 710032, China
| | - Liguo Sun
- Shaanxi Province Hospital of Traditional Chinese Medicine, Xi'an 710018, China
| | - Xiaoli Qu
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
- Rapid Manufacturing Research Center of Shaanxi Province, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hongbin Fan
- Department of Orthopedic Surgery, Xijing Hospital, the Air Force Military Medical University, Xi'an 710032, China
| |
Collapse
|
9
|
Hu Y, Li CY, Lu Q, Kuang Y. Multiplex miRNA reporting platform for real-time profiling of living cells. Cell Chem Biol 2024; 31:150-162.e7. [PMID: 38035883 DOI: 10.1016/j.chembiol.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/15/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023]
Abstract
Accurately characterizing cell types within complex cell structures provides invaluable information for comprehending the cellular status during biological processes. In this study, we have developed an miRNA-switch cocktail platform capable of reporting and tracking the activities of multiple miRNAs (microRNAs) at the single-cell level, while minimizing disruption to the cell culture. Drawing on the principles of traditional miRNA-sensing mRNA switches, our platform incorporates subcellular tags and employs intelligent engineering to segment three subcellular regions using two fluorescent proteins. These designs enable the quantification of multiple miRNAs within the same cell. Through our experiments, we have demonstrated the platform's ability to track marker miRNA levels during cell differentiation and provide spatial information of heterogeneity on outlier cells exhibiting extreme miRNA levels. Importantly, this platform offers real-time and in situ miRNA reporting, allowing for multidimensional evaluation of cell profile and paving the way for a comprehensive understanding of cellular events during biological processes.
Collapse
Affiliation(s)
- Yaxin Hu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Cheuk Yin Li
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Qiuyu Lu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China
| | - Yi Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong SAR, China.
| |
Collapse
|
10
|
Eftekhari BS, Song D, Janmey PA. Electrical Stimulation of Human Mesenchymal Stem Cells on Conductive Substrates Promotes Neural Priming. Macromol Biosci 2023; 23:e2300149. [PMID: 37571815 PMCID: PMC10880582 DOI: 10.1002/mabi.202300149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/29/2023] [Indexed: 08/13/2023]
Abstract
Electrical stimulation (ES) within a conductive scaffold is potentially beneficial in encouraging the differentiation of stem cells toward a neuronal phenotype. To improve stem cell-based regenerative therapies, it is essential to use electroconductive scaffolds with appropriate stiffnesses to regulate the amount and location of ES delivery. Herein, biodegradable electroconductive substrates with different stiffnesses are fabricated from chitosan-grafted-polyaniline (CS-g-PANI) copolymers. Human mesenchymal stem cells (hMSCs) cultured on soft conductive scaffolds show a morphological change with significant filopodial elongation after electrically stimulated culture along with upregulation of neuronal markers and downregulation of glial markers. Compared to stiff conductive scaffolds and non-conductive CS scaffolds, soft conductive CS-g-PANI scaffolds promote increased expression of microtubule-associated protein 2 (MAP2) and neurofilament heavy chain (NF-H) after application of ES. At the same time, there is a decrease in the expression of the glial markers glial fibrillary acidic protein (GFAP) and vimentin after ES. Furthermore, the elevation of intracellular calcium [Ca2+ ] during spontaneous, cell-generated Ca2+ transients further suggests that electric field stimulation of hMSCs cultured on conductive substrates can promote a neural-like phenotype. The findings suggest that the combination of the soft conductive CS-g-PANI substrate and ES is a promising new tool for enhancing neuronal tissue engineering outcomes.
Collapse
Affiliation(s)
| | - Dawei Song
- Department of Physiology and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A. Janmey
- Department of Bioengineering, University of Pennsylvania, Philadelphia, USA
- Department of Physiology and Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Samal P, Kumar Samal JR, Rho HS, van Beurden D, van Blitterswijk C, Truckenmüller R, Giselbrecht S. Direct deep UV lithography to micropattern PMMA for stem cell culture. Mater Today Bio 2023; 22:100779. [PMID: 37701129 PMCID: PMC10494264 DOI: 10.1016/j.mtbio.2023.100779] [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: 03/17/2023] [Revised: 06/22/2023] [Accepted: 08/25/2023] [Indexed: 09/14/2023] Open
Abstract
Microengineering is increasingly being used for controlling the microenvironment of stem cells. Here, a novel method for fabricating structures with subcellular dimensions in commonly available thermoplastic poly(methyl methacrylate) (PMMA) is shown. Microstructures are produced in PMMA substrates using Deep Ultraviolet lithography, and the effect of different developers is described. Microgrooves fabricated in PMMA are used for the neuronal differentiation of mouse embryonic stem cells (mESCs) directly on the polymer. The fabrication of 3D, curvilinear patterned surfaces is also highlighted. A 3D multilayered microfluidic chip is fabricated using this method, which includes a porous polycarbonate (PC) membrane as cell culture substrate. Besides directly manufacturing PMMA-based microfluidic devices, an application of the novel approach is shown where a reusable PMMA master is created for replicating microstructures with polydimethylsiloxane (PDMS). As an application example, microchannels fabricated in PDMS are used to selectively expose mESCs to soluble factors in a localized manner. The described microfabrication process offers a remarkably simple method to fabricate for example multifunctional topographical or microfluidic culture substrates outside cleanrooms, thereby using inexpensive and widely accessible equipment. The versatility of the underlying process could find various applications also in optical systems and surface modification of biomedical implants.
Collapse
Affiliation(s)
- Pinak Samal
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Jay Rabindra Kumar Samal
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Hoon Suk Rho
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
- Mepsgen Co., Ltd., 7F, Hanyang Tower, 12, Beobwon-ro-11-gil, Songpa-gu, Seoul, Republic of Korea
| | - Denis van Beurden
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Clemens van Blitterswijk
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Universiteitssingel 40, 6229 ER, Maastricht, the Netherlands
| |
Collapse
|
12
|
Gerardo‐Nava JL, Jansen J, Günther D, Klasen L, Thiebes AL, Niessing B, Bergerbit C, Meyer AA, Linkhorst J, Barth M, Akhyari P, Stingl J, Nagel S, Stiehl T, Lampert A, Leube R, Wessling M, Santoro F, Ingebrandt S, Jockenhoevel S, Herrmann A, Fischer H, Wagner W, Schmitt RH, Kiessling F, Kramann R, De Laporte L. Transformative Materials to Create 3D Functional Human Tissue Models In Vitro in a Reproducible Manner. Adv Healthc Mater 2023; 12:e2301030. [PMID: 37311209 PMCID: PMC11468549 DOI: 10.1002/adhm.202301030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/21/2023] [Indexed: 06/15/2023]
Abstract
Recreating human tissues and organs in the petri dish to establish models as tools in biomedical sciences has gained momentum. These models can provide insight into mechanisms of human physiology, disease onset, and progression, and improve drug target validation, as well as the development of new medical therapeutics. Transformative materials play an important role in this evolution, as they can be programmed to direct cell behavior and fate by controlling the activity of bioactive molecules and material properties. Using nature as an inspiration, scientists are creating materials that incorporate specific biological processes observed during human organogenesis and tissue regeneration. This article presents the reader with state-of-the-art developments in the field of in vitro tissue engineering and the challenges related to the design, production, and translation of these transformative materials. Advances regarding (stem) cell sources, expansion, and differentiation, and how novel responsive materials, automated and large-scale fabrication processes, culture conditions, in situ monitoring systems, and computer simulations are required to create functional human tissue models that are relevant and efficient for drug discovery, are described. This paper illustrates how these different technologies need to converge to generate in vitro life-like human tissue models that provide a platform to answer health-based scientific questions.
Collapse
|
13
|
Nattasit P, Niibe K, Yamada M, Ohori-Morita Y, Limraksasin P, Tiskratok W, Yamamoto M, Egusa H. Stiffness-Tunable Hydrogel-Sandwich Culture Modulates the YAP-Mediated Mechanoresponse in Induced-Pluripotent Stem Cell Embryoid Bodies and Augments Cardiomyocyte Differentiation. Macromol Biosci 2023:e2300021. [PMID: 36871184 DOI: 10.1002/mabi.202300021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Indexed: 03/06/2023]
Abstract
Microenvironmental factors, including substrate stiffness, regulate stem cell behavior and differentiation. However, the effects of substrate stiffness on the behavior of induced pluripotent stem cell (iPSC)- derived embryoid bodies (EB) remain unclear. To investigate the effects of mechanical cues on iPSC-EB differentiation, a 3D hydrogel-sandwich culture (HGSC) system is developed that controls the microenvironment surrounding iPSC-EBs using a stiffness-tunable polyacrylamide hydrogel assembly. Mouse iPSC-EBs are seeded between upper and lower polyacrylamide hydrogels of differing stiffness (Young's modulus [E'] = 54.3 ± 7.1 kPa [hard], 28.1 ± 2.3 kPa [moderate], and 5.1 ± 0.1 kPa [soft]) and cultured for 2 days. HGSC induces stiffness-dependent activation of the yes-associated protein (YAP) mechanotransducer and actin cytoskeleton rearrangement in the iPSC-EBs. Moreover, moderate-stiffness HGSC specifically upregulates the mRNA and protein expression of ectoderm and mesoderm lineage differentiation markers in iPSC-EBs via YAP-mediated mechanotransduction. Pretreatment of mouse iPSC-EBs with moderate-stiffness HGSC promotes cardiomyocyte (CM) differentiation and structural maturation of myofibrils. The proposed HGSC system provides a viable platform for investigating the role of mechanical cues on the pluripotency and differentiation of iPSCs that can be beneficial for research into tissue regeneration and engineering.
