1
|
Huang J, Chen J, Luo Y. Cell-Sheet Shape Transformation by Internally-Driven, Oriented Forces. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2416624. [PMID: 40165759 DOI: 10.1002/adma.202416624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/04/2025] [Indexed: 04/02/2025]
Abstract
During morphogenesis, cells collectively execute directional forces that drive the programmed folding and growth of the layers, forming tissues and organs. The ability to recapitulate aspects of these processes in vitro will constitute a significant leap forward in the field of tissue engineering. Free-standing, self-organizing, cell-laden matrices are fabricated using a sequential deposition approach that uses liquid crystal-templated hydrogel fibers to direct cell arrangements. The orientation of hydrogel fibers is controlled using flow or boundary cues, while their microstructures are controlled by depletion interaction and probed by scattering and microscopy. These fibers effectively direct cells embedded in a collagen matrix, creating multilayer structures through contact guidance and by leveraging steric interactions amongst the cells. In uniformly aligned cell matrices, oriented cells exert traction forces that can induce preferential contraction of the matrix. Simultaneously, the matrix densifies and develops anisotropy through cell remodeling. Such an approach can be extended to create cell arrangements with arbitrary in-plane patterns, allowing for coordinated cell forces and pre-programmed, macroscopic shape changes. This work reveals a fundamentally new path for controlled force generation, emphasizing the role of a carefully designed initial orientational field for manipulating shape transformations of reconstituted matrices.
Collapse
Affiliation(s)
- Junrou Huang
- Department of Mechanical Engineering and Materials Science, Yale University, 9 Hillhouse Ave, New Haven, CT, 06511, USA
| | - Juan Chen
- Department of Mechanical Engineering and Materials Science, Yale University, 9 Hillhouse Ave, New Haven, CT, 06511, USA
| | - Yimin Luo
- Department of Mechanical Engineering and Materials Science, Yale University, 9 Hillhouse Ave, New Haven, CT, 06511, USA
| |
Collapse
|
2
|
Singh N, Sharma A, Goel A, Kumar K, Solanki R, Bhatia D. DNA-based Precision Tools to Probe and Program Mechanobiology and Organ Engineering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410440. [PMID: 39887556 DOI: 10.1002/smll.202410440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/09/2025] [Indexed: 02/01/2025]
Abstract
DNA nanotechnology represents an innovative discipline that combines nanotechnology with biotechnology. It exploits the distinctive characteristics of deoxyribonucleic acid (DNA) to create nanoscale structures and devices with remarkable accuracy and functionality. Researchers may create complex nanostructures with precision and specialized functions using DNA's innate stability, adaptability, and capacity to self-assemble through complementary base-pairing interactions. Integrating multiple disciplines, known as nanobiotechnology, allows the production of sophisticated nanodevices with a broad range of applications. These include precise drug delivery systems, extremely sensitive biosensors, and the construction of intricate tissue scaffolds for regenerative medicine. Moreover, combining DNA nanotechnology with mechanobiology provides a new understanding of how small-scale mechanical stresses and molecular interactions affect cellular activity and tissue development. DNA nanotechnology has the potential to revolutionize molecular diagnostics, tissue engineering, and organ regeneration. This could lead to enormous improvements in biomedicine. This review emphasizes the most recent developments in DNA nanotechnology, explicitly highlighting its significant influence on mechanobiology and its growing involvement in organ engineering. It provides an extensive overview of present trends, obstacles, and future prospects in this fast-progressing area.
Collapse
Affiliation(s)
- Nihal Singh
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gujarat, 382355, India
| | - Ayushi Sharma
- College of Medicine, Taipei Medical University, Taipei City, 110, Taiwan
| | - Anjana Goel
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura, 281406, India
| | - Krishan Kumar
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gujarat, 382355, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Gujarat, 382355, India
| |
Collapse
|
3
|
Mehanna LE, Boyd JD, Remus-Williams S, Racca NM, Spraggins DP, Grady ME, Berron BJ. Improvement of cellular pattern organization and clarity through centrifugal force. Biomed Mater 2025; 20:025025. [PMID: 39746325 PMCID: PMC11823422 DOI: 10.1088/1748-605x/ada508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/04/2025]
Abstract
Rapid and strategic cell placement is necessary for high throughput tissue fabrication. Current adhesive cell patterning systems rely on fluidic shear flow to remove cells outside of the patterned regions, but limitations in washing complexity and uniformity prevent adhesive patterns from being widely applied. Centrifugation is commonly used to study the adhesive strength of cells to various substrates; however, the approach has not been applied to selective cell adhesion systems to create highly organized cell patterns. This study shows centrifugation as a promising method to wash cellular patterns after selective binding of cells to the surface has taken place. After patterning H9C2 cells using biotin-streptavidin as a model adhesive patterning system and washing with centrifugation, there is a significant number of cells removed outside of the patterned areas of the substrate compared to the initial seeding, while there is not a significant number removed from the desired patterned areas. This method is effective in patterning multiple size and linear structures from line widths of 50-200 μm without compromising immediate cell viability below 80%. We also test this procedure on a variety of tube-forming cell lines (MPCs, HUVECs) on various tissue-like surface materials (collagen 1 and Matrigel) with no significant differences in their respective tube formation metrics when the cells were seeded directly on their unconjugated surface versus patterned and washed through centrifugation. This result demonstrates that our patterning and centrifugation system can be adapted to a variety of cell types and substrates to create patterns tailored to many biological applications.
Collapse
Affiliation(s)
- Lauren E Mehanna
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, United States of America
| | - James D Boyd
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Shelley Remus-Williams
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Nicole M Racca
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Dawson P Spraggins
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Martha E Grady
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, United States of America
| | - Brad J Berron
- Department of Chemical and Materials Engineering, University of Kentucky, Lexington, KY, United States of America
| |
Collapse
|
4
|
Lan BQ, Wang YJ, Yu SX, Liu W, Liu YJ. Physical effects of 3-D microenvironments on confined cell behaviors. Am J Physiol Cell Physiol 2024; 327:C1192-C1201. [PMID: 39246142 DOI: 10.1152/ajpcell.00288.2024] [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: 04/30/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Cell migration is a fundamental and functional cellular process, influenced by a complex microenvironment consisting of different cells and extracellular matrix. Recent research has highlighted that, besides biochemical cues from the microenvironment, physical cues can also greatly alter cellular behavior. However, due to the complexity of the microenvironment, little is known about how the physical interactions between migrating cells and surrounding microenvironment instructs cell movement. Here, we explore various examples of three-dimensional microenvironment reconstruction models in vitro and describe how the physical interplay between migrating cells and the neighboring microenvironment controls cell behavior. Understanding this mechanical cooperation will provide key insights into organ development, regeneration, and tumor metastasis.
Collapse
Affiliation(s)
- Bao-Qiong Lan
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Sai-Xi Yu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Wei Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| |
Collapse
|
5
|
Singh A, Cho YK, Cohen DJ. Rapid Whole-Plate Cell and Tissue Micropatterning Using a Budget 3D Resin Printer. ACS OMEGA 2024; 9:43808-43816. [PMID: 39494000 PMCID: PMC11525498 DOI: 10.1021/acsomega.4c06539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 11/05/2024]
Abstract
The ability to precisely pattern cells and proteins is crucial in various scientific disciplines, including cell biology, bioengineering, and materials chemistry. Current techniques, such as microcontact stamping, 3D bioprinting, and direct photopatterning, have limitations in terms of cost, versatility, and throughput. In this Article, we present an accessible approach that combines the throughput of photomask systems with the versatility of programmable light patterning using a low-cost consumer LCD resin printer. The method involves utilizing a bioinert hydrogel, poly(ethylene glycol) diacrylate (PEGDA), and a 405 nm sensitive photoinitiator (LAP) that are selectively cross-linked to form a hydrogel upon light exposure, creating specific regions that are protein and cell-repellent. Our result highlights that a low-cost LCD resin printer can project virtual photomasks onto the hydrogel, allowing for reasonable resolution and large-area printing at a fraction of the cost of traditional systems. The study demonstrates the calibration of exposure times for optimal resolution and accuracy and shape corrections to overcome the inherent challenges of wide-field resin printing. The potential of this approach is validated through widely studied 2D and 3D stem cell applications, showcasing its biocompatibility and ability to replicate complex tissue engineering patterns. We also validate the method with a cell-adhesive polymer (gelatin methacrylate; GelMA). The combination of low cost, high throughput, and accessibility makes this method broadly applicable across fields for enabling rapid and precise fabrication of cells and tissues in standard laboratory culture vessels.
Collapse
Affiliation(s)
- Anamika Singh
- Department
of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Youn Kyoung Cho
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Daniel J. Cohen
- Department
of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
6
|
Sarikhani E, Meganathan DP, Larsen AKK, Rahmani K, Tsai CT, Lu CH, Marquez-Serrano A, Sadr L, Li X, Dong M, Santoro F, Cui B, Klausen LH, Jahed Z. Engineering the Cellular Microenvironment: Integrating Three-Dimensional Nontopographical and Two-Dimensional Biochemical Cues for Precise Control of Cellular Behavior. ACS NANO 2024; 18:19064-19076. [PMID: 38978500 PMCID: PMC11271182 DOI: 10.1021/acsnano.4c03743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 07/10/2024]
Abstract
The development of biomaterials capable of regulating cellular processes and guiding cell fate decisions has broad implications in tissue engineering, regenerative medicine, and cell-based assays for drug development and disease modeling. Recent studies have shown that three-dimensional (3D) nanoscale physical cues such as nanotopography can modulate various cellular processes like adhesion and endocytosis by inducing nanoscale curvature on the plasma and nuclear membranes. Two-dimensional (2D) biochemical cues such as protein micropatterns can also regulate cell function and fate by controlling cellular geometries. Development of biomaterials with precise control over nanoscale physical and biochemical cues can significantly influence programming cell function and fate. In this study, we utilized a laser-assisted micropatterning technique to manipulate the 2D architectures of cells on 3D nanopillar platforms. We performed a comprehensive analysis of cellular and nuclear morphology and deformation on both nanopillar and flat substrates. Our findings demonstrate the precise engineering of single cell architectures through 2D micropatterning on nanopillar platforms. We show that the coupling between the nuclear and cell shape is disrupted on nanopillar surfaces compared to flat surfaces. Furthermore, our results suggest that cell elongation on nanopillars enhances nanopillar-induced endocytosis. We believe our platform serves as a versatile tool for further explorations into programming cell function and fate through combined physical cues that create nanoscale curvature on cell membranes and biochemical cues that control the geometry of the cell.
