1
|
Wu D, Gong T, Sun Z, Yao X, Wang D, Chen Q, Guo Q, Li X, Guo Y, Lu Y. Dual-crosslinking gelatin-hyaluronic acid methacrylate based biomimetic PDAC desmoplastic niche enhances tumor-associated macrophages recruitment and M2-like polarization. Int J Biol Macromol 2024; 269:131826. [PMID: 38679256 DOI: 10.1016/j.ijbiomac.2024.131826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/18/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
The tumor microenvironment (TME) of pancreatic ductal adenocarcinoma (PDAC) is characterized by deposition of desmoplastic matrix (including collagen and hyaluronic acid). And the interactions between tumor-associated macrophages (TAMs) and tumor cells play a crucial role in progression of PDAC. Hence, the appropriate model of tumor cell-macrophage interaction within the unique PDAC TME is of significantly important. To this end, a 3D tumor niche based on dual-crosslinking gelatin methacrylate and hyaluronic acid methacrylate hydrogels was constructed to simulate the desmoplastic tumor matrix with matching compressive modulus and composition. The bionic 3D tumor niche creates an immunosuppressive microenvironment characterized by the downregulation of M1 markers and upregulation of M2 markers in TAMs. Mechanistically, RNA-seq analysis revealed that the PI3K-AKT signaling pathway might modulate the phenotypic balance and recruitment of macrophages through regulating SELE and VCAM-1. Furthermore, GO and GSEA revealed the biological process of leukocyte migration and the activation of cytokine-associated signaling were involved. Finally, the 3D tumor-macrophage niches with three different ratios were fabricated which displayed increased M2-like polarization and stemness. The utilization of the 3D tumor niche has the potential to provide a more accurate investigation of the interplay between PDAC tumor cells and macrophages within an in vivo setting.
Collapse
Affiliation(s)
- Di Wu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, PR China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu province 226001, PR China
| | - Tiancheng Gong
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, PR China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu province 226001, PR China
| | - Zhongxiang Sun
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu province 226001, PR China
| | - Xihao Yao
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, PR China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu province 226001, PR China
| | - Dongzhi Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, PR China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu province 226001, PR China
| | - Qiyang Chen
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, PR China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu province 226001, PR China
| | - Qingsong Guo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, PR China
| | - Xiaohong Li
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu province 226001, PR China
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu province 226001, PR China.
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, PR China.
| |
Collapse
|
2
|
Gomila Pelegri N, Stanczak AM, Bottomley AL, Cummins ML, Milthorpe BK, Gorrie CA, Padula MP, Santos J. Neural Marker Expression in Adipose-Derived Stem Cells Grown in PEG-Based 3D Matrix Is Enhanced in the Presence of B27 and CultureOne Supplements. Int J Mol Sci 2023; 24:16269. [PMID: 38003460 PMCID: PMC10671562 DOI: 10.3390/ijms242216269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) have incredible potential as an avenue to better understand and treat neurological disorders. While they have been successfully differentiated into neural stem cells and neurons, most such protocols involve 2D environments, which are not representative of in vivo physiology. In this study, human ADSCs were cultured in 1.1 kPa polyethylene-glycol 3D hydrogels for 10 days with B27, CultureOne (C1), and N2 neural supplements to examine the neural differentiation potential of ADSCs using both chemical and mechanical cues. Following treatment, cell viability, proliferation, morphology, and proteome changes were assessed. Results showed that cell viability was maintained during treatments, and while cells continued to proliferate over time, proliferation slowed down. Morphological changes between 3D untreated cells and treated cells were not observed. However, they were observed among 2D treatments, which exhibited cellular elongation and co-alignment. Proteome analysis showed changes consistent with early neural differentiation for B27 and C1 but not N2. No significant changes were detected using immunocytochemistry, potentially indicating a greater differentiation period was required. In conclusion, treatment of 3D-cultured ADSCs in PEG-based hydrogels with B27 and C1 further enhances neural marker expression, however, this was not observed using supplementation with N2.