Collapse
Affiliation(s)
- Praphawi Nattasit
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Kunimichi Niibe
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Masahiro Yamada
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Yumi Ohori-Morita
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Phoonsuk Limraksasin
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
- Dental Stem Cell Biology Research Unit, Center of Excellence for Regenerative Dentistry, and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Watcharaphol Tiskratok
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Masaya Yamamoto
- Department of Material Processing, Tohoku University Graduate School of Engineering, Sendai, Miyagi, 980-8579, Japan
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
14
|
Zeevaert K, Goetzke R, Elsafi Mabrouk MH, Schmidt M, Maaßen C, Henneke AC, He C, Gillner A, Zenke M, Wagner W. YAP1 is essential for self-organized differentiation of pluripotent stem cells. BIOMATERIALS ADVANCES 2023; 146:213308. [PMID: 36774716 DOI: 10.1016/j.bioadv.2023.213308] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/26/2023]
Abstract
Induced pluripotent stem cells (iPSCs) form aggregates that recapitulate aspects of the self-organization in early embryogenesis. Within few days, cells undergo a transition from epithelial-like structures to organized three-dimensional embryoid bodies (EBs) with upregulation of germ layer-specific genes. However, it is largely unclear, which signaling cascades regulate self-organized differentiation. The Yes-associated protein 1 (YAP1) is a downstream effector of the Hippo pathway and essential mechanotransducer. YAP1 has been suggested to play a crucial role for early embryo development, but the relevance for early germ layer commitment of human iPSCs remains to be elucidated. To gain insights into the function of YAP1 in early cell-fate decisions, we generated YAP1 knockout (YAP-/-) iPSC lines with CRISPR/Cas9 technology and analyzed transcriptomic and epigenetic modifications. YAP-/- iPSCs showed increased expression of several YAP1 targets and of NODAL, an important regulator of cell differentiation. Furthermore, YAP1 deficiency evoked global DNA methylation changes. Directed differentiation of adherent iPSC colonies towards endoderm, mesoderm, and ectoderm could be induced, albeit endodermal and ectodermal differentiation showed transcriptomic and epigenetic changes in YAP-/- lines. Notably, in undirected self-organized YAP-/- EBs germ layer specification was clearly impaired. This phenotype was rescued via lentiviral overexpression of YAP1 and also by NODAL inhibitors. Our results demonstrate that YAP1 plays an important role during early germ layer specification of iPSCs, particularly for the undirected self-organization of EBs, and this is at least partly attributed to activation of the NODAL signaling.
Collapse
Affiliation(s)
- Kira Zeevaert
- Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Institute for Stem Cell Biology, University Hospital of RWTH Aachen, 52074 Aachen, Germany.
| | - Roman Goetzke
- Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Institute for Stem Cell Biology, University Hospital of RWTH Aachen, 52074 Aachen, Germany; PL BioScience, Technology Centre Aachen, 52068 Aachen, Germany
| | - Mohamed H Elsafi Mabrouk
- Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Institute for Stem Cell Biology, University Hospital of RWTH Aachen, 52074 Aachen, Germany
| | - Marco Schmidt
- Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Institute for Stem Cell Biology, University Hospital of RWTH Aachen, 52074 Aachen, Germany
| | - Catharina Maaßen
- Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Institute for Stem Cell Biology, University Hospital of RWTH Aachen, 52074 Aachen, Germany
| | - Ann-Christine Henneke
- Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Institute for Stem Cell Biology, University Hospital of RWTH Aachen, 52074 Aachen, Germany
| | - Chao He
- Chair for Laser Technology LLT, RWTH Aachen University, 52074 Aachen, Germany
| | - Arnold Gillner
- Chair for Laser Technology LLT, RWTH Aachen University, 52074 Aachen, Germany
| | - Martin Zenke
- Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Institute for Stem Cell Biology, University Hospital of RWTH Aachen, 52074 Aachen, Germany; Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany
| | - Wolfgang Wagner
- Helmholtz-Institute for Biomedical Engineering, Medical Faculty of RWTH Aachen University, 52074 Aachen, Germany; Institute for Stem Cell Biology, University Hospital of RWTH Aachen, 52074 Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany.
| |
Collapse
|
15
|
Taheri S, Ghazali ZS, Montazeri L, Ebrahim FA, Javadpour J, Kamguyan K, Thormann E, Renaud P, Bonakdar S. Engineered substrates incapable of induction of chondrogenic differentiation compared to the chondrocyte imprinted substrates. Biomed Mater 2023; 18. [PMID: 36693281 DOI: 10.1088/1748-605x/acb5d7] [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: 07/07/2022] [Accepted: 01/24/2023] [Indexed: 01/26/2023]
Abstract
It is well established that surface topography can affect cell functions. However, finding a reproducible and reliable method for regulating stem cell behavior is still under investigation. It has been shown that cell imprinted substrates contain micro- and nanoscale structures of the cell membrane that serve as hierarchical substrates, can successfully alter stem cell fate. This study investigated the effect of the overall cell shape by fabricating silicon wafers containing pit structure in the average size of spherical-like chondrocytes using photolithography technique. We also used chondrocyte cell line (C28/I2) with spindle-like shape to produce cell imprinted substrates. The effect of all substrates on the differentiation of adipose-derived mesenchymal stem cells (ADSCs) has been studied. The AFM and scanning electron microscopy images of the prepared substrates demonstrated that the desired shapes were successfully transferred to the substrates. Differentiation of ADSCs was investigated by immunostaining for mature chondrocyte marker, collagen II, and gene expression of collagen II, Sox9, and aggrecan markers. C28/I2 imprinted substrate could effectively enhanced chondrogenic differentiation compared to regular pit patterns on the wafer. It can be concluded that cell imprinted substrates can induce differentiation signals better than engineered lithographic substrates. The nanostructures on the cell-imprinted patterns play a crucial role in harnessing cell fate. Therefore, the patterns must include the nano-topographies to have reliable and reproducible engineered substrates.
Collapse
Affiliation(s)
- Shiva Taheri
- National Cell Bank Department, Iran Pasteur Institute, Tehran, Iran.,School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Zahra Sadat Ghazali
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Jafar Javadpour
- School of Metallurgy and Materials Engineering, Iran University of Science and Technology, Tehran, Iran
| | - Khorshid Kamguyan
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Esben Thormann
- Department of Chemistry, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Philippe Renaud
- STI-IMT-LMIS4, École Polytechnique Fédérale de Lausanne, Station 17, 1015 Lausanne, Switzerland
| | - Shahin Bonakdar
- National Cell Bank Department, Iran Pasteur Institute, Tehran, Iran
| |
Collapse
|
16
|
Moxon SR, Richards D, Dobre O, Wong LS, Swift J, Richardson SM. Regulation of Mesenchymal Stem Cell Morphology Using Hydrogel Substrates with Tunable Topography and Photoswitchable Stiffness. Polymers (Basel) 2022; 14:polym14245338. [PMID: 36559706 PMCID: PMC9788018 DOI: 10.3390/polym14245338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Cell function can be directly influenced by the mechanical and structural properties of the extracellular environment. In particular, cell morphology and phenotype can be regulated via the modulation of both the stiffness and surface topography of cell culture substrates. Previous studies have highlighted the ability to design cell culture substrates to optimise cell function. Many such examples, however, employ photo-crosslinkable polymers with a terminal stiffness or surface profile. This study presents a system of polyacrylamide hydrogels, where the surface topography can be tailored and the matrix stiffness can be altered in situ with photoirradiation. The process allows for the temporal regulation of the extracellular environment. Specifically, the surface topography can be tailored via reticulation parameters to include creased features with control over the periodicity, length and branching. The matrix stiffness can also be dynamically tuned via exposure to an appropriate dosage and wavelength of light, thus, allowing for the temporal regulation of the extracellular environment. When cultured on the surface of the hydrogels, the morphology and alignment of immortalised human mesenchymal stem cells can be directly influenced through the tailoring of surface creases, while cell size can be altered via changes in matrix stiffness. This system offers a new platform to study cellular mechanosensing and the influence of extracellular cues on cell phenotype and function.
Collapse
Affiliation(s)
- Samuel R. Moxon
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
- The Henry Royce Institute, University of Manchester, Manchester M13 9PL, UK
| | - David Richards
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Oana Dobre
- Centre for the Cellular Microenvironment, Advanced Research Centre, University of Glasgow, Glasgow G12 8LT, UK
| | - Lu Shin Wong
- Manchester Institute of Biotechnology and School of Chemistry, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
- Correspondence: (L.S.W.); (J.S.); (S.M.R.)
| | - Joe Swift
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
- Wellcome Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Correspondence: (L.S.W.); (J.S.); (S.M.R.)
| | - Stephen M. Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
- Correspondence: (L.S.W.); (J.S.); (S.M.R.)
| |
Collapse
|
17
|
Damkham N, Issaragrisil S, Lorthongpanich C. Role of YAP as a Mechanosensing Molecule in Stem Cells and Stem Cell-Derived Hematopoietic Cells. Int J Mol Sci 2022; 23:14634. [PMID: 36498961 PMCID: PMC9737411 DOI: 10.3390/ijms232314634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/11/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022] Open
Abstract
Yes-associated protein (YAP) and WW domain-containing transcription regulator protein 1 (WWTR1, also known as TAZ) are transcriptional coactivators in the Hippo signaling pathway. Both are well-known regulators of cell proliferation and organ size control, and they have significant roles in promoting cell proliferation and differentiation. The roles of YAP and TAZ in stem cell pluripotency and differentiation have been extensively studied. However, the upstream mediators of YAP and TAZ are not well understood. Recently, a novel role of YAP in mechanosensing and mechanotransduction has been reported. The present review updates information on the regulation of YAP by mechanical cues such as extracellular matrix stiffness, fluid shear stress, and actin cytoskeleton tension in stem cell behaviors and differentiation. The review explores mesenchymal stem cell fate decisions, pluripotent stem cells (PSCs), self-renewal, pluripotency, and differentiation to blood products. Understanding how cells sense their microenvironment or niche and mimic those microenvironments in vitro could improve the efficiency of producing stem cell products and the efficacy of the products.
Collapse
Affiliation(s)
- Nattaya Damkham
- Siriraj Center of Excellence for Stem cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Bangkok Hematology Center, Wattanosoth Hospital, BDMS Center of Excellence for Cancer, Bangkok 10310, Thailand
| | - Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
18
|
Lee G, Oh Y, Nam JT, Ji S, Jang AR, Jeong DW, Kang M, Lee SS, Chae S, Cho D, Hwang JY, Lee K, Lee JO. Multifunctional-high resolution imaging plate based on hydrophilic graphene for digital pathology. NANOTECHNOLOGY 2022; 33:505101. [PMID: 36095982 DOI: 10.1088/1361-6528/ac9143] [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: 06/17/2022] [Accepted: 09/12/2022] [Indexed: 06/15/2023]
Abstract
In the present study, we showed that hydrophilic graphene can serve as an ideal imaging plate for biological specimens. Graphene being a single-atom-thick semi-metal with low secondary electron emission, array tomography analysis of serial sections of biological specimens on a graphene substrate showed excellent image quality with improvedz-axis resolution, without including any conductive surface coatings. However, the hydrophobic nature of graphene makes the placement of biological specimens difficult; graphene functionalized with polydimethylsiloxane oligomer was fabricated using a simple soft lithography technique and then processed with oxygen plasma to provide hydrophilic graphene with minimal damage to graphene. High-quality scanning electron microscopy images of biological specimens free from charging effects or distortion were obtained, and the optical transparency of graphene enabled fluorescence imaging of the specimen; high-resolution correlated electron and light microscopy analysis of the specimen became possible with the hydrophilic graphene plate.