Collapse
Affiliation(s)
- Einollah Sarikhani
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | - Dhivya Pushpa Meganathan
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | | | - Keivan Rahmani
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | - Ching-Ting Tsai
- Department
of Chemistry, Stanford University, Stanford ,California 94305, United States
| | - Chih-Hao Lu
- Department
of Chemistry, Stanford University, Stanford ,California 94305, United States
| | - Abel Marquez-Serrano
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | - Leah Sadr
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
| | - Xiao Li
- Department
of Chemistry, Stanford University, Stanford ,California 94305, United States
| | - Mingdong Dong
- Interdisciplinary
Nanoscience Center (iNANO), Aarhus University, Aarhus C 8000, Denmark
| | - Francesca Santoro
- Center
for Advanced Biomaterials for Healthcare, Tissue Electronics, Instituto Italiano di Tecnologia, Naples 80125, Italy
- Faculty
of Electrical Engineering and IT, RWTH, Aachen 52074, Germany
- Institute
for Biological Information Processing-Bioelectronics, Forschungszentrum
Juelich, Julich 52428, Germany
| | - Bianxiao Cui
- Department
of Chemistry, Stanford University, Stanford ,California 94305, United States
| | | | - Zeinab Jahed
- Department
of NanoEngineering, University of California
San Diego, La Jolla ,California 92093, United States
- Department
of Bioengineering, University of California
San Diego, La Jolla ,California 92093, United States
| |
Collapse
|
7
|
Heyn JCJ, Rädler JO, Falcke M. Mesenchymal cell migration on one-dimensional micropatterns. Front Cell Dev Biol 2024; 12:1352279. [PMID: 38694822 PMCID: PMC11062138 DOI: 10.3389/fcell.2024.1352279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/29/2024] [Indexed: 05/04/2024] Open
Abstract
Quantitative studies of mesenchymal cell motion are important to elucidate cytoskeleton function and mechanisms of cell migration. To this end, confinement of cell motion to one dimension (1D) significantly simplifies the problem of cell shape in experimental and theoretical investigations. Here we review 1D migration assays employing micro-fabricated lanes and reflect on the advantages of such platforms. Data are analyzed using biophysical models of cell migration that reproduce the rich scenario of morphodynamic behavior found in 1D. We describe basic model assumptions and model behavior. It appears that mechanical models explain the occurrence of universal relations conserved across different cell lines such as the adhesion-velocity relation and the universal correlation between speed and persistence (UCSP). We highlight the unique opportunity of reproducible and standardized 1D assays to validate theory based on statistical measures from large data of trajectories and discuss the potential of experimental settings embedding controlled perturbations to probe response in migratory behavior.
Collapse
Affiliation(s)
- Johannes C. J. Heyn
- Fakultät für Physik, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Joachim O. Rädler
- Fakultät für Physik, Ludwig-Maximilians-Universität München (LMU), Munich, Germany
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Physics, Humboldt University, Berlin, Germany
| |
Collapse
|
8
|
Wistner SC, Rashad L, Slaughter G. Advances in tissue engineering and biofabrication for in vitro skin modeling. BIOPRINTING (AMSTERDAM, NETHERLANDS) 2023; 35:e00306. [PMID: 38645432 PMCID: PMC11031264 DOI: 10.1016/j.bprint.2023.e00306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The global prevalence of skin disease and injury is continually increasing, yet conventional cell-based models used to study these conditions do not accurately reflect the complexity of human skin. The lack of inadequate in vitro modeling has resulted in reliance on animal-based models to test pharmaceuticals, biomedical devices, and industrial and environmental toxins to address clinical needs. These in vivo models are monetarily and morally expensive and are poor predictors of human tissue responses and clinical trial outcomes. The onset of three-dimensional (3D) culture techniques, such as cell-embedded and decellularized approaches, has offered accessible in vitro alternatives, using innovative scaffolds to improve cell-based models' structural and histological authenticity. However, these models lack adequate organizational control and complexity, resulting in variations between structures and the exclusion of physiologically relevant vascular and immunological features. Recently, biofabrication strategies, which combine biology, engineering, and manufacturing capabilities, have emerged as instrumental tools to recreate the heterogeneity of human skin precisely. Bioprinting uses computer-aided design (CAD) to yield robust and reproducible skin prototypes with unprecedented control over tissue design and assembly. As the interdisciplinary nature of biofabrication grows, we look to the promise of next-generation biofabrication technologies, such as organ-on-a-chip (OOAC) and 4D modeling, to simulate human tissue behaviors more reliably for research, pharmaceutical, and regenerative medicine purposes. This review aims to discuss the barriers to developing clinically relevant skin models, describe the evolution of skin-inspired in vitro structures, analyze the current approaches to biofabricating 3D human skin mimetics, and define the opportunities and challenges in biofabricating skin tissue for preclinical and clinical uses.
Collapse
Affiliation(s)
- Sarah C. Wistner
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Layla Rashad
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Gymama Slaughter
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, 23508, USA
| |
Collapse
|
9
|
Engrácia DM, Pinto CIG, Mendes F. Cancer 3D Models for Metallodrug Preclinical Testing. Int J Mol Sci 2023; 24:11915. [PMID: 37569291 PMCID: PMC10418685 DOI: 10.3390/ijms241511915] [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: 06/04/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/13/2023] Open
Abstract
Despite being standard tools in research, the application of cellular and animal models in drug development is hindered by several limitations, such as limited translational significance, animal ethics, and inter-species physiological differences. In this regard, 3D cellular models can be presented as a step forward in biomedical research, allowing for mimicking tissue complexity more accurately than traditional 2D models, while also contributing to reducing the use of animal models. In cancer research, 3D models have the potential to replicate the tumor microenvironment, which is a key modulator of cancer cell behavior and drug response. These features make cancer 3D models prime tools for the preclinical study of anti-tumoral drugs, especially considering that there is still a need to develop effective anti-cancer drugs with high selectivity, minimal toxicity, and reduced side effects. Metallodrugs, especially transition-metal-based complexes, have been extensively studied for their therapeutic potential in cancer therapy due to their distinctive properties; however, despite the benefits of 3D models, their application in metallodrug testing is currently limited. Thus, this article reviews some of the most common types of 3D models in cancer research, as well as the application of 3D models in metallodrug preclinical studies.
Collapse
Affiliation(s)
- Diogo M. Engrácia
- Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal; (D.M.E.); (C.I.G.P.)
| | - Catarina I. G. Pinto
- Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal; (D.M.E.); (C.I.G.P.)
| | - Filipa Mendes
- Center for Nuclear Sciences and Technologies, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal; (D.M.E.); (C.I.G.P.)
- Department of Nuclear Sciences and Engineering, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| |
Collapse
|
10
|
Kopeć K, Podgórski R, Ciach T, Wojasiński M. System for Patterning Polydopamine and VAPG Peptide on Polytetrafluoroethylene and Biodegradable Polyesters for Patterned Growth of Smooth Muscle Cells In Vitro. ACS OMEGA 2023; 8:22055-22066. [PMID: 37360448 PMCID: PMC10285958 DOI: 10.1021/acsomega.3c02114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
Biomaterial's surface functionalization for selective adhesion and patterned cell growth remains essential in developing novel implantable medical devices for regenerative medicine applications. We built and applied a 3D-printed microfluidic device to fabricate polydopamine (PDA) patterns on the surface of polytetrafluoroethylene (PTFE), poly(l-lactic acid-co-D,l-lactic acid) (PLA), and poly(lactic acid-co-glycolic acid) (PLGA). Then, we covalently attached the Val-Ala-Pro-Gly (VAPG) peptide to the created PDA pattern to promote the adhesion of the smooth muscle cells (SMCs). We proved that the fabrication of PDA patterns allows for the selective adhesion of mouse fibroblast and human SMCs to PDA-patterned surfaces after only 30 min of in vitro cultivation. After 7 days of SMC culture, we observed the proliferation of cells only along the patterns on PTFE but over the entire surface of the PLA and PLGA, regardless of patterning. This means that the presented approach is beneficial for application to materials resistant to cell adhesion and proliferation. The additional attachment of the VAPG peptide to the PDA patterns did not bring measurable benefits due to the high increase in adhesion and patterned cell proliferation by PDA itself.
Collapse
Affiliation(s)
- Kamil Kopeć
- Warsaw
University of Technology, Faculty of Chemical and Process Engineering,
Department of Biotechnology and Bioprocess Engineering, Waryńskiego 1, 00-645 Warsaw, Poland
| | - Rafał Podgórski
- Warsaw
University of Technology, Faculty of Chemical and Process Engineering,
Department of Biotechnology and Bioprocess Engineering, Waryńskiego 1, 00-645 Warsaw, Poland
| | - Tomasz Ciach
- Warsaw
University of Technology, Faculty of Chemical and Process Engineering,
Department of Biotechnology and Bioprocess Engineering, Waryńskiego 1, 00-645 Warsaw, Poland
- Warsaw
University of Technology, CEZAMAT, Poleczki 19, 02-822 Warsaw, Poland
| | - Michał Wojasiński
- Warsaw
University of Technology, Faculty of Chemical and Process Engineering,
Department of Biotechnology and Bioprocess Engineering, Waryńskiego 1, 00-645 Warsaw, Poland
| |
Collapse
|
11
|
Becher JE, Lautenschläger F, Thalla DG. A low-cost alternative method of generating fibronectin micropatterned lines for cellular applications. MethodsX 2023; 10:102240. [PMID: 37305805 PMCID: PMC10251141 DOI: 10.1016/j.mex.2023.102240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 05/31/2023] [Indexed: 06/13/2023] Open
Abstract
The cellular microenvironment contributes to the architecture, differentiation, polarity, mechanics and functions of the cell [1]. Spatial confinement of cells using micropatterning techniques allows to alter and regulate the cellular microenvironment for a better understanding of cellular mechanisms [2]. However, commercially available micropatterned consumables such as coverslips, dishes, plates etc. are expensive. These methods are complex and based on deep UV patterning [3,4]. In this study, we establish a low-cost method for effective micropatterning using Polydimethylsiloxane (PDMS) chips.•We demonstrate this method by generating fibronectin-coated micropatterned lines (width, 5 µm) on a glass bottom dish.•As a proof of concept, we culture macrophages on these lines. We additionally show that this method allows to determine the cellular polarity by measuring the position of the nucleus within a cell on a micropatterned line.
Collapse
Affiliation(s)
| | - Franziska Lautenschläger
- Experimental Physics, Saarland University, 66123 Saarbrücken, Germany
- Centre for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| | | |
Collapse
|
12
|
Abuarqoub D, Theeb LS, Omari MB, Hamadneh YI, Alrawabdeh JA, Aslam N, Jafar H, Awidi A. The Osteogenic Role of Biomaterials Combined with Human-Derived Dental Stem Cells in Bone Tissue Regeneration. Tissue Eng Regen Med 2023; 20:251-270. [PMID: 36808303 PMCID: PMC10070593 DOI: 10.1007/s13770-022-00514-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 02/23/2023] Open
Abstract
The use of stem cells in regenerative medicine had great potential for clinical applications. However, cell delivery strategies have critical importance in stimulating the differentiation of stem cells and enhancing their potential to regenerate damaged tissues. Different strategies have been used to investigate the osteogenic potential of dental stem cells in conjunction with biomaterials through in vitro and in vivo studies. Osteogenesis has a broad implication in regenerative medicine, particularly for maxillofacial defects. This review summarizes some of the most recent developments in the field of tissue engineering using dental stem cells.