Collapse
Affiliation(s)
- Neus Gomila Pelegri
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
- Neural Injury Research Unit, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Aleksandra M. Stanczak
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (A.M.S.); (M.P.P.)
| | - Amy L. Bottomley
- Microbial Imaging Facility, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Max L. Cummins
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Ultimo, NSW 2007, Australia;
- The Australian Centre for Genomic Epidemiological Microbiology, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Bruce K. Milthorpe
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
| | - Catherine A. Gorrie
- Neural Injury Research Unit, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Matthew P. Padula
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (A.M.S.); (M.P.P.)
| | - Jerran Santos
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
| |
Collapse
|
3
|
Kasahara K, Muramatsu J, Kurashina Y, Miura S, Miyata S, Onoe H. Spatiotemporal single-cell tracking analysis in 3D tissues to reveal heterogeneous cellular response to mechanical stimuli. SCIENCE ADVANCES 2023; 9:eadf9917. [PMID: 37831766 PMCID: PMC10575577 DOI: 10.1126/sciadv.adf9917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 09/08/2023] [Indexed: 10/15/2023]
Abstract
Mechanical stimuli have been recognized as important for tissue maturation, homeostasis and constructing engineered three-dimensional (3D) tissues. However, we know little about the cellular mechanical response in tissues that could be considerably heterogeneous and spatiotemporally dynamic due to the complex structure of tissues. Here, we report a spatiotemporal single-cell tracking analysis of in vitro 3D tissues under mechanical stretch, to reveal the heterogeneous cellular behavior by using a developed stretch and optical live imaging system. The system could affect the cellular orientation and directly measure the distance of cells in in vitro 3D myoblast tissues (3DMTs) at the single-cell level. Moreover, we observed the spatiotemporal heterogeneous cellular locomotion and shape changes under mechanical stretch in 3DMTs. This single-cell tracking analysis can become a principal method to investigate the heterogeneous cellular response in tissues and provide insights that conventional analyses have not yet offered.
Collapse
Affiliation(s)
- Keitaro Kasahara
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Jumpei Muramatsu
- School of Integrated Design Engineering, Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Yuta Kurashina
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
- Division of Advanced Mechanical Systems Engineering, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan
| | - Shigenori Miura
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shogo Miyata
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| | - Hiroaki Onoe
- Department of Mechanical Engineering, Faculty of Science and Technology, Keio University, 3-14-1 Hiyoshi, Kohoku-ku, Yokohama 223-8522, Japan
| |
Collapse
|
4
|
Gomila Pelegri N, Stanczak AM, Bottomley AL, Milthorpe BK, Gorrie CA, Padula MP, Santos J. Adipose-Derived Stem Cells Spontaneously Express Neural Markers When Grown in a PEG-Based 3D Matrix. Int J Mol Sci 2023; 24:12139. [PMID: 37569515 PMCID: PMC10418654 DOI: 10.3390/ijms241512139] [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: 07/03/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Neurological diseases are among the leading causes of disability and death worldwide and remain difficult to treat. Tissue engineering offers avenues to test potential treatments; however, the development of biologically accurate models of brain tissues remains challenging. Given their neurogenic potential and availability, adipose-derived stem cells (ADSCs) are of interest for creating neural models. While progress has been made in differentiating ADSCs into neural cells, their differentiation in 3D environments, which are more representative of the in vivo physiological conditions of the nervous system, is crucial. This can be achieved by modulating the 3D matrix composition and stiffness. Human ADSCs were cultured for 14 days in a 1.1 kPa polyethylene glycol-based 3D hydrogel matrix to assess effects on cell morphology, cell viability, proteome changes and spontaneous neural differentiation. Results showed that cells continued to proliferate over the 14-day period and presented a different morphology to 2D cultures, with the cells elongating and aligning with one another. The proteome analysis revealed 439 proteins changed in abundance by >1.5 fold. Cyclic nucleotide 3'-phosphodiesterase (CNPase) markers were identified using immunocytochemistry and confirmed with proteomics. Findings indicate that ADSCs spontaneously increase neural marker expression when grown in an environment with similar mechanical properties to the central nervous system.