Collapse
Affiliation(s)
- Geonhee Lee
- Advanced Materials Division, Korea Research Institute of Chemical Technology, Gajeongro 141, Daejeon, Republic of Korea
| | - Yuna Oh
- Korea Institute of Science and Technology, 5. Hwarang-ro 14-gil, Seongbuk-gu, Seoul, Republic of Korea
| | - Jung Tae Nam
- Institute of Advanced Composite Materials, Korea Institute of Science and Technology, Jeonbuk, 55324, Republic of Korea
| | - Seulgi Ji
- Advanced Materials Division, Korea Research Institute of Chemical Technology, Gajeongro 141, Daejeon, Republic of Korea
| | - A-Rang Jang
- Division of Electrical, Electronic and Control Engineering, Kongju National University, Cheonan 31080, Republic of Korea
| | - Du Won Jeong
- Advanced Materials Division, Korea Research Institute of Chemical Technology, Gajeongro 141, Daejeon, Republic of Korea
| | - MinSoung Kang
- Advanced Materials Division, Korea Research Institute of Chemical Technology, Gajeongro 141, Daejeon, Republic of Korea
| | - Sun Sook Lee
- Advanced Materials Division, Korea Research Institute of Chemical Technology, Gajeongro 141, Daejeon, Republic of Korea
| | - Soosang Chae
- Department of Nanostructured Materials, Leibniz Institute of Polymer Research Dresden, D-01069, Dresden, Germany
| | - Donghwi Cho
- Advanced Materials Division, Korea Research Institute of Chemical Technology, Gajeongro 141, Daejeon, Republic of Korea
| | - Jun Yeon Hwang
- Institute of Advanced Composite Materials, Korea Institute of Science and Technology, Jeonbuk, 55324, Republic of Korea
| | - Kyungeun Lee
- Korea Institute of Science and Technology, 5. Hwarang-ro 14-gil, Seongbuk-gu, Seoul, Republic of Korea
| | - Jeong-O Lee
- Advanced Materials Division, Korea Research Institute of Chemical Technology, Gajeongro 141, Daejeon, Republic of Korea
| |
Collapse
|
19
|
Zhao M, Gao X, Wei J, Tu C, Zheng H, Jing K, Chu J, Ye W, Groth T. Chondrogenic differentiation of mesenchymal stem cells through cartilage matrix-inspired surface coatings. Front Bioeng Biotechnol 2022; 10:991855. [PMID: 36246378 PMCID: PMC9557131 DOI: 10.3389/fbioe.2022.991855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
The stem cell niche comprises soluble molecules and extracellular matrix components which provide chemical and mechanical cues that determine the differentiation of stem cells. Here, the effect of polyelectrolyte multilayer (PEM) composition and terminal layer fabricated with layer-by-layer technique (LBL) pairing either hyaluronan [in its native (nHA) and oxidized form (oHA)] or chondroitin sulfate (CS) with type I collagen (Col I) is investigated on chondrogenic differentiation of human umbilical mesenchymal stem cells (hUC-MSCs). Physical studies performed to investigate the establishment and structure of the surface coatings show that PEM composed of HA and Col I show a dominance of nHA or oHA with considerably lesser organization of Col I fibrils. In contrast, distinguished fibrilized Col I is found in nCS-containing PEM. Generally, Col I-terminated PEM promote the adhesion, migration, and growth of hUC-MSCs more than GAG-terminated surfaces due to the presence of fibrillar Col I but show a lower degree of differentiation towards the chondrogenic lineage. Notably, the Col I/nHA PEM not only supports adhesion and growth of hUC-MSCs but also significantly promotes cartilage-associated gene and protein expression as found by histochemical and molecular biology studies, which is not seen on the Col I/oHA PEM. This is related to ligation of HA to the cell receptor CD44 followed by activation of ERK/Sox9 and noncanonical TGF-β signaling-p38 pathways that depends on the molecular weight of HA as found by immune histochemical and western blotting. Hence, surface coatings on scaffolds and other implants by PEM composed of nHA and Col I may be useful for programming MSC towards cartilage regeneration.
Collapse
Affiliation(s)
- Mingyan Zhao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- *Correspondence: Mingyan Zhao, ; Thomas Groth,
| | - Xiang Gao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jinsong Wei
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Chenlin Tu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hong Zheng
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Kaipeng Jing
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Chu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wei Ye
- Department of Obstetrics and Gynecology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Thomas Groth
- Department Biomedical Materials, Institute of Pharmacy, Martin Luther University Halle Wittenberg, Halle (Saale), Germany
- *Correspondence: Mingyan Zhao, ; Thomas Groth,
| |
Collapse
|
20
|
Nakashima Y, Yoshida S, Tsukahara M. Semi-3D cultures using Laminin 221 as a coating material for human induced pluripotent stem cells. Regen Biomater 2022; 9:rbac060. [PMID: 36176714 PMCID: PMC9514851 DOI: 10.1093/rb/rbac060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/09/2022] [Accepted: 08/21/2022] [Indexed: 11/19/2022] Open
Abstract
It was previously believed that human induced pluripotent stem cells (hiPSCs) did not show adhesion to the coating material Laminin 221, which is known to have specific affinity for cardiomyocytes. In this study, we report that human mononuclear cell-derived hiPSCs, established with Sendai virus vector, form peninsular-like colonies rather than embryonic stem cell-like colonies; these peninsular-like colonies can be passaged more than 10 times after establishment. Additionally, initialization-deficient cells with residual Sendai virus vector adhered to the coating material Laminin 511 but not to Laminin 221. Therefore, the expression of undifferentiated markers tended to be higher in hiPSCs established on Laminin 221 than on Laminin 511. On Laminin 221, hiPSCs15M66 showed a semi-floating colony morphology. The expression of various markers of cell polarity was significantly lower in hiPSCs cultured on Laminin 221 than in hiPSCs cultured on Laminin 511. Furthermore, 201B7 and 15M66 hiPSCs showed 3D cardiomyocyte differentiation on Laminin 221. Thus, the coating material Laminin 221 provides semi-floating culture conditions for the establishment, culture and induced differentiation of hiPSCs.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Shinsuke Yoshida
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| | - Masayoshi Tsukahara
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto 606-8397, Japan
| |
Collapse
|
21
|
Rizzo R, Bonato A, Chansoria P, Zenobi-Wong M. Macroporous Aligned Hydrogel Microstrands for 3D Cell Guidance. ACS Biomater Sci Eng 2022; 8:3871-3882. [PMID: 35977074 DOI: 10.1021/acsbiomaterials.2c00370] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tissue engineering strongly relies on the use of hydrogels as highly hydrated 3D matrices to support the maturation of laden cells. However, because of the lack of microarchitecture and sufficient porosity, common hydrogel systems do not provide physical cell-instructive guidance cues and efficient transport of nutrients and oxygen to the inner part of the construct. A controlled, organized cellular alignment and resulting alignment of secreted ECM are hallmarks of muscle, tendons, and nerves and play an important role in determining their functional properties. Although several strategies to induce cellular alignment have been investigated in 2D systems, the generation of cell-instructive 3D hydrogels remains a challenge. Here, we report on the development of a simple and scalable method to efficiently generate highly macroporous constructs featuring aligned guidance cues. A precross-linked bulk hydrogel is pressed through a grid with variable opening sizes, thus deconstructing it into an array of aligned, high aspect ratio microgels (microstrands) with tunable diameter that are eventually stabilized by a second photoclick cross-linking step. This method has been investigated and optimized both in silico and in vitro, thereby leading to conditions with excellent viability and organized cellular alignment. Finally, as proof of concept, the method has been shown to direct aligned muscle tissue maturation. These findings demonstrate the 3D physical guidance potential of our system, which can be used for a variety of anisotropic tissues and applications.
Collapse
Affiliation(s)
- Riccardo Rizzo
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich 8093, Switzerland
| | - Angela Bonato
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich 8093, Switzerland
| | - Parth Chansoria
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich 8093, Switzerland
| | - Marcy Zenobi-Wong
- Tissue Engineering + Biofabrication Laboratory, Department of Health Sciences and Technology, ETH Zürich, Otto-Stern-Weg 7, Zürich 8093, Switzerland
| |
Collapse
|
22
|
Zhang Y, Habibovic P. Delivering Mechanical Stimulation to Cells: State of the Art in Materials and Devices Design. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2110267. [PMID: 35385176 DOI: 10.1002/adma.202110267] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/19/2022] [Indexed: 06/14/2023]
Abstract
Biochemical signals, such as growth factors, cytokines, and transcription factors are known to play a crucial role in regulating a variety of cellular activities as well as maintaining the normal function of different tissues and organs. If the biochemical signals are assumed to be one side of the coin, the other side comprises biophysical cues. There is growing evidence showing that biophysical signals, and in particular mechanical cues, also play an important role in different stages of human life ranging from morphogenesis during embryonic development to maturation and maintenance of tissue and organ function throughout life. In order to investigate how mechanical signals influence cell and tissue function, tremendous efforts have been devoted to fabricating various materials and devices for delivering mechanical stimuli to cells and tissues. Here, an overview of the current state of the art in the design and development of such materials and devices is provided, with a focus on their design principles, and challenges and perspectives for future research directions are highlighted.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Instructive Biomaterials Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, Maastricht University, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, Maastricht, 6229 ER, The Netherlands
| |
Collapse
|
23
|
Dede Eren A, Lucassen AWA, Tuvshindorj U, Truckenmüller R, Giselbrecht S, Eren ED, Tas MO, Sudarsanam P, de Boer J. Cells Dynamically Adapt to Surface Geometry by Remodeling Their Focal Adhesions and Actin Cytoskeleton. Front Cell Dev Biol 2022; 10:863721. [PMID: 35721512 PMCID: PMC9203963 DOI: 10.3389/fcell.2022.863721] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/02/2022] [Indexed: 01/16/2023] Open
Abstract
Cells probe their environment and adapt their shape accordingly via the organization of focal adhesions and the actin cytoskeleton. In an earlier publication, we described the relationship between cell shape and physiology, for example, shape-induced differentiation, metabolism, and proliferation in mesenchymal stem cells and tenocytes. In this study, we investigated how these cells organize their adhesive machinery over time when exposed to microfabricated surfaces of different topographies and adhesive island geometries. We further examined the reciprocal interaction between stress fiber and focal adhesion formation by pharmacological perturbations. Our results confirm the current literature that spatial organization of adhesive sites determines the ability to form focal adhesions and stress fibers. Therefore, cells on roughened surfaces have smaller focal adhesion and fewer stress fibers. Our results further highlight the importance of integrin-mediated adhesion in the adaptive properties of cells and provide clear links to the development of bioactive materials.