Collapse
Affiliation(s)
- Duaa Abuarqoub
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.
- Cell Therapy Center, The University of Jordan, Amman, Jordan.
| | - Laith S Theeb
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Mohammad B Omari
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Yazan I Hamadneh
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | | | - Nazneen Aslam
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Hanan Jafar
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan.
- School of Medicine, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
13
|
Carpentier N, Urbani L, Dubruel P, Van Vlierberghe S. The native liver as inspiration to create superior in vitro hepatic models. Biomater Sci 2023; 11:1091-1115. [PMID: 36594602 DOI: 10.1039/d2bm01646j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Drug induced liver injury (DILI) is one of the major reasons of drug withdrawal during the different phases of drug development. The later in the drug development a drug is discovered to be toxic, the higher the economical as well as the ethical impact will be. In vitro models for early detection of drug liver toxicity are under constant development, however to date a superior model of the liver is still lacking. Ideally, a highly reliable model should be established to maintain the different hepatic cell functionalities to the greatest extent possible, during a period of time long enough to allow for tracking of the toxicity of compounds. In the case of DILI, toxicity can appear even after months of exposure. To reach this goal, an in vitro model should be developed that mimics the in vivo liver environment, function and response to external stimuli. The different approaches for the development of liver models currently used in the field of tissue engineering will be described in this review. Combining different technologies, leading to optimal materials, cells and 3D-constructs will ultimately lead to an ideal superior model that fully recapitulates the liver.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Luca Urbani
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK.,Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| |
Collapse
|
14
|
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
|
15
|
Cotner M, Meng S, Jost T, Gardner A, De Santiago C, Brock A. Integration of quantitative methods and mathematical approaches for the modeling of cancer cell proliferation dynamics. Am J Physiol Cell Physiol 2023; 324:C247-C262. [PMID: 36503241 PMCID: PMC9886359 DOI: 10.1152/ajpcell.00185.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
Physiological processes rely on the control of cell proliferation, and the dysregulation of these processes underlies various pathological conditions, including cancer. Mathematical modeling can provide new insights into the complex regulation of cell proliferation dynamics. In this review, we first examine quantitative experimental approaches for measuring cell proliferation dynamics in vitro and compare the various types of data that can be obtained in these settings. We then explore the toolbox of common mathematical modeling frameworks that can describe cell behavior, dynamics, and interactions of proliferation. We discuss how these wet-laboratory studies may be integrated with different mathematical modeling approaches to aid the interpretation of the results and to enable the prediction of cell behaviors, specifically in the context of cancer.
Collapse
Affiliation(s)
- Michael Cotner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Sarah Meng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Tyler Jost
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Andrea Gardner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Carolina De Santiago
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas
| |
Collapse
|
16
|
Levario-Diaz V, Alvarado RE, Rodriguez-Quinteros CM, Fink A, Christian J, Feng W, Cavalcanti-Adam EA. 1D micro-nanopatterned integrin ligand surfaces for directed cell movement. Front Cell Dev Biol 2022; 10:972624. [PMID: 36531964 PMCID: PMC9755580 DOI: 10.3389/fcell.2022.972624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2023] Open
Abstract
Cell-extracellular matrix (ECM) adhesion mediated by integrins is a highly regulated process involved in many vital cellular functions such as motility, proliferation and survival. However, the influence of lateral integrin clustering in the coordination of cell front and rear dynamics during cell migration remains unresolved. For this purpose, we describe a novel protocol to fabricate 1D micro-nanopatterned stripes by integrating the block copolymer micelle nanolithography (BCMNL) technique and maskless near UV lithography-based photopatterning. The photopatterned 10 μm-wide stripes consist of a quasi-perfect hexagonal arrangement of gold nanoparticles, decorated with the RGD (arginine-glycine-aspartate) motif for single integrin heterodimer binding, and placed at a distance of 50, 80, and 100 nm to regulate integrin clustering and focal adhesion dynamics. By employing time-lapse microscopy and immunostaining, we show that the displacement and speed of fibroblasts changes according to the nanoscale spacing of adhesion sites. We found that as the lateral spacing of adhesive peptides increased, fibroblast morphology was more elongated. This was accompanied by a decreased formation of mature focal adhesions and stress fibers, which increased cell displacement and speed. These results provide new insights into the migratory behavior of fibroblasts in 1D environments and our protocol offers a new platform to design and manufacture confined environments in 1D for integrin-mediated cell adhesion.
Collapse
Affiliation(s)
- Victoria Levario-Diaz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg, Germany
| | | | | | - Andreas Fink
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Joel Christian
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Wenqian Feng
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg, Germany
- College of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | | |
Collapse
|
17
|
Ortiz-Cárdenas JE, Zatorski JM, Arneja A, Montalbine AN, Munson JM, Luckey CJ, Pompano RR. Towards spatially-organized organs-on-chip: Photopatterning cell-laden thiol-ene and methacryloyl hydrogels in a microfluidic device. ORGANS-ON-A-CHIP 2022; 4:100018. [PMID: 35535262 PMCID: PMC9078144 DOI: 10.1016/j.ooc.2022.100018] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Micropatterning techniques for 3D cell cultures enable the recreation of tissue-level structures, but the combination of patterned hydrogels with organs-on-chip to generate organized 3D cultures under microfluidic perfusion remains challenging. To address this technological gap, we developed a user-friendly in-situ micropatterning protocol that integrates photolithography of crosslinkable, cell-laden hydrogels with a simple microfluidic housing, and tested the impact of crosslinking chemistry on stability and spatial resolution. Working with gelatin functionalized with photo-crosslinkable moieties, we found that inclusion of cells at high densities (≥ 107/mL) did not impede thiol-norbornene gelation, but decreased the storage moduli of methacryloyl hydrogels. Hydrogel composition and light dose were selected to match the storage moduli of soft tissues. To generate the desired pattern on-chip, the cell-laden precursor solution was flowed into a microfluidic chamber and exposed to 405 nm light through a photomask. The on-chip 3D cultures were self-standing and the designs were interchangeable by simply swapping out the photomask. Thiol-ene hydrogels yielded highly accurate feature sizes from 100 - 900 μm in diameter, whereas methacryloyl hydrogels yielded slightly enlarged features. Furthermore, only thiol-ene hydrogels were mechanically stable under perfusion overnight. Repeated patterning readily generated multi-region cultures, either separately or adjacent, including non-linear boundaries that are challenging to obtain on-chip. As a proof-of-principle, primary human T cells were patterned on-chip with high regional specificity. Viability remained high (> 85%) after 12-hr culture with constant perfusion. We envision that this technology will enable researchers to pattern 3D co-cultures to mimic organ-like structures that were previously difficult to obtain.
Collapse
Affiliation(s)
| | - Jonathan M. Zatorski
- Department of Chemistry, University of Virginia, PO BOX 400319, Charlottesville, VA, USA 22904
| | - Abhinav Arneja
- Department of Pathology, University of Virginia, Charlottesville, VA, USA 22904
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, PO BOX 400319, Charlottesville, VA, USA 22904
| | - Jennifer M. Munson
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | - Chance John Luckey
- Department of Pathology, University of Virginia, Charlottesville, VA, USA 22904
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, PO BOX 400319, Charlottesville, VA, USA 22904
- Department of Chemistry, Carter Immunology Center, University of Virginia, PO BOX 400319, Charlottesville, VA, USA 22904
| |
Collapse
|
18
|
Chi J, Wang M, Chen J, Hu L, Chen Z, Backman LJ, Zhang W. Topographic Orientation of Scaffolds for Tissue Regeneration: Recent Advances in Biomaterial Design and Applications. Biomimetics (Basel) 2022; 7:131. [PMID: 36134935 PMCID: PMC9496066 DOI: 10.3390/biomimetics7030131] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/30/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Tissue engineering to develop alternatives for the maintenance, restoration, or enhancement of injured tissues and organs is gaining more and more attention. In tissue engineering, the scaffold used is one of the most critical elements. Its characteristics are expected to mimic the native extracellular matrix and its unique topographical structures. Recently, the topographies of scaffolds have received increasing attention, not least because different topographies, such as aligned and random, have different repair effects on various tissues. In this review, we have focused on various technologies (electrospinning, directional freeze-drying, magnetic freeze-casting, etching, and 3-D printing) to fabricate scaffolds with different topographic orientations, as well as discussed the physicochemical (mechanical properties, porosity, hydrophilicity, and degradation) and biological properties (morphology, distribution, adhesion, proliferation, and migration) of different topographies. Subsequently, we have compiled the effect of scaffold orientation on the regeneration of vessels, skin, neural tissue, bone, articular cartilage, ligaments, tendons, cardiac tissue, corneas, skeletal muscle, and smooth muscle. The compiled information in this review will facilitate the future development of optimal topographical scaffolds for the regeneration of certain tissues. In the majority of tissues, aligned scaffolds are more suitable than random scaffolds for tissue repair and regeneration. The underlying mechanism explaining the various effects of aligned and random orientation might be the differences in "contact guidance", which stimulate certain biological responses in cells.
Collapse
Affiliation(s)
- Jiayu Chi
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Mingyue Wang
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| | - Lizhi Hu
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhixuan Chen
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Ludvig J. Backman
- Department of Integrative Medical Biology, Anatomy, Umeå University, SE-901 87 Umeå, Sweden
- Department of Community Medicine and Rehabilitation, Physiotherapy, Umeå University, SE-901 87 Umeå, Sweden
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing 210009, China
- Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing 210096, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China
| |
Collapse
|
19
|
Molugu K, Battistini GA, Heaster TM, Rouw J, Guzman EC, Skala MC, Saha K. Label-Free Imaging to Track Reprogramming of Human Somatic Cells. GEN BIOTECHNOLOGY 2022; 1:176-191. [PMID: 35586336 PMCID: PMC9092522 DOI: 10.1089/genbio.2022.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/28/2022] [Indexed: 11/12/2022]
Abstract
The process of reprogramming patient samples to human-induced pluripotent stem cells (iPSCs) is stochastic, asynchronous, and inefficient, leading to a heterogeneous population of cells. In this study, we track the reprogramming status of patient-derived erythroid progenitor cells (EPCs) at the single-cell level during reprogramming with label-free live-cell imaging of cellular metabolism and nuclear morphometry to identify high-quality iPSCs. EPCs isolated from human peripheral blood of three donors were used for our proof-of-principle study. We found distinct patterns of autofluorescence lifetime for the reduced form of nicotinamide adenine dinucleotide (phosphate) and flavin adenine dinucleotide during reprogramming. Random forest models classified iPSCs with ∼95% accuracy, which enabled the successful isolation of iPSC lines from reprogramming cultures. Reprogramming trajectories resolved at the single-cell level indicated significant reprogramming heterogeneity along different branches of cell states. This combination of micropatterning, autofluorescence imaging, and machine learning provides a unique, real-time, and nondestructive method to assess the quality of iPSCs in a biomanufacturing process, which could have downstream impacts in regenerative medicine, cell/gene therapy, and disease modeling.