Collapse
Affiliation(s)
- Neus Gomila Pelegri
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
- Neural Injury Research Unit, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Aleksandra M. Stanczak
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (A.M.S.); (M.P.P.)
| | - Amy L. Bottomley
- Microbial Imaging Facility, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Bruce K. Milthorpe
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
| | - Catherine A. Gorrie
- Neural Injury Research Unit, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Matthew P. Padula
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (A.M.S.); (M.P.P.)
| | - Jerran Santos
- Advanced Tissue Engineering and Stem Cell Biology Group, School of Life Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia; (N.G.P.); (B.K.M.)
| |
Collapse
|
5
|
Effect of Cyclic Uniaxial Mechanical Strain on Endothelial Progenitor Cell Differentiation. Cardiovasc Eng Technol 2022; 13:872-885. [PMID: 35501625 DOI: 10.1007/s13239-022-00623-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/30/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE Endothelial progenitor cells (EPCs) have been used as an autologous or allogeneic source in multiple tissue engineering applications. EPCs possess high proliferative and tissue regeneration potential. The effect of shear stress on EPCs has been extensively studied but the role of cyclic mechanical strain on EPCs remains to be understood. In this study, we focused on examining the role of uniaxial cyclic strain on EPCs cultured on three-dimensional (3D) anisotropic composites that mimic healthy and diseased aortic valve tissue matrix compositions. METHODS AND RESULTS The composites were fabricated by combining centrifugal jet spun fibers with photocrosslinkable gelatin and glycosaminoglycan hydrogels. A custom-designed uniaxial cyclic stretcher was used to provide the necessary cyclic stimulation to the EPC-seeded 3D composites. The samples were cyclically strained at a rate of 1 Hz at 15% strain mimicking the physiological condition experienced by aortic valve, with static conditions serving as controls. Cell viability was high in all conditions. Immunostaining revealed reduced endothelial marker (CD31) expression with increased smooth muscle cell marker, SM22α, expression when subjected to cyclic strain. Functional analysis through Matrigel assay agreed with the immunostaining findings with reduced tubular structure formation in strained conditions compared to EPC controls. Additionally, the cells showed reduced acLDL uptake compared to controls which are in alignment with the EPCs undergoing differentiation. CONCLUSION Overall, we show that EPCs lose their endothelial progenitor phenotype, and have the potential to be differentiated into mesenchymal-like cells through cyclic mechanical stimulation.
Collapse
|
6
|
Friedland F, Babu S, Springer R, Konrad J, Herfs Y, Gerlach S, Gehlen J, Krause HJ, De Laporte L, Merkel R, Noetzel E. ECM-transmitted shear stress induces apoptotic cell extrusion in early breast gland development. Front Cell Dev Biol 2022; 10:947430. [PMID: 36105352 PMCID: PMC9465044 DOI: 10.3389/fcell.2022.947430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial cells of human breast glands are exposed to various mechanical ECM stresses that regulate tissue development and homeostasis. Mechanoadaptation of breast gland tissue to ECM-transmitted shear stress remained poorly investigated due to the lack of valid experimental approaches. Therefore, we created a magnetic shear strain device that enabled, for the first time, to analyze the instant shear strain response of human breast gland cells. MCF10A-derived breast acini with basement membranes (BM) of defined maturation state and basoapical polarization were used to resemble breast gland morphogenesis in vitro. The novel biophysical tool was used to apply cyclic shear strain with defined amplitudes (≤15%, 0.2 Hz) over 22 h on living spheroids embedded in an ultrasoft matrix (<60 Pa). We demonstrated that breast spheroids gain resistance to shear strain, which increased with BM maturation and basoapical polarization. Most intriguingly, poorly developed spheroids were prone to cyclic strain-induced extrusion of apoptotic cells from the spheroid body. In contrast, matured spheroids were insensitive to this mechanoresponse—indicating changing mechanosensing or mechanotransduction mechanisms during breast tissue morphogenesis. Together, we introduced a versatile tool to study cyclic shear stress responses of 3D cell culture models. It can be used to strain, in principle, all kinds of cell clusters, even those that grow only in ultrasoft hydrogels. We believe that this approach opens new doors to gain new insights into dynamic shear strain-induced mechanobiological regulation circuits between cells and their ECM.