Collapse
Affiliation(s)
- Aysegul Dede Eren
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Amy W. A. Lucassen
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Urandelger Tuvshindorj
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Roman Truckenmüller
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Stefan Giselbrecht
- MERLN Institute for Technology Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - E. Deniz Eren
- Laboratory of Physical Chemistry, Department of Chemical Engineering and Chemistry, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mehmet Orhan Tas
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Phanikrishna Sudarsanam
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- *Correspondence: Jan de Boer,
| |
Collapse
|
24
|
Jiao F, Xu J, Zhao Y, Ye C, Sun Q, Liu C, Huo B. Synergistic effects of fluid shear stress and adhesion morphology on the apoptosis and osteogenesis of mesenchymal stem cells. J Biomed Mater Res A 2022; 110:1636-1644. [DOI: 10.1002/jbm.a.37413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/03/2022] [Accepted: 05/13/2022] [Indexed: 12/25/2022]
Affiliation(s)
- Fei Jiao
- Biomechanics Lab, Department of Mechanics School of Aerospace Engineering, Beijing Institute of Technology Beijing People's Republic of China
| | - Jiayi Xu
- Biomechanics Lab, Department of Mechanics School of Aerospace Engineering, Beijing Institute of Technology Beijing People's Republic of China
| | - Yang Zhao
- Biomechanics Lab, Department of Mechanics School of Aerospace Engineering, Beijing Institute of Technology Beijing People's Republic of China
| | - Chongyang Ye
- Biomechanics Lab, Department of Mechanics School of Aerospace Engineering, Beijing Institute of Technology Beijing People's Republic of China
| | - Qing Sun
- Biomechanics Lab, Department of Mechanics School of Aerospace Engineering, Beijing Institute of Technology Beijing People's Republic of China
| | - Chenglin Liu
- Sports Artificial Intelligence Institute Capital University of Physical Education and Sports Beijing People's Republic of China
| | - Bo Huo
- Biomechanics Lab, Department of Mechanics School of Aerospace Engineering, Beijing Institute of Technology Beijing People's Republic of China
- Sports Artificial Intelligence Institute Capital University of Physical Education and Sports Beijing People's Republic of China
| |
Collapse
|
25
|
Cao S, Yuan Q. An update of nanotopographical surfaces in modulating stem cell fate: a narrative review. BIOMATERIALS TRANSLATIONAL 2022; 3:55-64. [PMID: 35837345 PMCID: PMC9255793 DOI: 10.12336/biomatertransl.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/06/2022] [Accepted: 03/10/2022] [Indexed: 11/20/2022]
Abstract
Stem cells have been one of the ideal sources for tissue regeneration owing to their capability of self-renewal and differentiation. In vivo, the extracellular microenvironment plays a vital role in modulating stem cell fate. When developing biomaterials for regenerative medicine, incorporating biochemical and biophysical cues to mimic extracellular matrix can enhance stem cell lineage differentiation. More specifically, modulating the stem cell fate can be achieved by controlling the nanotopographic features on synthetic surfaces. Optimization of nanotopographical features leads to desirable stem cell functions, which can maximize the effectiveness of regenerative treatment. In this review, nanotopographical surfaces, including static patterned surface, dynamic patterned surface, and roughness are summarized, and their fabrication, as well as the impact on stem cell behaviour, are discussed. Later, the recent progress of applying nanotopographical featured biomaterials for altering different types of stem cells is presented, which directs the design and fabrication of functional biomaterial. Last, the perspective in fundamental research and for clinical application in this field is discussed.
Collapse
|
26
|
Zhou P, Qin L, Ge Z, Xie B, Huang H, He F, Ma S, Ren L, Shi J, Pei S, Dong G, Qi Y, Lan F. Design of chemically defined synthetic substrate surfaces for the in vitro maintenance of human pluripotent stem cells: A review. J Biomed Mater Res B Appl Biomater 2022; 110:1968-1990. [PMID: 35226397 DOI: 10.1002/jbm.b.35034] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 11/11/2022]
Abstract
Human pluripotent stem cells (hPSCs) have the potential of long-term self-renewal and differentiation into nearly all cell types in vitro. Prior to the downstream applications, the design of chemically defined synthetic substrates for the large-scale proliferation of quality-controlled hPSCs is critical. Although great achievements have been made, Matrigel and recombinant proteins are still widely used in the fundamental research and clinical applications. Therefore, much effort is still needed to improve the performance of synthetic substrates in the culture of hPSCs, realizing their commercial applications. In this review, we summarized the design of reported synthetic substrates and especially their limitations in terms of cell culture. Moreover, much attention was paid to the development of promising peptide displaying surfaces. Besides, the biophysical regulation of synthetic substrate surfaces as well as the three-dimensional culture systems were described.
Collapse
Affiliation(s)
- Ping Zhou
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Liying Qin
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Zhangjie Ge
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Biyao Xie
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Hongxin Huang
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Fei He
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Shengqin Ma
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Lina Ren
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Jiamin Shi
- Department of Laboratory Animal Centre, Changzhi Medical College, Changzhi, China
| | - Suying Pei
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Genxi Dong
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Yongmei Qi
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Feng Lan
- Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen Key Laboratory of Cardiovascular Disease, State Key Laboratory of Cardiovascular Disease, Shenzhen, China
| |
Collapse
|
27
|
Carthew J, Taylor JBJ, Garcia-Cruz MR, Kiaie N, Voelcker NH, Cadarso VJ, Frith JE. The Bumpy Road to Stem Cell Therapies: Rational Design of Surface Topographies to Dictate Stem Cell Mechanotransduction and Fate. ACS APPLIED MATERIALS & INTERFACES 2022; 14:23066-23101. [PMID: 35192344 DOI: 10.1021/acsami.1c22109] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cells sense and respond to a variety of physical cues from their surrounding microenvironment, and these are interpreted through mechanotransductive processes to inform their behavior. These mechanisms have particular relevance to stem cells, where control of stem cell proliferation, potency, and differentiation is key to their successful application in regenerative medicine. It is increasingly recognized that surface micro- and nanotopographies influence stem cell behavior and may represent a powerful tool with which to direct the morphology and fate of stem cells. Current progress toward this goal has been driven by combined advances in fabrication technologies and cell biology. Here, the capacity to generate precisely defined micro- and nanoscale topographies has facilitated the studies that provide knowledge of the mechanotransducive processes that govern the cellular response as well as knowledge of the specific features that can drive cells toward a defined differentiation outcome. However, the path forward is not fully defined, and the "bumpy road" that lays ahead must be crossed before the full potential of these approaches can be fully exploited. This review focuses on the challenges and opportunities in applying micro- and nanotopographies to dictate stem cell fate for regenerative medicine. Here, key techniques used to produce topographic features are reviewed, such as photolithography, block copolymer lithography, electron beam lithography, nanoimprint lithography, soft lithography, scanning probe lithography, colloidal lithography, electrospinning, and surface roughening, alongside their advantages and disadvantages. The biological impacts of surface topographies are then discussed, including the current understanding of the mechanotransductive mechanisms by which these cues are interpreted by the cells, as well as the specific effects of surface topographies on cell differentiation and fate. Finally, considerations in translating these technologies and their future prospects are evaluated.
Collapse
Affiliation(s)
- James Carthew
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Jason B J Taylor
- Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Maria R Garcia-Cruz
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Nasim Kiaie
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Nicolas H Voelcker
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
- ARC Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, Victoria 3800, Australia
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3168, Australia
| | - Victor J Cadarso
- Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria 3800, Australia
- Centre to Impact Antimicrobial Resistance, Monash University, Clayton, Victoria 3800, Australia
| | - Jessica E Frith
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
- ARC Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, Victoria 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
28
|
Vermeulen S, Birgani ZT, Habibovic P. Biomaterial-induced pathway modulation for bone regeneration. Biomaterials 2022; 283:121431. [DOI: 10.1016/j.biomaterials.2022.121431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022]
|
29
|
The spatial self-organization within pluripotent stem cell colonies is continued in detaching aggregates. Biomaterials 2022; 282:121389. [PMID: 35121357 DOI: 10.1016/j.biomaterials.2022.121389] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/13/2021] [Accepted: 01/23/2022] [Indexed: 12/13/2022]
Abstract
Colonies of induced pluripotent stem cells (iPSCs) reveal aspects of self-organization even under culture conditions that maintain pluripotency. To investigate the dynamics of this process under spatial confinement, we used either polydimethylsiloxane (PDMS) pillars or micro-contact printing of vitronectin. There was a progressive upregulation of OCT4, E-cadherin, and NANOG within 70 μm from the outer rim of iPSC colonies. Single-cell RNA-sequencing and spatial reconstruction of gene expression demonstrated that OCT4high subsets, residing at the edge of the colony, have pronounced up-regulation of the TGF-β pathway, particularly of NODAL and its inhibitor LEFTY. Interestingly, after 5-7 days, iPSC colonies detached spontaneously from micro-contact printed substrates to form 3D aggregates. This new method allowed generation of embryoid bodies (EBs) of controlled size without enzymatic or mechanical treatment. Within the early 3D aggregates, radial organization and differential gene expression continued in analogy to the changes observed during self-organization of iPSC colonies. Early self-detached aggregates revealed up-regulated germline-specific gene expression patterns as compared to conventional EBs. However, there were no marked differences after further directed differentiation toward hematopoietic, mesenchymal, and neuronal lineages. Our results provide further insight into the gradual self-organization within iPSC colonies and at their transition into EBs.