Collapse
Affiliation(s)
- Kaivalya Molugu
- Biophysics Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
| | - Giovanni A. Battistini
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
| | - Tiffany M. Heaster
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; and Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Jacob Rouw
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; and Madison, Wisconsin, USA
| | | | - Melissa C. Skala
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; and Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Krishanu Saha
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA; Madison, Wisconsin, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA; and Madison, Wisconsin, USA
| |
Collapse
|
20
|
Jagiełło A, Hu Q, Castillo U, Botvinick E. Patterned photocrosslinking to establish stiffness anisotropies in fibrous 3D hydrogels. Acta Biomater 2022; 141:39-47. [PMID: 34971786 DOI: 10.1016/j.actbio.2021.12.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/04/2021] [Accepted: 12/22/2021] [Indexed: 11/28/2022]
Abstract
Cells are known to constantly interact with their local extracellular matrix (ECM) and respond to a variety of biochemical and mechanical cues received from the ECM. Nonetheless, comprehensive understanding of cell-ECM interactions has been elusive. Many studies rely on analysis of cell behavior on 2D substrates, which do not reflect a natural cell environment. Further, lack of dynamic control over local stiffness anisotropies and fiber alignment hinders progress in studies in naturally derived fibrous 3D cultures. Here, we present a cell-safe method of patterned photocrosslinking, which can aid in studying biological hypotheses related to mechanotransduction in 3D hydrogels. As previously described by our group, ruthenium-catalyzed photocrosslinking (RCP) of selected ECM regions promotes localized increase in stiffness mediated by focused blue laser light in a confocal microscope. In this study, we further demonstrate that RCP can induce localized strain stiffening and fiber alignment outside of the selected crosslinked region and induce stiffness anisotropy biased towards the direction of fiber alignment. MDA-MB-231 cells are shown to respond to RCP-induced changes in local ECM architecture and display directional bias towards the direction of fiber alignment, as compared to control cells. Further, the effect of patterned crosslinking on a stiffness landscape is measured using multi-axes optical tweezers active microrheology (AMR) with backscattered laser beam illumination. AMR validates RCP as a suitable tool for creating distinct stiffness anisotropies which promote directed migration of cells, further underscoring the usefulness of RCP in cell-ECM studies. STATEMENT OF SIGNIFICANCE: Studies on cell-ECM interactions in 3D cultures have often been hindered by the lack of available tools to dynamically alter local ECM stiffness and fiber alignment. Here, we present a non-invasive, cell-safe and easily applicable method of patterned photocrosslinking, which can aid in studying biological hypotheses in fibrous 3D hydrogels. Ruthenium-catalyzed crosslinking (RCP) of selected fibrin ECM regions promotes localized increase in stiffness and creates distinct stiffness anisotropies in the presence of the focused blue laser light. Outside of the crosslinked region, RCP causes fiber alignment and strain stiffening in the ECM, verified using multi-axes optical tweezers active microrheology (AMR). Following RCP, human breast cancer MDA-MB-231 exhibit directed cell migration, validating usefulness of this method in cell-ECM studies.
Collapse
Affiliation(s)
- Alicja Jagiełło
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697-2730, United States
| | - Qingda Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697-2730, United States; Center for Complex Biological Systems, University of California Irvine, Irvine, CA, 92697-2280, United States
| | - Ulysses Castillo
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697-2730, United States
| | - Elliot Botvinick
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, 92697-2730, United States; Center for Complex Biological Systems, University of California Irvine, Irvine, CA, 92697-2280, United States; Beckman Laser Institute and Medical Clinic, University of California Irvine, Irvine, CA, 92612, United States; Department of Surgery, University of California Irvine, 333 City Boulevard, Suite 700, Orange, CA, 92868, United States; The Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California Irvine, Irvine, CA, 92697-2730, United States.
| |
Collapse
|
21
|
Nitsche KS, Müller I, Malcomber S, Carmichael PL, Bouwmeester H. Implementing organ-on-chip in a next-generation risk assessment of chemicals: a review. Arch Toxicol 2022; 96:711-741. [PMID: 35103818 PMCID: PMC8850248 DOI: 10.1007/s00204-022-03234-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 12/17/2022]
Abstract
Organ-on-chip (OoC) technology is full of engineering and biological challenges, but it has the potential to revolutionize the Next-Generation Risk Assessment of novel ingredients for consumer products and chemicals. A successful incorporation of OoC technology into the Next-Generation Risk Assessment toolbox depends on the robustness of the microfluidic devices and the organ tissue models used. Recent advances in standardized device manufacturing, organ tissue cultivation and growth protocols offer the ability to bridge the gaps towards the implementation of organ-on-chip technology. Next-Generation Risk Assessment is an exposure-led and hypothesis-driven tiered approach to risk assessment using detailed human exposure information and the application of appropriate new (non-animal) toxicological testing approaches. Organ-on-chip presents a promising in vitro approach by combining human cell culturing with dynamic microfluidics to improve physiological emulation. Here, we critically review commercial organ-on-chip devices, as well as recent tissue culture model studies of the skin, intestinal barrier and liver as the main metabolic organ to be used on-chip for Next-Generation Risk Assessment. Finally, microfluidically linked tissue combinations such as skin-liver and intestine-liver in organ-on-chip devices are reviewed as they form a relevant aspect for advancing toxicokinetic and toxicodynamic studies. We point to recent achievements and challenges to overcome, to advance non-animal, human-relevant safety studies.
Collapse
Affiliation(s)
- Katharina S Nitsche
- Division of Toxicology, Wageningen University, P.O. Box 8000, 6700 EA, Wageningen, The Netherlands.
| | - Iris Müller
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - Sophie Malcomber
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - Paul L Carmichael
- Division of Toxicology, Wageningen University, P.O. Box 8000, 6700 EA, Wageningen, The Netherlands
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University, P.O. Box 8000, 6700 EA, Wageningen, The Netherlands
| |
Collapse
|
22
|
Cecchini A, Cornelison DDW. Eph/Ephrin-Based Protein Complexes: The Importance of cis Interactions in Guiding Cellular Processes. Front Mol Biosci 2022; 8:809364. [PMID: 35096972 PMCID: PMC8793696 DOI: 10.3389/fmolb.2021.809364] [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: 11/09/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Although intracellular signal transduction is generally represented as a linear process that transmits stimuli from the exterior of a cell to the interior via a transmembrane receptor, interactions with additional membrane-associated proteins are often critical to its success. These molecules play a pivotal role in mediating signaling via the formation of complexes in cis (within the same membrane) with primary effectors, particularly in the context of tumorigenesis. Such secondary effectors may act to promote successful signaling by mediating receptor-ligand binding, recruitment of molecular partners for the formation of multiprotein complexes, or differential signaling outcomes. One signaling family whose contact-mediated activity is frequently modulated by lateral interactions at the cell surface is Eph/ephrin (EphA and EphB receptor tyrosine kinases and their ligands ephrin-As and ephrin-Bs). Through heterotypic interactions in cis, these molecules can promote a diverse range of cellular activities, including some that are mutually exclusive (cell proliferation and cell differentiation, or adhesion and migration). Due to their broad expression in most tissues and their promiscuous binding within and across classes, the cellular response to Eph:ephrin interaction is highly variable between cell types and is dependent on the cellular context in which binding occurs. In this review, we will discuss interactions between molecules in cis at the cell membrane, with emphasis on their role in modulating Eph/ephrin signaling.
Collapse
Affiliation(s)
- Alessandra Cecchini
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - D. D. W. Cornelison
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
- *Correspondence: D. D. W. Cornelison,
| |
Collapse
|
23
|
Simon G, Plouhinec JL, Sorre B. Differentiation of EpiLCs on Micropatterned Substrates Generated by Micro-Contact Printing. Methods Mol Biol 2022; 2490:251-268. [PMID: 35486251 DOI: 10.1007/978-1-0716-2281-0_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
During the last decades, signaling pathways responsible for the initiation of gastrulation in mammalian embryos have been identified. However, the physical rules governing the tissue spatial patterning and the extensive morphogenetic movements occurring during that process are still elusive. Progress on these issues is slowed by the difficulty to record or perturb the patterning events in real time, especially in mammalian embryos that develop in utero. Because they permit easy observation and manipulation, in vitro model systems offer an exciting opportunity to dissect the rules governing the organization of the mammalian gastrula. For instance, it is sufficient to cultivate human embryonic stem cells on micropatterned substrates to reveal their self-organization potential. We present here a method to obtain micropatterned mouse Epiblast Like Cells colonies, providing a convenient way to compare spatial organization of mouse and human pluripotent stem cells and to complement the characterization of mutant embryos in a controlled environment.
Collapse
Affiliation(s)
- Gaël Simon
- Institut Jacques Monod, UMR 7592, CNRS, Université de Paris, Paris, France
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, Paris, France
| | - Jean-Louis Plouhinec
- Laboratoire "Matière et Systèmes Complexes" (MSC), UMR 7057 CNRS, Université de Paris, Paris, France
| | - Benoit Sorre
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, Paris, France.