Collapse
Affiliation(s)
- F. Friedland
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, Jülich, Germany
| | - S. Babu
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry (ITMC), Polymeric Biomaterials, RWTH University Aachen, Aachen, Germany
| | - R. Springer
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, Jülich, Germany
| | - J. Konrad
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, Jülich, Germany
| | - Y. Herfs
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, Jülich, Germany
| | - S. Gerlach
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, Jülich, Germany
| | - J. Gehlen
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, Jülich, Germany
| | - H.-J. Krause
- Institute of Biological Information Processing 3 (IBI-3): Bioelectronics, Forschungszentrum Jülich, Jülich, Germany
| | - L. De Laporte
- DWI-Leibniz Institute for Interactive Materials, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry (ITMC), Polymeric Biomaterials, RWTH University Aachen, Aachen, Germany
- Advanced Materials for Biomedicine (AMB), Institute of Applied Medical Engineering (AME), University Hospital RWTH Aachen, Center for Biohybrid Medical Systems (CMBS), Aachen, Germany
| | - R. Merkel
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, Jülich, Germany
| | - E. Noetzel
- Institute of Biological Information Processing 2 (IBI-2): Mechanobiology, Forschungszentrum Jülich, Jülich, Germany
- *Correspondence: E. Noetzel,
| |
Collapse
|
7
|
Blood-Vessel-Inspired Hierarchical Trilayer Scaffolds: PCL/Gelatin-Driven Protein Adsorption and Cellular Interaction. Polymers (Basel) 2022; 14:polym14112135. [PMID: 35683808 PMCID: PMC9182901 DOI: 10.3390/polym14112135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 12/23/2022] Open
Abstract
Fabrication of scaffolds with hierarchical structures exhibiting the blood vessel topological and biochemical features of the native extracellular matrix that maintain long-term patency remains a major challenge. Within this context, scaffold assembly using biodegradable synthetic polymers (BSPs) via electrospinning had led to soft-tissue-resembling microstructures that allow cell infiltration. However, BSPs fail to exhibit the sufficient surface reactivity, limiting protein adsorption and/or cell adhesion and jeopardizing the overall graft performance. Here, we present a methodology for the fabrication of three-layered polycaprolactone (PCL)-based tubular structures with biochemical cues to improve protein adsorption and cell adhesion. For this purpose, PCL was backbone-oxidized (O-PCL) and cast over a photolithography-manufactured microgrooved mold to obtain a bioactive surface as demonstrated using a protein adsorption assay (BSA), Fourier transform infrared spectroscopy (FTIR) and calorimetric analyses. Then, two layers of PCL:gelatin (75:25 and 95:5 w/w), obtained using a novel single-desolvation method, were electrospun over the casted O-PCL to mimic a vascular wall with a physicochemical gradient to guide cell adhesion. Furthermore, tensile properties were shown to withstand the physiological mechanical stresses and strains. In vitro characterization, using L929 mouse fibroblasts, demonstrated that the multilayered scaffold is a suitable platform for cell infiltration and proliferation from the innermost to the outermost layer as is needed for vascular wall regeneration. Our work holds promise as a strategy for the low-cost manufacture of next-generation polymer-based hierarchical scaffolds with high bioactivity and resemblance of ECM’s microstructure to accurately guide cell attachment and proliferation.