Collapse
|
30
|
Lee G, Cho Y, Kim EH, Choi JM, Chae SS, Lee MG, Kim J, Choi WJ, Kwon J, Han EH, Kim SH, Park S, Chung YH, Chi SG, Jung BH, Shin JH, Lee JO. Pillar-Based Mechanical Induction of an Aggressive Tumorigenic Lung Cancer Cell Model. ACS APPLIED MATERIALS & INTERFACES 2022; 14:20-31. [PMID: 34914354 DOI: 10.1021/acsami.1c12380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tissue microarchitecture imposes physical constraints to the migration of individual cells. Especially in cancer metastasis, three-dimensional structural barriers within the extracellular matrix are known to affect the migratory behavior of cells, regulating the pathological state of the cells. Here, we employed a culture platform with micropillar arrays of 2 μm diameter and 16 μm pitch (2.16 micropillar) as a mechanical stimulant. Using this platform, we investigated how a long-term culture of A549 human lung carcinoma cells on the (2.16) micropillar-embossed dishes would influence the pathological state of the cell. A549 cells grown on the (2.16) micropillar array with 10 μm height exhibited a significantly elongated morphology and enhanced migration even after the detachment and reattachment, as evidenced in the conventional wound-healing assay, single-cell tracking analysis, and in vivo tumor colonization assays. Moreover, the pillar-induced morphological deformation in nuclei was accompanied by cell-cycle arrest in the S phase, leading to suppressed proliferation. While these marked traits of morphology-migration-proliferation support more aggressive characteristics of metastatic cancer cells, typical indices of epithelial-mesenchymal transition were not found, but instead, remarkable traces of amoeboidal transition were confirmed. Our study also emphasizes the importance of mechanical stimuli from the microenvironment during pathogenesis and how gained traits can be passed onto subsequent generations, ultimately affecting their pathophysiological behavior. Furthermore, this study highlights the potential use of pillar-based mechanical stimuli as an in vitro cell culture strategy to induce more aggressive tumorigenic cancer cell models.
Collapse
Affiliation(s)
- Geonhee Lee
- Advanced Materials Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Youngbin Cho
- Department of Mechanical Engineering, Korea Advanced of Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Eun Hye Kim
- Laboratory of Translational Therapeutics, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Jong Min Choi
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Soo Sang Chae
- Advanced Materials Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Min-Goo Lee
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Jonghyun Kim
- Division of Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Won Jin Choi
- Advanced Materials Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Joseph Kwon
- Division of Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Eun Hee Han
- Division of Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Seong Hwan Kim
- Laboratory of Translational Therapeutics, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Young-Ho Chung
- Division of Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Sung-Gil Chi
- Department of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Byung Hwa Jung
- Molecular Recognition Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Jennifer H Shin
- Department of Mechanical Engineering, Korea Advanced of Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jeong-O Lee
- Advanced Materials Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| |
Collapse
|
31
|
Bian Q, Chen J, Weng Y, Li S. Endothelialization strategy of implant materials surface: The newest research in recent 5 years. J Appl Biomater Funct Mater 2022; 20:22808000221105332. [PMID: 35666145 DOI: 10.1177/22808000221105332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In recent years, more and more metal or non-metal materials have been used in the treatment of cardiovascular diseases, but the vascular complications after transplantation are still the main factors restricting the clinical application of most grafts, such as acute thrombosis and graft restenosis. Implant materials have been extensively designed and surface optimized by researchers, but it is still too difficult to avoid complications. Natural vascular endodermis has excellent function, anti-coagulant and anti-intimal hyperplasia, and it is also the key to maintaining the homeostasis of normal vascular microenvironment. Therefore, how to promote the adhesion of endothelial cells (ECs) on the surface of cardiovascular materials to achieve endothelialization of the surface is the key to overcoming the complications after implant materialization. At present, the surface endothelialization design of materials based on materials surface science, bioactive molecules, and biological function intervention and feedback has attracted much attention. In this review, we summarize the related research on the surface modification of materials by endothelialization in recent years, and analyze the advantages and challenges of current endothelialization design ideas, explain the relationship between materials, cells, and vascular remodeling in order to find a more ideal endothelialization surface modification strategy for future researchers to meet the requirements of clinical biocompatibility of cardiovascular materials.
Collapse
Affiliation(s)
- Qihao Bian
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, China.,School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Junying Chen
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, China.,School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Yajun Weng
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, China.,School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Suiyan Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
32
|
Nazbar A, Samani S, Yazdian Kashani S, Amanzadeh A, Shoeibi S, Bonakdar S. Molecular imprinting as a simple way for the long-term maintenance of the stemness and proliferation potential of adipose-derived stem cells: an in vitro study. J Mater Chem B 2022; 10:6816-6830. [DOI: 10.1039/d2tb00279e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Culturing adipose-derived stem cells (ADSCs) on the biomimetic ADSC-imprinted substrate is a simple way for long-term maintenance of their stemness and proliferation potential.
Collapse
Affiliation(s)
- Abolfazl Nazbar
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Saeed Samani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Yazdian Kashani
- Department of Chemical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Amir Amanzadeh
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| | - Shahram Shoeibi
- Food and Drug Laboratory Research Center (FDLRC), Iran Food and Drug Administration (IFDA), MOH & ME, Tehran, Iran
| | - Shahin Bonakdar
- National Cell Bank Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
33
|
Chalmantzi V, Simitzi C, Papadopoulos A, Bagli E, Murphy C, Stratakis E, Fotsis T. Culturing Human Pluripotent Stem Cells on Micropatterned Silicon Surfaces. Methods Mol Biol 2022; 2454:49-59. [PMID: 34907510 DOI: 10.1007/7651_2021_428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Human pluripotent stem cell culture conditions are constantly being optimized, thus providing insight to the environmental cues that affect cell choices. A wide variety of media, coating materials, and substrates is now available for use, serving different scientific needs. Factors such as material stiffness, roughness, and topography are being recognized to contribute or even direct the acquisition of specific phenotypes. Here, we describe the use of patterned silicon substrates coated with Matrigel for the propagation and differentiation of human pluripotent stem cells.
Collapse
Affiliation(s)
- Varvara Chalmantzi
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
- Department of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, Ioannina, Greece
| | - Chara Simitzi
- Institute of Electronic Structure and Laser (IESL), Foundation of Research and Technology Hellas (FORTH), Vassilika Vouton, Heraklion, Greece
- Institute of Materials Discovery, University College London, London, UK
| | - Angelos Papadopoulos
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
- Department of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, Ioannina, Greece
| | - Eleni Bagli
- Department of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, Ioannina, Greece
| | - Carol Murphy
- School of Biosciences, College of Life and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
- Department of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, Ioannina, Greece
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser (IESL), Foundation of Research and Technology Hellas (FORTH), Vassilika Vouton, Heraklion, Greece.
- Materials Science and Technology Department, University of Crete, Vassilika Voutes, Heraklion, Greece.
| | - Theodore Fotsis
- Department of Biomedical Research, Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, Ioannina, Greece.
- Laboratory of Biological Chemistry, Medical School, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
34
|
Daga KR, Priyadarshani P, Larey AM, Rui K, Mortensen LJ, Marklein RA. Shape up before you ship out: morphology as a potential critical quality attribute for cellular therapies. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
35
|
Petzold J, Gentleman E. Intrinsic Mechanical Cues and Their Impact on Stem Cells and Embryogenesis. Front Cell Dev Biol 2021; 9:761871. [PMID: 34820380 PMCID: PMC8606660 DOI: 10.3389/fcell.2021.761871] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
Although understanding how soluble cues direct cellular processes revolutionised the study of cell biology in the second half of the 20th century, over the last two decades, new insights into how mechanical cues similarly impact cell fate decisions has gained momentum. During development, extrinsic cues such as fluid flow, shear stress and compressive forces are essential for normal embryogenesis to proceed. Indeed, both adult and embryonic stem cells can respond to applied forces, but they can also detect intrinsic mechanical cues from their surrounding environment, such as the stiffness of the extracellular matrix, which impacts differentiation and morphogenesis. Cells can detect changes in their mechanical environment using cell surface receptors such as integrins and focal adhesions. Moreover, dynamic rearrangements of the cytoskeleton have been identified as a key means by which forces are transmitted from the extracellular matrix to the cell and vice versa. Although we have some understanding of the downstream mechanisms whereby mechanical cues are translated into changes in cell behaviour, many of the signalling pathways remain to be defined. This review discusses the importance of intrinsic mechanical cues on adult cell fate decisions, the emerging roles of cell surface mechano-sensors and the cytoskeleton in enabling cells to sense its microenvironment, and the role of intracellular signalling in translating mechanical cues into transcriptional outputs. In addition, the contribution of mechanical cues to fundamental processes during embryogenesis such as apical constriction and convergent extension is discussed. The continued development of tools to measure the biomechanical properties of soft tissues in vivo is likely to uncover currently underestimated contributions of these cues to adult stem cell fate decisions and embryogenesis, and may inform on regenerative strategies for tissue repair.
Collapse
Affiliation(s)
- Jonna Petzold
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
36
|
Zhang Y, Wang X, Zhang Y, Liu Y, Wang D, Yu X, Wang H, Bai Z, Jiang YC, Li X, Zheng W, Li Q. Endothelial Cell Migration Regulated by Surface Topography of Poly(ε-caprolactone) Nanofibers. ACS Biomater Sci Eng 2021; 7:4959-4970. [PMID: 34543012 DOI: 10.1021/acsbiomaterials.1c00951] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The study of cell migration on biomaterials is of great significance in tissue engineering and regenerative medicine. In recent years, there has been increasing evidence that the physical properties of the extracellular matrix (ECM), such as surface topography, affect various cellular behaviors such as proliferation, adhesion, and migration. However, the biological mechanism of surface topography influencing cellular behavior is still unclear. In this study, we prepared polycaprolactone (PCL) fibrous materials with different surface microstructures by solvent casting, electrospinning, and self-induced crystallization. The corresponding topographical structure obtained is a two-dimensional (2D) flat surface, 2.5-dimensional (2.5D) fibers, and three-dimensional (3D) fibers with a multilevel microstructure. We then investigated the effects of the complex topographical structure on endothelial cell migration. Our study demonstrates that cells can sense the changes of micro- and nanomorphology on the surface of materials, adapt to the physical environment through biochemical reactions, and regulate actin polymerization and directional migration through Rac1 and Cdc42. The cells on the nanofibers are elongated spindles, and the positive feedback of cell adhesion and actin polymerization along the fiber direction makes the plasma membrane continue to protrude, promoting cell polarization and directional migration. This study might provide new insights into the biomaterial design, especially those used for artificial vascular grafts.