- Laboratoire "Matière et Systèmes Complexes" (MSC), UMR 7057 CNRS, Université de Paris, Paris, France.
| |
Collapse
|
24
|
Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6:426. [PMID: 34916490 PMCID: PMC8674418 DOI: 10.1038/s41392-021-00830-x] [Citation(s) in RCA: 410] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Hydrogel is a type of versatile platform with various biomedical applications after rational structure and functional design that leverages on material engineering to modulate its physicochemical properties (e.g., stiffness, pore size, viscoelasticity, microarchitecture, degradability, ligand presentation, stimulus-responsive properties, etc.) and influence cell signaling cascades and fate. In the past few decades, a plethora of pioneering studies have been implemented to explore the cell-hydrogel matrix interactions and figure out the underlying mechanisms, paving the way to the lab-to-clinic translation of hydrogel-based therapies. In this review, we first introduced the physicochemical properties of hydrogels and their fabrication approaches concisely. Subsequently, the comprehensive description and deep discussion were elucidated, wherein the influences of different hydrogels properties on cell behaviors and cellular signaling events were highlighted. These behaviors or events included integrin clustering, focal adhesion (FA) complex accumulation and activation, cytoskeleton rearrangement, protein cyto-nuclei shuttling and activation (e.g., Yes-associated protein (YAP), catenin, etc.), cellular compartment reorganization, gene expression, and further cell biology modulation (e.g., spreading, migration, proliferation, lineage commitment, etc.). Based on them, current in vitro and in vivo hydrogel applications that mainly covered diseases models, various cell delivery protocols for tissue regeneration and disease therapy, smart drug carrier, bioimaging, biosensor, and conductive wearable/implantable biodevices, etc. were further summarized and discussed. More significantly, the clinical translation potential and trials of hydrogels were presented, accompanied with which the remaining challenges and future perspectives in this field were emphasized. Collectively, the comprehensive and deep insights in this review will shed light on the design principles of new biomedical hydrogels to understand and modulate cellular processes, which are available for providing significant indications for future hydrogel design and serving for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China.
| | - Kun Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China.
| |
Collapse
|
25
|
Gu T, Meesrisom A, Luo Y, Dinh QN, Lin S, Yang M, Sharma A, Tang R, Zhang J, Jia Z, Millner PD, Pearlstein AJ, Zhang B. Listeria monocytogenes biofilm formation as affected by stainless steel surface topography and coating composition. Food Control 2021. [DOI: 10.1016/j.foodcont.2021.108275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
26
|
Abstract
Micropatterning encompasses a set of methods aimed at precisely controlling the spatial distribution of molecules onto the surface of materials. Biologists have borrowed the idea and adapted these methods, originally developed for electronics, to impose physical constraints on biological systems with the aim of addressing fundamental questions across biological scales from molecules to multicellular systems. Here, I approach this topic from a developmental biologist's perspective focusing specifically on how and why micropatterning has gained in popularity within the developmental biology community in recent years. Overall, this Primer provides a concise overview of how micropatterns are used to study developmental processes and emphasises how micropatterns are a useful addition to the developmental biologist's toolbox.
Collapse
Affiliation(s)
- Guillaume Blin
- Institute for Regeneration and Repair, Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| |
Collapse
|
27
|
Kukla DA, Khetani SR. Bioengineered Liver Models for Investigating Disease Pathogenesis and Regenerative Medicine. Semin Liver Dis 2021; 41:368-392. [PMID: 34139785 DOI: 10.1055/s-0041-1731016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Owing to species-specific differences in liver pathways, in vitro human liver models are utilized for elucidating mechanisms underlying disease pathogenesis, drug development, and regenerative medicine. To mitigate limitations with de-differentiated cultures, bioengineers have developed advanced techniques/platforms, including micropatterned cocultures, spheroids/organoids, bioprinting, and microfluidic devices, for perfusing cell cultures and liver slices. Such techniques improve mature functions and culture lifetime of primary and stem-cell human liver cells. Furthermore, bioengineered liver models display several features of liver diseases including infections with pathogens (e.g., malaria, hepatitis C/B viruses, Zika, dengue, yellow fever), alcoholic/nonalcoholic fatty liver disease, and cancer. Here, we discuss features of bioengineered human liver models, their uses for modeling aforementioned diseases, and how such models are being augmented/adapted for fabricating implantable human liver tissues for clinical therapy. Ultimately, continued advances in bioengineered human liver models have the potential to aid the development of novel, safe, and efficacious therapies for liver disease.
Collapse
Affiliation(s)
- David A Kukla
- Deparment of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R Khetani
- Deparment of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
28
|
Al Halawani A, Wang Z, Liu L, Zhang M, Weiss AS. Applications of Engineering Techniques in Microvasculature Design. Front Cardiovasc Med 2021; 8:660958. [PMID: 33981737 PMCID: PMC8107229 DOI: 10.3389/fcvm.2021.660958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/30/2021] [Indexed: 11/17/2022] Open
Abstract
Achieving successful microcirculation in tissue engineered constructs in vitro and in vivo remains a challenge. Engineered tissue must be vascularized in vitro for successful inosculation post-implantation to allow instantaneous perfusion. To achieve this, most engineering techniques rely on engineering channels or pores for guiding angiogenesis and capillary tube formation. However, the chosen materials should also exhibit properties resembling the native extracellular matrix (ECM) in providing mechanical and molecular cues for endothelial cells. This review addresses techniques that can be used in conjunction with matrix-mimicking materials to further advance microvasculature design. These include electrospinning, micropatterning and bioprinting. Other techniques implemented for vascularizing organoids are also considered for their potential to expand on these approaches.
Collapse
Affiliation(s)
- Aleen Al Halawani
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Ziyu Wang
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Linyang Liu
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Miao Zhang
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- School of Biomedical Engineering, University of Sydney, Sydney, NSW, Australia
| | - Anthony S. Weiss
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- Sydney Nano Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
29
|
Shinde A, Illath K, Gupta P, Shinde P, Lim KT, Nagai M, Santra TS. A Review of Single-Cell Adhesion Force Kinetics and Applications. Cells 2021; 10:577. [PMID: 33808043 PMCID: PMC8000588 DOI: 10.3390/cells10030577] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Cells exert, sense, and respond to the different physical forces through diverse mechanisms and translating them into biochemical signals. The adhesion of cells is crucial in various developmental functions, such as to maintain tissue morphogenesis and homeostasis and activate critical signaling pathways regulating survival, migration, gene expression, and differentiation. More importantly, any mutations of adhesion receptors can lead to developmental disorders and diseases. Thus, it is essential to understand the regulation of cell adhesion during development and its contribution to various conditions with the help of quantitative methods. The techniques involved in offering different functionalities such as surface imaging to detect forces present at the cell-matrix and deliver quantitative parameters will help characterize the changes for various diseases. Here, we have briefly reviewed single-cell mechanical properties for mechanotransduction studies using standard and recently developed techniques. This is used to functionalize from the measurement of cellular deformability to the quantification of the interaction forces generated by a cell and exerted on its surroundings at single-cell with attachment and detachment events. The adhesive force measurement for single-cell microorganisms and single-molecules is emphasized as well. This focused review should be useful in laying out experiments which would bring the method to a broader range of research in the future.
Collapse
Affiliation(s)
- Ashwini Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Kavitha Illath
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Pallavi Shinde
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon-Si, Gangwon-Do 24341, Korea;
| | - Moeto Nagai
- Department of Mechanical Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempaku-cho, Toyohashi, Aichi 441-8580, Japan;
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India; (A.S.); (K.I.); (P.G.); (P.S.)
| |
Collapse
|
30
|
Biofabrication of aligned structures that guide cell orientation and applications in tissue engineering. Biodes Manuf 2021. [DOI: 10.1007/s42242-020-00104-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Amirghasemi F, Adjei-Sowah E, Pockaj BA, Nikkhah M. Microengineered 3D Tumor Models for Anti-Cancer Drug Discovery in Female-Related Cancers. Ann Biomed Eng 2021; 49:1943-1972. [PMID: 33403451 DOI: 10.1007/s10439-020-02704-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
The burden of cancer continues to increase in society and negatively impacts the lives of numerous patients. Due to the high cost of current treatment strategies, there is a crucial unmet need to develop inexpensive preclinical platforms to accelerate the process of anti-cancer drug discovery to improve outcomes in cancer patients, most especially in female patients. Many current methods employ expensive animal models which not only present ethical concerns but also do not often accurately predict human physiology and the outcomes of anti-cancer drug responsiveness. Conventional treatment approaches for cancer generally include systemic therapy after a surgical procedure. Although this treatment technique is effective, the outcome is not always positive due to various complex factors such as intratumor heterogeneity and confounding factors within the tumor microenvironment (TME). Patients who develop metastatic disease still have poor prognosis. To that end, recent efforts have attempted to use 3D microengineered platforms to enhance the predictive power and efficacy of anti-cancer drug screening, ultimately to develop personalized therapies. Fascinating features of microengineered assays, such as microfluidics, have led to the advancement in the development of the tumor-on-chip technology platforms, which have shown tremendous potential for meaningful and physiologically relevant anti-cancer drug discovery and screening. Three dimensional microscale models provide unprecedented ability to unveil the biological complexities of cancer and shed light into the mechanism of anti-cancer drug resistance in a timely and resource efficient manner. In this review, we discuss recent advances in the development of microengineered tumor models for anti-cancer drug discovery and screening in female-related cancers. We specifically focus on female-related cancers to draw attention to the various approaches being taken to improve the survival rate of women diagnosed with cancers caused by sex disparities. We also briefly discuss other cancer types like colon adenocarcinomas and glioblastoma due to their high rate of occurrence in females, as well as the high likelihood of sex-biased mutations which complicate current treatment strategies for women. We highlight recent advances in the development of 3D microscale platforms including 3D tumor spheroids, microfluidic platforms as well as bioprinted models, and discuss how they have been utilized to address major challenges in the process of drug discovery, such as chemoresistance, intratumor heterogeneity, drug toxicity, etc. We also present the potential of these platform technologies for use in high-throughput drug screening approaches as a replacements of conventional assays. Within each section, we will provide our perspectives on advantages of the discussed platform technologies.
Collapse
Affiliation(s)
- Farbod Amirghasemi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287-9709, USA
| | - Emmanuela Adjei-Sowah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287-9709, USA
| | - Barbara A Pockaj
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic, Phoenix, AZ, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287-9709, USA. .,Biodesign Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
32
|
Manufacturing micropatterned collagen scaffolds with chemical-crosslinking for development of biomimetic tissue-engineered oral mucosa. Sci Rep 2020; 10:22192. [PMID: 33335194 PMCID: PMC7747639 DOI: 10.1038/s41598-020-79114-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 12/01/2020] [Indexed: 12/19/2022] Open
Abstract
The junction between the epithelium and the underlying connective tissue undulates, constituting of rete ridges, which lack currently available soft tissue constructs. In this study, using a micro electro mechanical systems process and soft lithography, fifteen negative molds, with different dimensions and aspect ratios in grid- and pillar-type configurations, were designed and fabricated to create three-dimensional micropatterns and replicated onto fish-scale type I collagen scaffolds treated with chemical crosslinking. Image analyses showed the micropatterns were well-transferred onto the scaffold surfaces, showing the versatility of our manufacturing system. With the help of rheological test, the collagen scaffold manufactured in this study was confirmed to be an ideal gel and have visco-elastic features. As compared with our previous study, its mechanical and handling properties were improved by chemical cross-linking, which is beneficial for grafting and suturing into the complex structures of oral cavity. Histologic evaluation of a tissue-engineered oral mucosa showed the topographical microstructures of grid-type were well-preserved, rather than pillar-type, a well-stratified epithelial layer was regenerated on all scaffolds and the epithelial rete ridge-like structure was developed. As this three-dimensional microstructure is valuable for maintaining epithelial integrity, our micropatterned collagen scaffolds can be used not only intraorally but extraorally as a graft material for human use.