Collapse
|
8
|
Cho J, Lee H, Rah W, Chang HJ, Yoon YS. From engineered heart tissue to cardiac organoid. Theranostics 2022; 12:2758-2772. [PMID: 35401829 PMCID: PMC8965483 DOI: 10.7150/thno.67661] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 03/01/2022] [Indexed: 12/03/2022] Open
Abstract
The advent of human pluripotent stem cells (hPSCs) presented a new paradigm to employ hPSC-derived cardiomyocytes (hPSC-CMs) in drug screening and disease modeling. However, hPSC-CMs differentiated in conventional two-dimensional systems are structurally and functionally immature. Moreover, these differentiation systems generate predominantly one type of cell. Since the heart includes not only CMs but other cell types, such monolayer cultures have limitations in simulating the native heart. Accordingly, three-dimensional (3D) cardiac tissues have been developed as a better platform by including various cardiac cell types and extracellular matrices. Two advances were made for 3D cardiac tissue generation. One type is engineered heart tissues (EHTs), which are constructed by 3D cell culture of cardiac cells using an engineering technology. This system provides a convenient real-time analysis of cardiac function, as well as a precise control of the input/output flow and mechanical/electrical stimulation. The other type is cardiac organoids, which are formed through self-organization of differentiating cardiac lineage cells from hPSCs. While mature cardiac organoids are more desirable, at present only primitive forms of organoids are available. In this review, we discuss various models of hEHTs and cardiac organoids emulating the human heart, focusing on their unique features, utility, and limitations.
Collapse
Affiliation(s)
- Jaeyeaon Cho
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyein Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Woongchan Rah
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyuk Jae Chang
- Division of Cardiology, Department of Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Young-sup Yoon
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Karis Bio Inc., Seoul, Republic of Korea
| |
Collapse
|
9
|
Özkan H, Öztürk DG, Korkmaz G. Transcriptional Factor Repertoire of Breast Cancer in 3D Cell Culture Models. Cancers (Basel) 2022; 14:cancers14041023. [PMID: 35205770 PMCID: PMC8870600 DOI: 10.3390/cancers14041023] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Knowledge of the transcriptional regulation of breast cancer tumorigenesis is largely based on studies performed in two-dimensional (2D) monolayer culture models, which lack tissue architecture and therefore fail to represent tumor heterogeneity. However, three-dimensional (3D) cell culture models are better at mimicking in vivo tumor microenvironment, which is critical in regulating cellular behavior. Hence, 3D cell culture models hold great promise for translational breast cancer research. Abstract Intratumor heterogeneity of breast cancer is driven by extrinsic factors from the tumor microenvironment (TME) as well as tumor cell–intrinsic parameters including genetic, epigenetic, and transcriptomic traits. The extracellular matrix (ECM), a major structural component of the TME, impacts every stage of tumorigenesis by providing necessary biochemical and biomechanical cues that are major regulators of cell shape/architecture, stiffness, cell proliferation, survival, invasion, and migration. Moreover, ECM and tissue architecture have a profound impact on chromatin structure, thereby altering gene expression. Considering the significant contribution of ECM to cellular behavior, a large body of work underlined that traditional two-dimensional (2D) cultures depriving cell–cell and cell–ECM interactions as well as spatial cellular distribution and organization of solid tumors fail to recapitulate in vivo properties of tumor cells residing in the complex TME. Thus, three-dimensional (3D) culture models are increasingly employed in cancer research, as these culture systems better mimic the physiological microenvironment and shape the cellular responses according to the microenvironmental cues that will regulate critical cell functions such as cell shape/architecture, survival, proliferation, differentiation, and drug response as well as gene expression. Therefore, 3D cell culture models that better resemble the patient transcriptome are critical in defining physiologically relevant transcriptional changes. This review will present the transcriptional factor (TF) repertoire of breast cancer in 3D culture models in the context of mammary tissue architecture, epithelial-to-mesenchymal transition and metastasis, cell death mechanisms, cancer therapy resistance and differential drug response, and stemness and will discuss the impact of culture dimensionality on breast cancer research.