Collapse
Affiliation(s)
- Yang Zhang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yan Zhang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yajing Liu
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Dongfang Wang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xueke Yu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Haonan Wang
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Zhiyuan Bai
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Yong-Chao Jiang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Wei Zheng
- Engineering and Technology Department, University of Wisconsin-STOUT, Menomonie, Wisconsin 54751, United States
| | - Qian Li
- School of Mechanics and Safety Engineering, National Center for International Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
37
|
A blueprint of the topology and mechanics of the human ovary for next-generation bioengineering and diagnosis. Nat Commun 2021; 12:5603. [PMID: 34556652 PMCID: PMC8460685 DOI: 10.1038/s41467-021-25934-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 09/07/2021] [Indexed: 02/06/2023] Open
Abstract
Although the first dissection of the human ovary dates back to the 17th century, the biophysical characteristics of the ovarian cell microenvironment are still poorly understood. However, this information is vital to deciphering cellular processes such as proliferation, morphology and differentiation, as well as pathologies like tumor progression, as demonstrated in other biological tissues. Here, we provide the first readout of human ovarian fiber morphology, interstitial and perifollicular fiber orientation, pore geometry, topography and surface roughness, and elastic and viscoelastic properties. By determining differences between healthy prepubertal, reproductive-age, and menopausal ovarian tissue, we unravel and elucidate a unique biophysical phenotype of reproductive-age tissue, bridging biophysics and female fertility. While these data enable to design of more biomimetic scaffolds for the tissue-engineered ovary, our analysis pipeline is applicable for the characterization of other organs in physiological or pathological states to reveal their biophysical markers or design their bioinspired analogs. Although the first dissection of the human ovary dates back to the 17th century, its characterization is still limited. Here, the authors have unraveled a unique biophysical and topological phenotype of reproductive-age tissue, bridging biophysics and female fertility and providing a blueprint for the artificial ovary.
Collapse
|
38
|
Riegert J, Töpel A, Schieren J, Coryn R, Dibenedetto S, Braunmiller D, Zajt K, Schalla C, Rütten S, Zenke M, Pich A, Sechi A. Guiding cell adhesion and motility by modulating cross-linking and topographic properties of microgel arrays. PLoS One 2021; 16:e0257495. [PMID: 34555082 PMCID: PMC8460069 DOI: 10.1371/journal.pone.0257495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Biomaterial-driven modulation of cell adhesion and migration is a challenging aspect of tissue engineering. Here, we investigated the impact of surface-bound microgel arrays with variable geometry and adjustable cross-linking properties on cell adhesion and migration. We show that cell migration is inversely correlated with microgel array spacing, whereas directionality increases as array spacing increases. Focal adhesion dynamics is also modulated by microgel topography resulting in less dynamic focal adhesions on surface-bound microgels. Microgels also modulate the motility and adhesion of Sertoli cells used as a model for cell migration and adhesion. Both focal adhesion dynamics and speed are reduced on microgels. Interestingly, Gas2L1, a component of the cytoskeleton that mediates the interaction between microtubules and microfilaments, is dispensable for the regulation of cell adhesion and migration on microgels. Finally, increasing microgel cross-linking causes a clear reduction of focal adhesion turnover in Sertoli cells. These findings not only show that spacing and rigidity of surface-grafted microgels arrays can be effectively used to modulate cell adhesion and motility of diverse cellular systems, but they also form the basis for future developments in the fields of medicine and tissue engineering.
Collapse
Affiliation(s)
- Janine Riegert
- Dept. of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen
University, Aachen, Germany
| | - Alexander Töpel
- Functional and Interactive Polymers, Institute of Technical and
Macromolecular Chemistry, RWTH Aachen University, Aachen,
Germany
- DWI, Leibniz Institute for Interactive Materials e.V., Aachen,
Germany
| | - Jana Schieren
- Dept. of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen
University, Aachen, Germany
| | - Renee Coryn
- Dept. of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen
University, Aachen, Germany
| | - Stella Dibenedetto
- Dept. of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen
University, Aachen, Germany
| | - Dominik Braunmiller
- Functional and Interactive Polymers, Institute of Technical and
Macromolecular Chemistry, RWTH Aachen University, Aachen,
Germany
- DWI, Leibniz Institute for Interactive Materials e.V., Aachen,
Germany
| | - Kamil Zajt
- Dept. of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen
University, Aachen, Germany
| | - Carmen Schalla
- Dept. of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen
University, Aachen, Germany
| | - Stephan Rütten
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen
University, Aachen, Germany
| | - Martin Zenke
- Dept. of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen
University, Aachen, Germany
| | - Andrij Pich
- Functional and Interactive Polymers, Institute of Technical and
Macromolecular Chemistry, RWTH Aachen University, Aachen,
Germany
- DWI, Leibniz Institute for Interactive Materials e.V., Aachen,
Germany
| | - Antonio Sechi
- Dept. of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen
University, Aachen, Germany
| |
Collapse
|
39
|
Tayler IM, Stowers RS. Engineering hydrogels for personalized disease modeling and regenerative medicine. Acta Biomater 2021; 132:4-22. [PMID: 33882354 DOI: 10.1016/j.actbio.2021.04.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/26/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
Technological innovations and advances in scientific understanding have created an environment where data can be collected, analyzed, and interpreted at scale, ushering in the era of personalized medicine. The ability to isolate cells from individual patients offers tremendous promise if those cells can be used to generate functional tissue replacements or used in disease modeling to determine optimal treatment strategies. Here, we review recent progress in the use of hydrogels to create artificial cellular microenvironments for personalized tissue engineering and regenerative medicine applications, as well as to develop personalized disease models. We highlight engineering strategies to control stem cell fate through hydrogel design, and the use of hydrogels in combination with organoids, advanced imaging methods, and novel bioprinting techniques to generate functional tissues. We also discuss the use of hydrogels to study molecular mechanisms underlying diseases and to create personalized in vitro disease models to complement existing pre-clinical models. Continued progress in the development of engineered hydrogels, in combination with other emerging technologies, will be essential to realize the immense potential of personalized medicine. STATEMENT OF SIGNIFICANCE: In this review, we cover recent advances in hydrogel engineering strategies with applications in personalized medicine. Specifically, we focus on material systems to expand or control differentiation of patient-derived stem cells, and hydrogels to reprogram somatic cells to pluripotent states. We then review applications of hydrogels in developing personalized engineered tissues. We also highlight the use of hydrogel systems as personalized disease models, focusing on specific examples in fibrosis and cancer, and more broadly on drug screening strategies using patient-derived cells and hydrogels. We believe this review will be a valuable contribution to the Special Issue and the readership of Acta Biomaterialia will appreciate the comprehensive overview of the utility of hydrogels in the developing field of personalized medicine.
Collapse
|
40
|
Jin Y, Cho SW. Bioengineering platforms for cell therapeutics derived from pluripotent and direct reprogramming. APL Bioeng 2021; 5:031501. [PMID: 34258498 PMCID: PMC8263070 DOI: 10.1063/5.0040621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
Pluripotent and direct reprogramming technologies hold great potential for tissue repair and restoration of tissue and organ function. The implementation of induced pluripotent stem cells and directly reprogrammed cells in biomedical research has resulted in a significant leap forward in the highly promising area of regenerative medicine. While these therapeutic strategies are promising, there are several obstacles to overcome prior to the introduction of these therapies into clinical settings. Bioengineering technologies, such as biomaterials, bioprinting, microfluidic devices, and biostimulatory systems, can enhance cell viability, differentiation, and function, in turn the efficacy of cell therapeutics generated via pluripotent and direct reprogramming. Therefore, cellular reprogramming technologies, in combination with tissue-engineering platforms, are poised to overcome current bottlenecks associated with cell-based therapies and create new ways of producing engineered tissue substitutes.
Collapse
Affiliation(s)
- Yoonhee Jin
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | | |
Collapse
|
41
|
Zhang X, van Rijt S. 2D biointerfaces to study stem cell-ligand interactions. Acta Biomater 2021; 131:80-96. [PMID: 34237424 DOI: 10.1016/j.actbio.2021.06.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023]
Abstract
Stem cells have great potential in the field of tissue engineering and regenerative medicine due to their inherent regenerative capabilities. However, an ongoing challenge within their clinical translation is to elicit or predict the desired stem cell behavior once transplanted. Stem cell behavior and function are regulated by their interaction with biophysical and biochemical signals present in their natural environment (i.e., stem cell niches). To increase our understanding about the interplay between stem cells and their resident microenvironments, biointerfaces have been developed as tools to study how these substrates can affect stem cell behaviors. This article aims to review recent developments on fabricating cell-instructive interfaces to control cell adhesion processes towards directing stem cell behavior. After an introduction on stem cells and their natural environment, static surfaces exhibiting predefined biochemical signals to probe the effect of chemical features on stem cell behaviors are discussed. In the third section, we discuss more complex dynamic platforms able to display biochemical cues with spatiotemporal control using on-off ligand display, reversible ligand display, and ligand mobility. In the last part of the review, we provide the reader with an outlook on future designs of biointerfaces. STATEMENT OF SIGNIFICANCE: Stem cells have great potential as treatments for many degenerative disorders prevalent in our aging societies. However, an ongoing challenge within their clinical translation is to promote stem cell mediated regeneration once they are transplanted in the body. Stem cells reside within our bodies where their behavior and function are regulated by interactions with their natural environment called the stem cell niche. To increase our understanding about the interplay between stem cells and their niche, 2D materials have been developed as tools to study how specific signals can affect stem cell behaviors. This article aims to review recent developments on fabricating cell-instructive interfaces to control cell adhesion processes towards directing stem cell behavior.
Collapse
|
42
|
Martínez A, González-Lana S, Asín L, de la Fuente JM, Bastiaansen CWM, Broer DJ, Sánchez-Somolinos C. Nano-Second Laser Interference Photoembossed Microstructures for Enhanced Cell Alignment. Polymers (Basel) 2021; 13:polym13172958. [PMID: 34502998 PMCID: PMC8434024 DOI: 10.3390/polym13172958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 11/16/2022] Open
Abstract
Photoembossing is a powerful photolithographic technique to prepare surface relief structures relying on polymerization-induced diffusion in a solventless development step. Conveniently, surface patterns are formed by two or more interfering laser beams without the need for a lithographic mask. The use of nanosecond pulsed light-based interference lithography strengthens the pattern resolution through the absence of vibrational line pattern distortions. Typically, a conventional photoembossing protocol consists of an exposure step at room temperature that is followed by a thermal development step at high temperature. In this work, we explore the possibility to perform the pulsed holographic exposure directly at the development temperature. The surface relief structures generated using this modified photoembossing protocol are compared with those generated using the conventional one. Importantly, the enhancement of surface relief height has been observed by exposing the samples directly at the development temperature, reaching approximately double relief heights when compared to samples obtained using the conventional protocol. Advantageously, the light dose needed to reach the optimum height and the amount of photoinitiator can be substantially reduced in this modified protocol, demonstrating it to be a more efficient process for surface relief generation in photopolymers. Kidney epithelial cell alignment studies on substrates with relief-height optimized structures generated using the two described protocols demonstrate improved cell alignment in samples generated with exposure directly at the development temperature, highlighting the relevance of the height enhancement reached by this method. Although cell alignment is well-known to be enhanced by increasing the relief height of the polymeric grating, our work demonstrates nano-second laser interference photoembossing as a powerful tool to easily prepare polymeric gratings with tunable topography in the range of interest for fundamental cell alignment studies.