Collapse
|
33
|
Erben A, Hörning M, Hartmann B, Becke T, Eisler SA, Southan A, Cranz S, Hayden O, Kneidinger N, Königshoff M, Lindner M, Tovar GEM, Burgstaller G, Clausen‐Schaumann H, Sudhop S, Heymann M. Precision 3D-Printed Cell Scaffolds Mimicking Native Tissue Composition and Mechanics. Adv Healthc Mater 2020; 9:e2000918. [PMID: 33025765 DOI: 10.1002/adhm.202000918] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/29/2020] [Indexed: 12/20/2022]
Abstract
Cellular dynamics are modeled by the 3D architecture and mechanics of the extracellular matrix (ECM) and vice versa. These bidirectional cell-ECM interactions are the basis for all vital tissues, many of which have been investigated in 2D environments over the last decades. Experimental approaches to mimic in vivo cell niches in 3D with the highest biological conformity and resolution can enable new insights into these cell-ECM interactions including proliferation, differentiation, migration, and invasion assays. Here, two-photon stereolithography is adopted to print up to mm-sized high-precision 3D cell scaffolds at micrometer resolution with defined mechanical properties from protein-based resins, such as bovine serum albumin or gelatin methacryloyl. By modifying the manufacturing process including two-pass printing or post-print crosslinking, high precision scaffolds with varying Young's moduli ranging from 7-300 kPa are printed and quantified through atomic force microscopy. The impact of varying scaffold topographies on the dynamics of colonizing cells is observed using mouse myoblast cells and a 3D-lung microtissue replica colonized with primary human lung fibroblast. This approach will allow for a systematic investigation of single-cell and tissue dynamics in response to defined mechanical and bio-molecular cues and is ultimately scalable to full organs.
Collapse
Affiliation(s)
- Amelie Erben
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Heinz‐Nixdorf‐Chair of Biomedical Electronics, TranslaTUM, Campus Klinikum rechts der Isar Technical University of Munich Einsteinstraße 25 Munich 81675 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Marcel Hörning
- Institute of Biomaterials and Biomolecular Systems University of Stuttgart Pfaffenwaldring 57 Stuttgart 70569 Germany
| | - Bastian Hartmann
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Tanja Becke
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Stephan A. Eisler
- Stuttgart Research Center Systems Biology University of Stuttgart Nobelstr. 15 Stuttgart 70569 Germany
| | - Alexander Southan
- Institute of Interfacial Process Engineering and Plasma Technology IGVP University of Stuttgart Nobelstr. 12 Stuttgart 70569 Germany
| | - Séverine Cranz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Research Unit Lung Repair and Regeneration Helmholtz Zentrum München Max‐Lebsche‐Platz 31 Munich 81377 Germany
| | - Oliver Hayden
- Heinz‐Nixdorf‐Chair of Biomedical Electronics, TranslaTUM, Campus Klinikum rechts der Isar Technical University of Munich Einsteinstraße 25 Munich 81675 Germany
| | - Nikolaus Kneidinger
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Department of Internal Medicine V Ludwig‐Maximillians‐University Munich Marchioninistr. 15 Munich 81377 Germany
| | - Melanie Königshoff
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Research Unit Lung Repair and Regeneration Helmholtz Zentrum München Max‐Lebsche‐Platz 31 Munich 81377 Germany
- University of Colorado Department of Pulmonary Sciences and Critical Care Medicine 13001 E. 17th Pl. Aurora CO 80045 USA
| | - Michael Lindner
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- University Department of Visceral and Thoracic Surgery Salzburg Paracelsus Medical University Müllner Hauptstraße 48 Salzburg A‐5020 Austria
| | - Günter E. M. Tovar
- Institute of Interfacial Process Engineering and Plasma Technology IGVP University of Stuttgart Nobelstr. 12 Stuttgart 70569 Germany
| | - Gerald Burgstaller
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Institute of Lung Biology and Disease (ILBD) Helmholtz Zentrum München Max‐Lebsche‐Platz 31 Munich 81377 Germany
| | - Hauke Clausen‐Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Stefanie Sudhop
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Michael Heymann
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
- Institute of Biomaterials and Biomolecular Systems University of Stuttgart Pfaffenwaldring 57 Stuttgart 70569 Germany
- Department of Cellular and Molecular Biophysics MPI of Biochemistry Martinsried Am Klopferspitz 18 Planegg 82152 Germany
| |
Collapse
|
34
|
Abstract
Brain tumors' severity ranges from benign to highly aggressive and invasive. Bioengineering tools can assist in understanding the pathophysiology of these tumors from outside the body and facilitate development of suitable antitumoral treatments. Here, we first describe the physiology and cellular composition of brain tumors. Then, we discuss the development of three-dimensional tissue models utilizing brain tumor cells. In particular, we highlight the role of hydrogels in providing a biomimetic support for the cells to grow into defined structures. Microscale technologies, such as electrospinning and bioprinting, and advanced cellular models aim to mimic the extracellular matrix and natural cellular localization in engineered tumor tissues. Lastly, we review current applications and prospects of hydrogels for therapeutic purposes, such as drug delivery and co-administration with other therapies. Through further development, hydrogels can serve as a reliable option for in vitro modeling and treatment of brain tumors for translational medicine.
Collapse
|
35
|
Orozco P, Montoya Y, Bustamante J. Development of endomyocardial fibrosis model using a cell patterning technique: In vitro interaction of cell coculture of 3T3 fibroblasts and RL-14 cardiomyocytes. PLoS One 2020; 15:e0229158. [PMID: 32092082 PMCID: PMC7039516 DOI: 10.1371/journal.pone.0229158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/30/2020] [Indexed: 01/07/2023] Open
Abstract
Cardiac functions can be altered by changes in the microstructure of the heart, i.e., remodeling of the cardiac tissue, which may activate pathologies such as hypertrophy, dilation, or cardiac fibrosis. Cardiac fibrosis can develop due to an excessive deposition of extracellular matrix proteins, which are products of the activation of fibroblasts. In this context, the anatomical-histological change may interfere with the functioning of the cardiac tissue, which requires specialized cells for its operation. The purpose of the present study was to determine the cellular interactions and morphological changes in cocultures of 3T3 fibroblasts and RL-14 cardiomyocytes via the generation of a platform an in vitro model. For this purpose, a platform emulating the biological characteristics of endomyocardial fibrosis was generated using a cell patterning technique to study morphological cellular changes in compact and irregular patterns of fibrosis. It was found that cellular patterns emulating the geometrical distributions of endomyocardial fibrosis generated morphological changes after interaction of the RL-14 cardiomyocytes with the 3T3 fibroblasts. Through this study, it was possible to evaluate biological characteristics such as cell proliferation, adhesion, and spatial distribution, which are directly related to the type of emulated endomyocardial fibrosis. This research concluded that fibroblasts inhibited the proliferation of cardiomyocytes via their interaction with specific microarchitectures. This behavior is consistent with the histopathological distribution of cardiac fibrosis; therefore, the platform developed in this research could be useful for the in vitro assessment of cellular microdomains. This would allow for the experimental determination of interactions with drugs, substrates, or biomaterials within the engineering of cardiac tissues.
Collapse
Affiliation(s)
- Paola Orozco
- Centro de Bioingeniería, Grupo de Dinámica Cardiovascular, Universidad Pontificia Bolivariana, Medellín, Colombia
| | - Yuliet Montoya
- Centro de Bioingeniería, Grupo de Dinámica Cardiovascular, Universidad Pontificia Bolivariana, Medellín, Colombia
- Comité de Trabajo de Bioingeniería Cardiovascular, Sociedad Colombiana de Cardiología y Cirugía Cardiovascular, Bogotá, Colombia
| | - John Bustamante
- Centro de Bioingeniería, Grupo de Dinámica Cardiovascular, Universidad Pontificia Bolivariana, Medellín, Colombia
- Comité de Trabajo de Bioingeniería Cardiovascular, Sociedad Colombiana de Cardiología y Cirugía Cardiovascular, Bogotá, Colombia
| |
Collapse
|
36
|
Bello AB, Kim D, Kim D, Park H, Lee SH. Engineering and Functionalization of Gelatin Biomaterials: From Cell Culture to Medical Applications. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:164-180. [PMID: 31910095 DOI: 10.1089/ten.teb.2019.0256] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Health care and medicine were revolutionized in recent years by the development of biomaterials, such as stents, implants, personalized drug delivery systems, engineered grafts, cell sheets, and other transplantable materials. These materials not only support the growth of cells before transplantation but also serve as replacements for damaged tissues in vivo. Among the various biomaterials available, those made from natural biological sources such as extracellular proteins (collagen, fibronectin, laminin) have shown significant benefits, and thus are widely used. However, routine biomaterial-based research requires copious quantities of proteins and the use of pure and intact extracellular proteins could be highly cost ineffective. Gelatin is a molecular derivative of collagen obtained through the irreversible denaturation of collagen proteins. Gelatin shares a very close molecular structure and function with collagen and thus is often used in cell and tissue culture to replace collagen for biomaterial purposes. Recent technological advancements such as additive manufacturing, rapid prototyping, and three-dimensional printing, in general, have resulted in great strides toward the generation of functional gelatin-based materials for medical purposes. In this review, the structural and molecular similarities of gelatin to other extracellular matrix proteins are compared and analyzed. Current strategies for gelatin crosslinking and production are described and recent applications of gelatin-based biomaterials in cell culture and tissue regeneration are discussed. Finally, recent improvements in gelatin-based biomaterials for medical applications and future directions are elaborated. Impact statement In this study, we described gelatin's biochemical properties and compared its advantages and drawbacks over other extracellular matrix proteins and polymers used for biomaterial application. We also described how gelatin can be used with other polymers in creating gelatin composite materials that have enhanced mechanical properties, increased biocompatibility, and boosted bioactivity, maximizing its benefits for biomedical purposes. The article is relevant, as it discussed not only the chemistry of gelatin, but also listed the current techniques in gelatin/biomaterial manufacturing and described the most recent trends in gelatin-based biomaterials for biomedical applications.
Collapse
Affiliation(s)
- Alvin Bacero Bello
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea.,Department of Biomedical Science, Dongguk University, Gyeonggi, Republic of Korea
| | - Deogil Kim
- Department of Biomedical Science, CHA University, Seongnam-Si, Republic of Korea
| | - Dohyun Kim
- Department of Biomedical Science, Dongguk University, Gyeonggi, Republic of Korea
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, Dongguk University, Gyeonggi, Republic of Korea
| |
Collapse
|
37
|
Schulte C. Cluster-assembled nanostructured materials for cell biology. CLUSTER BEAM DEPOSITION OF FUNCTIONAL NANOMATERIALS AND DEVICES 2020. [DOI: 10.1016/b978-0-08-102515-4.00010-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
38
|
Chen L, Li P, Lu X, Wang S, Zheng Z. Binary polymer brush patterns from facile initiator stickiness for cell culturing. Faraday Discuss 2019; 219:189-202. [PMID: 31317169 DOI: 10.1039/c9fd00013e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report a new initiator stickiness method to fabricate micropatterned binary polymer brush surfaces, which are ideal platforms for studying cell adhesion behavior. The atom transfer radical polymerization (ATRP) initiator, ω-mercaptoundecyl bromoisobutyrate (MUDBr), is found to adsorb on several hosting polymer brushes, including poly[oligo(ethylene glycol)methyl ether methacrylate] (POEGMA), poly(2-hydroxyethyl methacrylate) (PHEMA), and poly(glycidyl methacrylate) (PGMA) brushes. Based on the initiator stickiness, micropatterned initiator molecules are printed onto a layer of homogenous hosting polymer brushes via microcontact printing (μCP), and then, vertically, a patterned second layer of polymer brushes is grown from the initiator areas. With this simple, fast, and additive method, we demonstrate the fabrication of various binary polymer brushes, and show their applications for patterning cell microarrays and controlling cell orientation. This new approach to generating binary polymer brushes shows great potential for the manipulation of interfacial phenomena, facilitating a range of applications from semiconductors and lubrication to fundamental cell biology studies.