Collapse
Affiliation(s)
- Hande Özkan
- School of Medicine, Koç University, Istanbul 34450, Turkey;
- Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| | - Deniz Gülfem Öztürk
- School of Medicine, Koç University, Istanbul 34450, Turkey;
- Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
- Correspondence: (D.G.Ö.); (G.K.)
| | - Gozde Korkmaz
- School of Medicine, Koç University, Istanbul 34450, Turkey;
- Research Centre for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
- Correspondence: (D.G.Ö.); (G.K.)
| |
Collapse
|
10
|
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. 3D Cell Culture Systems: Tumor Application, Advantages, and Disadvantages. Int J Mol Sci 2021; 22:12200. [PMID: 34830082 PMCID: PMC8618305 DOI: 10.3390/ijms222212200] [Citation(s) in RCA: 211] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 01/09/2023] Open
Abstract
The traditional two-dimensional (2D) in vitro cell culture system (on a flat support) has long been used in cancer research. However, this system cannot be fully translated into clinical trials to ideally represent physiological conditions. This culture cannot mimic the natural tumor microenvironment due to the lack of cellular communication (cell-cell) and interaction (cell-cell and cell-matrix). To overcome these limitations, three-dimensional (3D) culture systems are increasingly developed in research and have become essential for tumor research, tissue engineering, and basic biology research. 3D culture has received much attention in the field of biomedicine due to its ability to mimic tissue structure and function. The 3D matrix presents a highly dynamic framework where its components are deposited, degraded, or modified to delineate functions and provide a platform where cells attach to perform their specific functions, including adhesion, proliferation, communication, and apoptosis. So far, various types of models belong to this culture: either the culture based on natural or synthetic adherent matrices used to design 3D scaffolds as biomaterials to form a 3D matrix or based on non-adherent and/or matrix-free matrices to form the spheroids. In this review, we first summarize a comparison between 2D and 3D cultures. Then, we focus on the different components of the natural extracellular matrix that can be used as supports in 3D culture. Then we detail different types of natural supports such as matrigel, hydrogels, hard supports, and different synthetic strategies of 3D matrices such as lyophilization, electrospiding, stereolithography, microfluid by citing the advantages and disadvantages of each of them. Finally, we summarize the different methods of generating normal and tumor spheroids, citing their respective advantages and disadvantages in order to obtain an ideal 3D model (matrix) that retains the following characteristics: better biocompatibility, good mechanical properties corresponding to the tumor tissue, degradability, controllable microstructure and chemical components like the tumor tissue, favorable nutrient exchange and easy separation of the cells from the matrix.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Liban;
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| |
Collapse
|
11
|
Özkale B, Sakar MS, Mooney DJ. Active biomaterials for mechanobiology. Biomaterials 2021; 267:120497. [PMID: 33129187 PMCID: PMC7719094 DOI: 10.1016/j.biomaterials.2020.120497] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 02/06/2023]
Abstract
Active biomaterials offer novel approaches to study mechanotransduction in mammalian cells. These material systems probe cellular responses by dynamically modulating their resistance to endogenous forces or applying exogenous forces on cells in a temporally controlled manner. Stimuli-responsive molecules, polymers, and nanoparticles embedded inside cytocompatible biopolymer networks transduce external signals such as light, heat, chemicals, and magnetic fields into changes in matrix elasticity (few kPa to tens of kPa) or forces (few pN to several μN) at the cell-material interface. The implementation of active biomaterials in mechanobiology has generated scientific knowledge and therapeutic potential relevant to a variety of conditions including but not limited to cancer metastasis, fibrosis, and tissue regeneration. We discuss the repertoire of cellular responses that can be studied using these platforms including receptor signaling as well as downstream events namely, cytoskeletal organization, nuclear shuttling of mechanosensitive transcriptional regulators, cell migration, and differentiation. We highlight recent advances in active biomaterials and comment on their future impact.