Collapse
Affiliation(s)
- Alba Martínez
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Advanced Manufacturing Laboratory, Departamento de Física de la Materia Condensada, C./Pedro Cerbuna 12, 50009 Zaragoza, Spain; (A.M.); (S.G.-L.)
| | - Sandra González-Lana
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Advanced Manufacturing Laboratory, Departamento de Física de la Materia Condensada, C./Pedro Cerbuna 12, 50009 Zaragoza, Spain; (A.M.); (S.G.-L.)
- BEONCHIP S.L., CEMINEM, Campus Rio Ebro. C./Mariano Esquillor Gómez s/n, 50018 Zaragoza, Spain
| | - Laura Asín
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, C./Pedro Cerbuna 12, 50009 Zaragoza, Spain; (L.A.); (J.M.d.l.F.)
- CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Jesús M. de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, C./Pedro Cerbuna 12, 50009 Zaragoza, Spain; (L.A.); (J.M.d.l.F.)
- CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Cees W. M. Bastiaansen
- Faculty of Chemistry and Chemical Engineering, Eindhoven University, P.O. Box 513, 5600 Eindhoven, The Netherlands; (C.W.M.B.); (D.J.B.)
| | - Dirk J. Broer
- Faculty of Chemistry and Chemical Engineering, Eindhoven University, P.O. Box 513, 5600 Eindhoven, The Netherlands; (C.W.M.B.); (D.J.B.)
| | - Carlos Sánchez-Somolinos
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, Advanced Manufacturing Laboratory, Departamento de Física de la Materia Condensada, C./Pedro Cerbuna 12, 50009 Zaragoza, Spain; (A.M.); (S.G.-L.)
- CIBER in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
- Correspondence:
| |
Collapse
|
43
|
Panwar V, Babu A, Sharma A, Thomas J, Chopra V, Malik P, Rajput S, Mittal M, Guha R, Chattopadhyay N, Mandal D, Ghosh D. Tunable, conductive, self-healing, adhesive and injectable hydrogels for bioelectronics and tissue regeneration applications. J Mater Chem B 2021; 9:6260-6270. [PMID: 34338263 DOI: 10.1039/d1tb01075a] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Conductive hydrogels are attracting considerable interest in view of their potential in a wide range of applications that include healthcare and electronics. Such hydrogels are generally incorporated with conductive materials/polymers. Herein, we present a series of conductive hydrogels (Ch-CMC-PDA), prepared with no additional conductive material. The hydrogels were synthesized using a combination of chitosan, cellulose (CMC) and dopamine (DA). The conductivity (0.01-3.4 × 10-3 S cm-1) in these gels is attributed to ionic conductivity. Very few conductive hydrogels are endowed with additional properties like injectability, adhesiveness and self-healing, which would help to widen their scope for applications. While the dynamic Schiff base coupling in our hydrogels facilitated self-healing and injectable properties, polydopamine imparted tissue adhesiveness. The porosity, rheological, mechanical and conductive properties of the hydrogels are regulated by the CMC-dialdehyde-polydopamine (CMC-D-PDA) content. The hydrogel was evaluated in various bioelectronics applications like ECG monitoring and triboelectric nanogenerators (TENG). The ability of the hydrogel to support cell growth and serve as a template for tissue regeneration was confirmed using in vitro and in vivo studies. In summary, the integration of such remarkable features in the ionic-conductive hydrogel would enable its usage in bioelectronics and biomedical applications.
Collapse
Affiliation(s)
- Vineeta Panwar
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector-81, Mohali-140306, Punjab, India.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Vermeulen S, Honig F, Vasilevich A, Roumans N, Romero M, Dede Eren A, Tuvshindorj U, Alexander M, Carlier A, Williams P, Uquillas J, de Boer J. Expanding Biomaterial Surface Topographical Design Space through Natural Surface Reproduction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102084. [PMID: 34165820 PMCID: PMC11468538 DOI: 10.1002/adma.202102084] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/15/2021] [Indexed: 06/13/2023]
Abstract
Surface topography is a tool to endow biomaterials with bioactive properties. However, the large number of possible designs makes it challenging to find the optimal surface structure to induce a specific cell response. The TopoChip platform is currently the largest collection of topographies with 2176 in silico designed microtopographies. Still, it is exploring only a small part of the design space due to design algorithm limitations and the surface engineering strategy. Inspired by the diversity of natural surfaces, it is assessed as to what extent the topographical design space and consequently the resulting cellular responses can be expanded using natural surfaces. To this end, 26 plant and insect surfaces are replicated in polystyrene and their surface properties are quantified using white light interferometry. Through machine-learning algorithms, it is demonstrated that natural surfaces extend the design space of the TopoChip, which coincides with distinct morphological and focal adhesion profiles in mesenchymal stem cells (MSCs) and Pseudomonas aeruginosa colonization. Furthermore, differentiation experiments reveal the strong potential of the holy lotus to improve osteogenesis in MSCs. In the future, the design algorithms will be trained with the results obtained by natural surface imprint experiments to explore the bioactive properties of novel surface topographies.
Collapse
Affiliation(s)
- Steven Vermeulen
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Floris Honig
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Aliaksei Vasilevich
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Nadia Roumans
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Manuel Romero
- National Biofilms Innovation CentreBiodiscovery Institute and School of Life SciencesUniversity of NottinghamNottinghamNG7 2RDUK
| | - Aysegul Dede Eren
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Urnaa Tuvshindorj
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Morgan Alexander
- Advanced Materials and Healthcare TechnologiesThe School of PharmacyUniversity of NottinghamNottinghamNG7 2RDUK
| | - Aurélie Carlier
- MERLN InstituteMaastricht UniversityMaastricht6229 ERThe Netherlands
| | - Paul Williams
- National Biofilms Innovation CentreBiodiscovery Institute and School of Life SciencesUniversity of NottinghamNottinghamNG7 2RDUK
| | - Jorge Uquillas
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| | - Jan de Boer
- Department of Biomedical Engineering and Institute for Complex Molecular SystemsEindhoven University of TechnologyEindhoven5600 MBThe Netherlands
| |
Collapse
|
45
|
Soheilmoghaddam F, Rumble M, Cooper-White J. High-Throughput Routes to Biomaterials Discovery. Chem Rev 2021; 121:10792-10864. [PMID: 34213880 DOI: 10.1021/acs.chemrev.0c01026] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many existing clinical treatments are limited in their ability to completely restore decreased or lost tissue and organ function, an unenviable situation only further exacerbated by a globally aging population. As a result, the demand for new medical interventions has increased substantially over the past 20 years, with the burgeoning fields of gene therapy, tissue engineering, and regenerative medicine showing promise to offer solutions for full repair or replacement of damaged or aging tissues. Success in these fields, however, inherently relies on biomaterials that are engendered with the ability to provide the necessary biological cues mimicking native extracellular matrixes that support cell fate. Accelerating the development of such "directive" biomaterials requires a shift in current design practices toward those that enable rapid synthesis and characterization of polymeric materials and the coupling of these processes with techniques that enable similarly rapid quantification and optimization of the interactions between these new material systems and target cells and tissues. This manuscript reviews recent advances in combinatorial and high-throughput (HT) technologies applied to polymeric biomaterial synthesis, fabrication, and chemical, physical, and biological screening with targeted end-point applications in the fields of gene therapy, tissue engineering, and regenerative medicine. Limitations of, and future opportunities for, the further application of these research tools and methodologies are also discussed.
Collapse
Affiliation(s)
- Farhad Soheilmoghaddam
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Madeleine Rumble
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| | - Justin Cooper-White
- Tissue Engineering and Microfluidics Laboratory (TEaM), Australian Institute for Bioengineering and Nanotechnology (AIBN), University Of Queensland, St. Lucia, Queensland, Australia 4072.,School of Chemical Engineering, University Of Queensland, St. Lucia, Queensland, Australia 4072
| |
Collapse
|
46
|
Kharbikar BN, Chendke GS, Desai TA. Modulating the foreign body response of implants for diabetes treatment. Adv Drug Deliv Rev 2021; 174:87-113. [PMID: 33484736 PMCID: PMC8217111 DOI: 10.1016/j.addr.2021.01.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/30/2020] [Accepted: 01/10/2021] [Indexed: 02/06/2023]
Abstract
Diabetes Mellitus is a group of diseases characterized by high blood glucose levels due to patients' inability to produce sufficient insulin. Current interventions often require implants that can detect and correct high blood glucose levels with minimal patient intervention. However, these implantable technologies have not reached their full potential in vivo due to the foreign body response and subsequent development of fibrosis. Therefore, for long-term function of implants, modulating the initial immune response is crucial in preventing the activation and progression of the immune cascade. This review discusses the different molecular mechanisms and cellular interactions involved in the activation and progression of foreign body response (FBR) and fibrosis, specifically for implants used in diabetes. We also highlight the various strategies and techniques that have been used for immunomodulation and prevention of fibrosis. We investigate how these general strategies have been applied to implants used for the treatment of diabetes, offering insights on how these devices can be further modified to circumvent FBR and fibrosis.