Collapse
Affiliation(s)
- Lina Chen
- Laboratory for Advanced Interfacial Materials and Devices, Institute of Textiles and Clothing, The Hong Kong Polytechnic University, Hong Kong SAR, P. R. China.
| | | | | | | | | |
Collapse
|
39
|
Ma X, Dewan S, Liu J, Tang M, Miller KL, Yu C, Lawrence N, McCulloch AD, Chen S. 3D printed micro-scale force gauge arrays to improve human cardiac tissue maturation and enable high throughput drug testing. Acta Biomater 2019; 95:319-327. [PMID: 30576862 PMCID: PMC6584548 DOI: 10.1016/j.actbio.2018.12.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/11/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022]
Abstract
Human induced pluripotent stem cell - derived cardiomyocytes (iPSC-CMs) are regarded as a promising cell source for establishing in-vitro personalized cardiac tissue models and developing therapeutics. However, analyzing cardiac force and drug response using mature human iPSC-CMs in a high-throughput format still remains a great challenge. Here we describe a rapid light-based 3D printing system for fabricating micro-scale force gauge arrays suitable for 24-well and 96-well plates that enable scalable tissue formation and measurement of cardiac force generation in human iPSC-CMs. We demonstrate consistent tissue band formation around the force gauge pillars with aligned sarcomeres. Among the different maturation treatment protocols we explored, 3D aligned cultures on force gauge arrays with in-culture pacing produced the highest expression of mature cardiac marker genes. We further demonstrated the utility of these micro-tissues to develop significantly increased contractile forces in response to treatment with isoproterenol, levosimendan, and omecamtiv mecarbil. Overall, this new 3D printing system allows for high flexibility in force gauge design and can be optimized to achieve miniaturization and promote cardiac tissue maturation with great potential for high-throughput in-vitro drug screening applications. STATEMENT OF SIGNIFICANCE: The application of iPSC-derived cardiac tissues in translatable drug screening is currently limited by the challenges in forming mature cardiac tissue and analyzing cardiac forces in a high-throughput format. We demonstrate the use of a rapid light-based 3D printing system to build a micro-scale force gauge array that enables scalable cardiac tissue formation from iPSC-CMs and measurement of contractile force development. With the capability to provide great flexibility over force gauge design as well as optimization to achieve miniaturization, our 3D printing system serves as a promising tool to build cardiac tissues for high-throughput in-vitro drug screening applications.
Collapse
Affiliation(s)
- Xuanyi Ma
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Sukriti Dewan
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Justin Liu
- Department of Materials Science and Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Min Tang
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kathleen L Miller
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Claire Yu
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Natalie Lawrence
- Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Shaochen Chen
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Materials Science and Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of NanoEngineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Chemical Engineering Program, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
40
|
Li N, Xie T, Sun Y. Towards organogenesis and morphogenesis in vitro: harnessing engineered microenvironment and autonomous behaviors of pluripotent stem cells. Integr Biol (Camb) 2019; 10:574-586. [PMID: 30225509 DOI: 10.1039/c8ib00116b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Recently, researchers have been attempting to control pluripotent stem cell fate or generate self-organized tissues from stem cells. Advances in bioengineering enable generation of organotypic structures, which capture the cellular components, spatial cell organization and even some functions of tissues or organs in development. However, only a few engineering tools have been utilized to regulate the formation and organization of spatially complex tissues derived from stem cells. Here, we provide a review of recent progress in the culture of organotypic structures in vitro, focusing on how microengineering approaches including geometric confinement, extracellular matrix (ECM) property modulation, spatially controlled biochemical factors, and external forces, can be utilized to generate organotypic structures. Moreover, we will discuss potential technologies that can be applied to further control both soluble and insoluble factors spatiotemporally in vitro. In summary, advanced engineered approaches have a great promise in generating miniaturized tissues and organs in a reproducible fashion, facilitating the cellular and molecular understanding of embryogenesis and morphogenesis processes.
Collapse
Affiliation(s)
- Ningwei Li
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, Massachusetts 01003, USA.
| | | | | |
Collapse
|
41
|
Buskermolen ABC, Suresh H, Shishvan SS, Vigliotti A, DeSimone A, Kurniawan NA, Bouten CVC, Deshpande VS. Entropic Forces Drive Cellular Contact Guidance. Biophys J 2019; 116:1994-2008. [PMID: 31053262 PMCID: PMC6531843 DOI: 10.1016/j.bpj.2019.04.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 12/30/2022] Open
Abstract
Contact guidance-the widely known phenomenon of cell alignment induced by anisotropic environmental features-is an essential step in the organization of adherent cells, but the mechanisms by which cells achieve this orientational ordering remain unclear. Here, we seeded myofibroblasts on substrates micropatterned with stripes of fibronectin and observed that contact guidance emerges at stripe widths much greater than the cell size. To understand the origins of this surprising observation, we combined morphometric analysis of cells and their subcellular components with a, to our knowledge, novel statistical framework for modeling nonthermal fluctuations of living cells. This modeling framework is shown to predict not only the trends but also the statistical variability of a wide range of biological observables, including cell (and nucleus) shapes, sizes, and orientations, as well as stress-fiber arrangements within the cells with remarkable fidelity with a single set of cell parameters. By comparing observations and theory, we identified two regimes of contact guidance: 1) guidance on stripe widths smaller than the cell size (w ≤ 160 μm), which is accompanied by biochemical changes within the cells, including increasing stress-fiber polarization and cell elongation; and 2) entropic guidance on larger stripe widths, which is governed by fluctuations in the cell morphology. Overall, our findings suggest an entropy-mediated mechanism for contact guidance associated with the tendency of cells to maximize their morphological entropy through shape fluctuations.
Collapse
Affiliation(s)
- Antonetta B C Buskermolen
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, North Brabant, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Hamsini Suresh
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Siamak S Shishvan
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom; Department of Structural Engineering, University of Tabriz, Tabriz, East Azarbayjan, Iran
| | - Andrea Vigliotti
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom; Innovative Materials, Italian Aerospace Research Center, Capua, Caserta, Italy
| | - Antonio DeSimone
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom; The BioRobotics Institute, Scuola Superiore Sant'Anna, Pontedera, Pisa, Italy; SISSA - International School for Advanced Studies, Trieste, Italy
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, North Brabant, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, North Brabant, Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Vikram S Deshpande
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
42
|
Ng FL, Ong YO, Chen HZ, Tran LQN, Cao Y, Tay BY, Tan LP. A facile method for fabricating a three-dimensional aligned fibrous scaffold for vascular application. RSC Adv 2019; 9:13054-13064. [PMID: 35520779 PMCID: PMC9063778 DOI: 10.1039/c9ra00661c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/05/2019] [Indexed: 01/03/2023] Open
Abstract
Collection of circumferentially aligned and 3D fibrous scaffold on a newly designed electrospinning auxiliary jig. The aligned fibres served as a signaling modality to induce cell alignment and the maintenance of a contractile phenotype for hSMCs.
Collapse
Affiliation(s)
- Feng Lin Ng
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
- Singapore Institute of Manufacturing Technology
| | - Yee Oon Ong
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
| | - Hui Zhi Chen
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
| | - Le Quan Ngoc Tran
- Singapore Institute of Manufacturing Technology
- Singapore 637662
- Singapore
| | - Ye Cao
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
| | - Bee Yen Tay
- Singapore Institute of Manufacturing Technology
- Singapore 637662
- Singapore
| | - Lay Poh Tan
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore 639798
- Singapore
| |
Collapse
|
43
|
Schulte C, Lamanna J, Moro AS, Piazzoni C, Borghi F, Chighizola M, Ortoleva S, Racchetti G, Lenardi C, Podestà A, Malgaroli A, Milani P. Neuronal Cells Confinement by Micropatterned Cluster-Assembled Dots with Mechanotransductive Nanotopography. ACS Biomater Sci Eng 2018; 4:4062-4075. [DOI: 10.1021/acsbiomaterials.8b00916] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Carsten Schulte
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Department of Physics, Università degli Studi di Milano, Milano, Italy
| | - Jacopo Lamanna
- Center for Behavioral Neuroscience and Communication (BNC), Università Vita-Salute San Raffaele and Neurobiology of Learning Unit, Division of Neuroscience, Scientific
Institute San Raffaele, Milano, Italy
| | - Andrea Stefano Moro
- Center for Behavioral Neuroscience and Communication (BNC), Università Vita-Salute San Raffaele and Neurobiology of Learning Unit, Division of Neuroscience, Scientific
Institute San Raffaele, Milano, Italy
| | - Claudio Piazzoni
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Department of Physics, Università degli Studi di Milano, Milano, Italy
| | - Francesca Borghi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Department of Physics, Università degli Studi di Milano, Milano, Italy
| | - Matteo Chighizola
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Department of Physics, Università degli Studi di Milano, Milano, Italy
| | - Serena Ortoleva
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Department of Physics, Università degli Studi di Milano, Milano, Italy
| | - Gabriella Racchetti
- Center for Behavioral Neuroscience and Communication (BNC), Università Vita-Salute San Raffaele and Neurobiology of Learning Unit, Division of Neuroscience, Scientific
Institute San Raffaele, Milano, Italy
| | - Cristina Lenardi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Department of Physics, Università degli Studi di Milano, Milano, Italy
| | - Alessandro Podestà
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Department of Physics, Università degli Studi di Milano, Milano, Italy
| | - Antonio Malgaroli
- Center for Behavioral Neuroscience and Communication (BNC), Università Vita-Salute San Raffaele and Neurobiology of Learning Unit, Division of Neuroscience, Scientific
Institute San Raffaele, Milano, Italy
| | - Paolo Milani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (CIMaINa) and Department of Physics, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
44
|
Impact of Three-Dimentional Culture Systems on Hepatic Differentiation of Puripotent Stem Cells and Beyond. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018. [PMID: 30357683 DOI: 10.1007/978-981-13-0947-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Generation of functional hepatocytes from human pluripotent stem cells (hPSCs) is a vital tool to produce large amounts of human hepatocytes, which hold a great promise for biomedical and regenerative medicine applications. Despite a tremendous progress in developing the differentiation protocols recapitulating the developmental signalling and stages, these resulting hepatocytes from hPSCs yet achieve maturation and functionality comparable to those primary hepatocytes. The absence of 3D milieu in the culture and differentiation of these hepatocytes may account for this, at least partly, thus developing an optimal 3D culture could be a step forward to achieve this aim. Hence, review focuses on current development of 3D culture systems for hepatic differentiation and maturation and the future perspectives of its application.