Collapse
Affiliation(s)
- Berna Özkale
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA, 02138, USA
| | - Mahmut Selman Sakar
- Institute of Mechanical Engineering and Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland.
| | - David J Mooney
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA, 02138, USA.
| |
Collapse
|
12
|
Zhang W, Huang G, Xu F. Engineering Biomaterials and Approaches for Mechanical Stretching of Cells in Three Dimensions. Front Bioeng Biotechnol 2020; 8:589590. [PMID: 33154967 PMCID: PMC7591716 DOI: 10.3389/fbioe.2020.589590] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Mechanical stretch is widely experienced by cells of different tissues in the human body and plays critical roles in regulating their behaviors. Numerous studies have been devoted to investigating the responses of cells to mechanical stretch, providing us with fruitful findings. However, these findings have been mostly observed from two-dimensional studies and increasing evidence suggests that cells in three dimensions may behave more closely to their in vivo behaviors. While significant efforts and progresses have been made in the engineering of biomaterials and approaches for mechanical stretching of cells in three dimensions, much work remains to be done. Here, we briefly review the state-of-the-art researches in this area, with focus on discussing biomaterial considerations and stretching approaches. We envision that with the development of advanced biomaterials, actuators and microengineering technologies, more versatile and predictive three-dimensional cell stretching models would be available soon for extensive applications in such fields as mechanobiology, tissue engineering, and drug screening.
Collapse
Affiliation(s)
- Weiwei Zhang
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Guoyou Huang
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing University, Chongqing, China
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center, Xi’an Jiaotong University, Xi’an, China
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Sciences and Technology, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
13
|
Ribeiro-Filho AC, Levy D, Ruiz JLM, Mantovani MDC, Bydlowski SP. Traditional and Advanced Cell Cultures in Hematopoietic Stem Cell Studies. Cells 2019; 8:cells8121628. [PMID: 31842488 PMCID: PMC6953118 DOI: 10.3390/cells8121628] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 01/09/2023] Open
Abstract
Hematopoiesis is the main function of bone marrow. Human hematopoietic stem and progenitor cells reside in the bone marrow microenvironment, making it a hotspot for the development of hematopoietic diseases. Numerous alterations that correspond to disease progression have been identified in the bone marrow stem cell niche. Complex interactions between the bone marrow microenvironment and hematopoietic stem cells determine the balance between the proliferation, differentiation and homeostasis of the stem cell compartment. Changes in this tightly regulated network can provoke malignant transformation. However, our understanding of human hematopoiesis and the associated niche biology remains limited due to accessibility to human material and the limits of in vitro culture models. Traditional culture systems for human hematopoietic studies lack microenvironment niches, spatial marrow gradients, and dense cellularity, rendering them incapable of effectively translating marrow physiology ex vivo. This review will discuss the importance of 2D and 3D culture as a physiologically relevant system for understanding normal and abnormal hematopoiesis.
Collapse
Affiliation(s)
- Antonio Carlos Ribeiro-Filho
- Organoid Development Team, Center of Innovation and Translational Medicine (CIMTRA), University of São Paulo School of Medicine, Sao Paulo 05360-130, Brazil; (A.C.R.-F.); (M.d.C.M.)
| | - Débora Levy
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), University of São Paulo School of Medicine, Sao Paulo 05403-900, Brazil;
| | - Jorge Luis Maria Ruiz
- Life and Nature Science Institute, Federal University of Latin American Integration-UNILA, Foz de Iguaçú, PR 858570-901, Brazil;
| | - Marluce da Cunha Mantovani
- Organoid Development Team, Center of Innovation and Translational Medicine (CIMTRA), University of São Paulo School of Medicine, Sao Paulo 05360-130, Brazil; (A.C.R.-F.); (M.d.C.M.)
| | - Sérgio Paulo Bydlowski
- Organoid Development Team, Center of Innovation and Translational Medicine (CIMTRA), University of São Paulo School of Medicine, Sao Paulo 05360-130, Brazil; (A.C.R.-F.); (M.d.C.M.)