Collapse
Affiliation(s)
- Bhushan N Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gauree S Chendke
- University of California Berkeley - University of California San Francisco Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94143, USA; University of California Berkeley - University of California San Francisco Graduate Program in Bioengineering, San Francisco, CA 94143, USA; Department of Bioengineering, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
47
|
Lomboni DJ, Steeves A, Schock S, Bonetti L, De Nardo L, Variola F. Compounded topographical and physicochemical cueing by micro-engineered chitosan substrates on rat dorsal root ganglion neurons and human mesenchymal stem cells. SOFT MATTER 2021; 17:5284-5302. [PMID: 34075927 DOI: 10.1039/d0sm02170a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Given the intertwined physicochemical effects exerted in vivo by both natural and synthetic (e.g., biomaterial) interfaces on adhering cells, the evaluation of structure-function relationships governing cellular response to micro-engineered surfaces for applications in neuronal tissue engineering requires the use of in vitro testing platforms which consist of a clinically translatable material with tunable physiochemical properties. In this work, we micro-engineered chitosan substrates with arrays of parallel channels with variable width (20 and 60 μm). A citric acid (CA)-based crosslinking approach was used to provide an additional level of synergistic cueing on adhering cells by regulating the chitosan substrate's stiffness. Morphological and physicochemical characterization was conducted to unveil the structure-function relationships which govern the activity of rat dorsal root ganglion neurons (DRGs) and human mesenchymal stem cells (hMSCs), ultimately singling out the key role of microtopography, roughness and substrate's stiffness. While substrate's stiffness predominantly affected hMSC spreading, the modulation of the channels' design affected the neuronal architecture's complexity and guided the morphological transition of hMSCs. Finally, the combined analysis of tubulin expression and cell morphology allowed us to cast new light on the predominant role of the microtopography over substrate's stiffness in the process of hMSCs neurogenic differentiation.
Collapse
Affiliation(s)
- David J Lomboni
- Department of Mechanical Engineering, University of Ottawa, K1N 6N5 Canada. and Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - Alexander Steeves
- Department of Mechanical Engineering, University of Ottawa, K1N 6N5 Canada. and Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada
| | - Sarah Schock
- Department of Cellular and Molecular Medicine, University of Ottawa, Canada and The Children's Hospital of Eastern Ontario (CHEO) Research Institute, Canada
| | - Lorenzo Bonetti
- Department of Chemistry, Materials and Chemical Engineering, "G. Natta", Politecnico di Milano, Italy
| | - Luigi De Nardo
- Department of Chemistry, Materials and Chemical Engineering, "G. Natta", Politecnico di Milano, Italy
| | - Fabio Variola
- Department of Mechanical Engineering, University of Ottawa, K1N 6N5 Canada. and Ottawa-Carleton Institute for Biomedical Engineering (OCIBME), Ottawa, Canada and Department of Cellular and Molecular Medicine, University of Ottawa, Canada and The Children's Hospital of Eastern Ontario (CHEO) Research Institute, Canada
| |
Collapse
|
48
|
Sankar D, Mony U, Rangasamy J. Combinatorial effect of plasma treatment, fiber alignment and fiber scale of poly (ε-caprolactone)/collagen multiscale fibers in inducing tenogenesis in non-tenogenic media. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 127:112206. [PMID: 34225858 DOI: 10.1016/j.msec.2021.112206] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/20/2022]
Abstract
Tendon being a hypocellular, low vascularized tissue often requires assistance for restoration after complete tear. Tendon tissue engineering aims in the development of suitable scaffold that could support the regeneration of tendon after damage. The success of such scaffolds is dependent on its integration with the native tissue which in turn is influenced by the cell-material interaction. In this work aligned poly(ε-caprolactone)/collagen (PCL/collagen) multiscale fibers were developed and plasma treatment using argon, nitrogen and its combination was accessed for inducing tenogenic differentiation in mesenchymal stem cells. The developed fibers mimicked tendon extracellular matrix (ECM) which upon plasma treatment maintained moderate hydrophilicity. Oxygen and nitrogen containing groups were observed to be incorporated after argon and nitrogen treatment respectively. Statistically significant (p < 0.001) enhancement was observed in average and root mean square (RMS) roughness after plasma treatment with the maximum in argon treated fibers. Vitronectin was competitively (statistically significant, p < 0.05) adsorbed after argon and combination treatment whereas nitrogen treatment led to the competitive adsorption of fibronectin (statistically significant, p < 0.05). Human mesenchymal stem cells (hMSCs) showed enhanced proliferation and attachment on plasma treated fibers. Increased porosity due to the presence of sacrificial collagen nanofibers improved cell infiltration which was further enhanced upon plasma treatment. RhoA activation was observed (statistically significant, p < 0.05) on aligned PCL/collagen multiscale fibers and PCL microfibers, which proved its impact on tenogenic differentiation. Further enhancement in rhoA expression was observed on argon (p < 0.01) and combination plasma (p < 0.05) treated fibers. Tenogenic differentiation of hMSCs was enhanced (statistically significant) on argon plasma treated aligned fibers which was confirmed by the expression of scleraxis, mohawk (early markers) and tenomodulin (late marker) at protein level and mohawk, collagen I, collagen III (early markers), thrombospondin 4 and tenascin C (late markers) at gene level. Thus argon plasma treatment on aligned fibers is an effective method to induce tenogenesis even in non-tenogenic media.
Collapse
Affiliation(s)
- Deepthi Sankar
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Ullas Mony
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India.
| | - Jayakumar Rangasamy
- Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi 682041, India.
| |
Collapse
|
49
|
Yang L, Pijuan-Galito S, Rho HS, Vasilevich AS, Eren AD, Ge L, Habibović P, Alexander MR, de Boer J, Carlier A, van Rijn P, Zhou Q. High-Throughput Methods in the Discovery and Study of Biomaterials and Materiobiology. Chem Rev 2021; 121:4561-4677. [PMID: 33705116 PMCID: PMC8154331 DOI: 10.1021/acs.chemrev.0c00752] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 02/07/2023]
Abstract
The complex interaction of cells with biomaterials (i.e., materiobiology) plays an increasingly pivotal role in the development of novel implants, biomedical devices, and tissue engineering scaffolds to treat diseases, aid in the restoration of bodily functions, construct healthy tissues, or regenerate diseased ones. However, the conventional approaches are incapable of screening the huge amount of potential material parameter combinations to identify the optimal cell responses and involve a combination of serendipity and many series of trial-and-error experiments. For advanced tissue engineering and regenerative medicine, highly efficient and complex bioanalysis platforms are expected to explore the complex interaction of cells with biomaterials using combinatorial approaches that offer desired complex microenvironments during healing, development, and homeostasis. In this review, we first introduce materiobiology and its high-throughput screening (HTS). Then we present an in-depth of the recent progress of 2D/3D HTS platforms (i.e., gradient and microarray) in the principle, preparation, screening for materiobiology, and combination with other advanced technologies. The Compendium for Biomaterial Transcriptomics and high content imaging, computational simulations, and their translation toward commercial and clinical uses are highlighted. In the final section, current challenges and future perspectives are discussed. High-throughput experimentation within the field of materiobiology enables the elucidation of the relationships between biomaterial properties and biological behavior and thereby serves as a potential tool for accelerating the development of high-performance biomaterials.
Collapse
Affiliation(s)
- Liangliang Yang
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Sara Pijuan-Galito
- School
of Pharmacy, Biodiscovery Institute, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Hoon Suk Rho
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Aliaksei S. Vasilevich
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aysegul Dede Eren
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Lu Ge
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Pamela Habibović
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Morgan R. Alexander
- School
of Pharmacy, Boots Science Building, University
of Nottingham, University Park, Nottingham NG7 2RD, U.K.
| | - Jan de Boer
- Department
of Biomedical Engineering, Eindhoven University
of Technology, 5600 MB Eindhoven, The Netherlands
| | - Aurélie Carlier
- Department
of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick van Rijn
- University
of Groningen, W. J. Kolff Institute for Biomedical Engineering and
Materials Science, Department of Biomedical Engineering, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Qihui Zhou
- Institute
for Translational Medicine, Department of Stomatology, The Affiliated
Hospital of Qingdao University, Qingdao
University, Qingdao 266003, China
| |
Collapse
|
50
|
Eftekhari BS, Eskandari M, Janmey PA, Samadikuchaksaraei A, Gholipourmalekabadi M. Conductive chitosan/polyaniline hydrogel with cell-imprinted topography as a potential substrate for neural priming of adipose derived stem cells. RSC Adv 2021; 11:15795-15807. [PMID: 35481217 PMCID: PMC9029165 DOI: 10.1039/d1ra00413a] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Biophysical characteristics of engineered scaffolds such as topography and electroconductivity have shown potentially beneficial effects on stem cell morphology, proliferation, and differentiation toward neural cells. In this study, we fabricated a conductive hydrogel made from chitosan (CS) and polyaniline (PANI) with induced PC12 cell surface topography using a cell imprinting technique to provide both topographical properties and conductivity in a platform. The engineered hydrogel's potential for neural priming of rat adipose-derived stem cells (rADSCs) was determined in vitro. The biomechanical analysis revealed that the electrical conductivity, stiffness, and hydrophobicity of flat (F) and cell-imprinted (CI) substrates increased with increased PANI content in the CS/PANI scaffold. The conductive substrates exhibited a lower degradation rate compared to non-conductive substrates. According to data obtained from F-actin staining and AFM micrographs, both CI(CS) and CI(CS-PANI) substrates induced the morphology of rADSCs from their irregular shape (on flat substrates) into the elongated and bipolar shape of the neuronal-like PC12 cells. Immunostaining analysis revealed that both CI(CS) and CI (CS-PANI) significantly upregulated the expression of GFAP and MAP2, two neural precursor-specific genes, in rADSCs compared with flat substrates. Although the results reveal that both cell-imprinted topography and electrical conductivity affect the neural lineage differentiation, some data demonstrate that the topography effects of the cell-imprinted surface have a more critical role than electrical conductivity on neural priming of ADSCs. The current study provides new insight into the engineering of scaffolds for nerve tissue engineering.
Collapse
Affiliation(s)
- Behnaz Sadat Eftekhari
- Department of Biomedical Engineering, Amirkabir University of Technology 424 Hafez Ave Tehran 15875-4413 Iran +98 21 6454 23 62
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania 1010 Vagelos Research Laboratories, 3340 Smith Walk Philadelphia PA 19104-6383 USA +1 215 573 6815 +1 215 573 7380
| | - Mahnaz Eskandari
- Department of Biomedical Engineering, Amirkabir University of Technology 424 Hafez Ave Tehran 15875-4413 Iran +98 21 6454 23 62
| | - Paul A Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania 1010 Vagelos Research Laboratories, 3340 Smith Walk Philadelphia PA 19104-6383 USA +1 215 573 6815 +1 215 573 7380
| | | | - Mazaher Gholipourmalekabadi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences Tehran Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences Tehran Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences Tehran Iran
| |
Collapse
|