Collapse
|
45
|
Chou WC, Wang RCC, Huang CL, Lee TM. The effect of plasma treatment on the osseointegration of rough titanium implant: A histo-morphometric study in rabbits. J Dent Sci 2018; 13:267-273. [PMID: 30895131 PMCID: PMC6388852 DOI: 10.1016/j.jds.2018.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/28/2018] [Indexed: 11/27/2022] Open
Abstract
Abstract Background/purpose The surface properties, such as hydrophilicity and functional OH groups, play an important role in bone fixation in vivo. In our previous study, the plasma treatments of large grit and acid etching (SLA) method produce functional OH groups on the rough surface. There is no report in discussing the integration between basic Ti—OH groups and bone-to-implant contact (BIC). The aim of this study was to evaluate the effect of the functional OH groups on the rough surface both in vitro and in vivo. Materials and methods Functional hydroxyl groups were produced on a SLA-treated surface. The surface topography, roughness, wettability, and chemical composition were examined using various techniques. Twenty-four implants were inserted into the proximal tibia of four New Zealand white rabbits. The biological responses were measured in terms of histomorphometric analysis 4 and 8 weeks post-implantation. Results The surface morphology and roughness were similar among all groups. However, the concentration of OH groups and hydrophilicity were found increased in the plasma treatment. The cell morphology in RF-plasma treated groups had more polygonal type and higher expression of actin and vinculin. The bone-to-implant contact (BIC) ratios of RF-200W were significantly higher than other groups (P < 0.05). The relationship between basic OH groups and BIC showed linear correspondence. Conclusion The Ti—OH groups introduced on the rough surface by plasma treatments can trigger cell adhesion which further initiate new bone apposition. We propose that RF-plasma treatment can help to enhance bone healing at 4 and 8 weeks.
Collapse
Affiliation(s)
- Wan-Ching Chou
- Institute of Manufacturing Information and Systems, National Cheng Kung University, Tainan, Taiwan
| | - Rex C-C Wang
- Institute of Manufacturing Information and Systems, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Ling Huang
- Center for Fundamental Science, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzer-Min Lee
- Institute of Oral Medicine, National Cheng Kung University Medical College, Tainan, Taiwan.,School of Dentistry, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
46
|
Gorkun AA, Shpichka AI, Zurina IM, Koroleva AV, Kosheleva NV, Nikishin DA, Butnaru DV, Timashev PS, Repin VS, Saburina IN. Angiogenic potential of spheroids from umbilical cord and adipose-derived multipotent mesenchymal stromal cells within fibrin gel. Biomed Mater 2018; 13:044108. [DOI: 10.1088/1748-605x/aac22d] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
47
|
A triple co-culture method to investigate the effect of macrophages and fibroblasts on myoblast proliferation and migration. Biotechniques 2018; 64:52-58. [PMID: 29571282 DOI: 10.2144/btn-2017-0100] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/11/2018] [Indexed: 11/23/2022] Open
Abstract
The communication between nonmyogenic cells, such as macrophages and fibroblasts, and myoblasts is crucial for successful skeletal muscle repair. In vitro co-culture methods can be used to increase our understanding of these cellular interactions; however, current protocols are restricted to two, often physically separate, cell populations. Here, we demonstrate a novel, inexpensive in vitro triple co-culture method that facilitates the co-culture of at least three cell populations with some degree of cell-cell contact. Using this method, we determined the effect of macrophages and fibroblasts on myoblast proliferation and migration. A significant increase in myoblast proliferation and migration was observed following co-culture with either macrophages or fibroblasts. However, triple co-culture of macrophages, fibroblasts, and myoblasts revealed that the presence of macrophages prevented fibroblasts from maintaining this positive effect on myoblast migration. Macrophages, on the other hand, continued to promote myoblast proliferation whether in the presence of fibroblasts or not. Our triple co-culture system highlights the significance of multicellular communication in regulating myoblast proliferation and migration and emphasizes the importance of more complex co-culture systems when investigating myogenesis in vitro.
Collapse
|
48
|
Fernandez A, Bautista M, Stanciauskas R, Chung T, Pinaud F. Cell-Shaping Micropatterns for Quantitative Super-Resolution Microscopy Imaging of Membrane Mechanosensing Proteins. ACS APPLIED MATERIALS & INTERFACES 2017; 9:27575-27586. [PMID: 28766344 DOI: 10.1021/acsami.7b09743] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Patterning cells on microcontact-printed substrates is a powerful approach to control cell morphology and introduce specific mechanical cues on a cell's molecular organization. Although global changes in cellular architectures caused by micropatterns can easily be probed with diffraction-limited optical microscopy, studying molecular reorganizations at the nanoscale demands micropatterned substrates that accommodate the optical requirements of single molecule microscopy techniques. Here, we developed a simple micropatterning strategy that provides control of cellular architectures and is optimized for nanometer accuracy single molecule tracking and three-dimensional super-resolution imaging of plasma and nuclear membrane proteins in cells. This approach, based on fibronectin microcontact printing on hydrophobic organosilane monolayers, allows evanescent wave and light-sheet microscopy of cells whilst fulfilling the stringent optical demands of point reconstruction optical microscopy. By imposing steady-state mechanical cues on cells grown in these micropatterns, we reveal nanoscale remodeling in the dynamics and the structural organizations of the nuclear envelope mechanotransducing protein emerin and of the plasma membrane mechanosensing protein caveolin-1 using single particle tracking photoactivated localization microscopy and direct stochastic optical reconstruction microscopy imaging. In addition to allowing quantitative biophysical studies of mechanoresponsive membrane proteins, this approach provides an easy means to probe mechanical regulations in cellular membranes with high optical resolution and nanometer precision.
Collapse
Affiliation(s)
- Anthony Fernandez
- Department of Biological Sciences, ‡Department of Chemistry, and §Department of Physics and Astronomy, University of Southern California , Los Angeles, California 90089, United States
| | - Markville Bautista
- Department of Biological Sciences, ‡Department of Chemistry, and §Department of Physics and Astronomy, University of Southern California , Los Angeles, California 90089, United States
| | - Ramunas Stanciauskas
- Department of Biological Sciences, ‡Department of Chemistry, and §Department of Physics and Astronomy, University of Southern California , Los Angeles, California 90089, United States
| | - Taerin Chung
- Department of Biological Sciences, ‡Department of Chemistry, and §Department of Physics and Astronomy, University of Southern California , Los Angeles, California 90089, United States
| | - Fabien Pinaud
- Department of Biological Sciences, ‡Department of Chemistry, and §Department of Physics and Astronomy, University of Southern California , Los Angeles, California 90089, United States
| |
Collapse
|
49
|
Abstract
There is no good science in bad models. Cell culture is especially prone to artifacts. A number of novel cell culture technologies have become more broadly available in the 21st century, which allow overcoming limitations of traditional culture and are more physiologically relevant. These include the use of stem-cell derived human cells, cocultures of different cell types, scaffolds and extracellular matrices, perfusion platforms (such as microfluidics), 3D culture, organ-on-chip technologies, tissue architecture, and organ functionality. The physiological relevance of such models is further enhanced by the measurement of biomarkers (e.g., key events of pathways), organ specific functionality, and more comprehensive assessment cell responses by high-content methods. These approaches are still rarely combined to create microphysiological systems. The complexity of the combination of these technologies can generate results closer to the in vivo situation but increases the number of parameters to control, bringing some new challenges. In fact, we do not argue that all cell culture needs to be that sophisticated. The efforts taken are determined by the purpose of our experiments and tests. If only a very specific molecular target to cell response is of interest, a very simple model, which reflects this, might be much more suited to allow standardization and high-throughput. However, the less defined the end point of interest and cellular response are, the better we should approximate organ- or tissue-like culture conditions to make physiological responses more probable. Besides these technologic advances, important progress in the quality assurance and reporting on cell cultures as well as the validation of cellular test systems brings the utility of cell cultures to a new level. The advancement and broader implementation of Good Cell Culture Practice (GCCP) is key here. In toxicology, this is a major prerequisite for meaningful and reliable results, ultimately supporting risk assessment and product development decisions.
Collapse
Affiliation(s)
- David Pamies
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland 21205, United States
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland 21205, United States.,CAAT-Europe, University of Konstanz , 78464 Konstanz, Germany
| |
Collapse
|
50
|
Ruprecht V, Monzo P, Ravasio A, Yue Z, Makhija E, Strale PO, Gauthier N, Shivashankar GV, Studer V, Albiges-Rizo C, Viasnoff V. How cells respond to environmental cues - insights from bio-functionalized substrates. J Cell Sci 2016; 130:51-61. [PMID: 27856508 DOI: 10.1242/jcs.196162] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Biomimetic materials have long been the (he)art of bioengineering. They usually aim at mimicking in vivo conditions to allow in vitro culture, differentiation and expansion of cells. The past decade has witnessed a considerable amount of progress in soft lithography, bio-inspired micro-fabrication and biochemistry, allowing the design of sophisticated and physiologically relevant micro- and nano-environments. These systems now provide an exquisite toolbox with which we can control a large set of physicochemical environmental parameters that determine cell behavior. Bio-functionalized surfaces have evolved from simple protein-coated solid surfaces or cellular extracts into nano-textured 3D surfaces with controlled rheological and topographical properties. The mechanobiological molecular processes by which cells interact and sense their environment can now be unambiguously understood down to the single-molecule level. This Commentary highlights recent successful examples where bio-functionalized substrates have contributed in raising and answering new questions in the area of extracellular matrix sensing by cells, cell-cell adhesion and cell migration. The use, the availability, the impact and the challenges of such approaches in the field of biology are discussed.
Collapse
Affiliation(s)
- Verena Ruprecht
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona 08003, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | | | - Andrea Ravasio
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 117411, Singapore
| | - Zhang Yue
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 117411, Singapore
| | - Ekta Makhija
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 117411, Singapore
| | - Pierre Olivier Strale
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux F-33000, France
| | | | - G V Shivashankar
- IFOM, Via Adamello, 16, Milano 20139, Italy.,Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 117411, Singapore
| | - Vincent Studer
- CNRS, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux F-33000, France
| | - Corinne Albiges-Rizo
- INSERM, U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Institute Albert Bonniot, University Grenoble Alpes, La Tronche F-38700, France
| | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, 117411, Singapore .,CNRS UMI 3639, 5A Engineering Drive 1, 117411 Singapore
| |
Collapse
|