- Lipids, Oxidation and Cell Biology Team, Laboratory of Immunology (LIM19), Heart Institute (InCor), University of São Paulo School of Medicine, Sao Paulo 05403-900, Brazil;
- National Institute of Science and Technology in Regenerative Medicine (INCT-Regenera), CNPq, Rio de Janeiro 21941-902, Brazil
- Correspondence:
| |
Collapse
|
14
|
Straining 3D Hydrogels with Uniform Z-Axis Strains While Enabling Live Microscopy Imaging. Ann Biomed Eng 2019; 48:868-880. [PMID: 31802281 DOI: 10.1007/s10439-019-02426-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 11/24/2019] [Indexed: 10/25/2022]
Abstract
External forces play an important role in the development and regulation of many tissues. Such effects are often studied using specialized stretchers-standardized commercial and novel laboratory-designed. While designs for 2D stretchers are abundant, the range of available 3D stretcher designs is more limited, especially when live imaging is required. This work presents a novel method and a stretching device that allow straining of 3D hydrogels from their circumference, using a punctured elastic silicone strip as the sample carrier. The system was primarily constructed from 3D-printed parts and low-cost electronics, rendering it simple and cost-efficient to reproduce in other labs. To demonstrate the system functionality, > 100 μm thick soft fibrin gels (< 1 KPa) were stretched, while performing live confocal imaging. The subsequent strains and fiber alignment were analyzed and found to be relatively homogenous throughout the gel's thickness (Z axis). The uniform Z-response enabled by our approach was found to be in contrast to a previously reported approach that utilizes an underlying elastic substrate to convey strain to a 3D thick sample. This work advances the ability to study the role of external forces on biological processes under more physiological 3D conditions, and can contribute to the field of tissue engineering.
Collapse
|
15
|
Jepsen ML, Nielsen LH, Boisen A, Almdal K, Dufva M. Characterization of thin gelatin hydrogel membranes with balloon properties for dynamic tissue engineering. Biopolymers 2018; 110:e23241. [PMID: 30536858 DOI: 10.1002/bip.23241] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/12/2018] [Accepted: 11/02/2018] [Indexed: 02/04/2023]
Abstract
Cell or tissue stretching and strain are present in any in vivo environment, but is difficult to reproduce in vitro. Here, we describe a simple method for casting a thin (about 500 μm) and soft (about 0.3 kPa) hydrogel of gelatin and a method for characterizing the mechanical properties of the hydrogel simply by changing pressure with a water column. The gelatin is crosslinked with mTransglutaminase and the area of the resulting hydrogel can be increased up 13-fold by increasing the radial water pressure. This is far beyond physiological stretches observed in vivo. Actuating the hydrogel with a radial force achieves both information about stiffness, stretchability, and contractability, which are relevant properties for tissue engineering purposes. Cells could be stretched and contracted using the gelatin membrane. Gelatin is a commonly used polymer for hydrogels in tissue engineering, and the discovered reversible stretching is particularly interesting for organ modeling applications.
Collapse
Affiliation(s)
- Morten Leth Jepsen
- Department of Micro- and Nanotechnology, The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Technical University of Denmark, Kongens Lyngby, Denmark
| | - Line Hagner Nielsen
- Department of Micro- and Nanotechnology, The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anja Boisen
- Department of Micro- and Nanotechnology, The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Technical University of Denmark, Kongens Lyngby, Denmark
| | - Kristoffer Almdal
- Department of Micro- and Nanotechnology, The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Technical University of Denmark, Kongens Lyngby, Denmark
| | - Martin Dufva
- Department of Micro- and Nanotechnology, The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
16
|
Ergir E, Bachmann B, Redl H, Forte G, Ertl P. Small Force, Big Impact: Next Generation Organ-on-a-Chip Systems Incorporating Biomechanical Cues. Front Physiol 2018; 9:1417. [PMID: 30356887 PMCID: PMC6190857 DOI: 10.3389/fphys.2018.01417] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 09/18/2018] [Indexed: 12/13/2022] Open
Abstract
Mechanobiology-on-a-chip is a growing field focusing on how mechanical inputs modulate physico-chemical output in microphysiological systems. It is well known that biomechanical cues trigger a variety of molecular events and adjustment of mechanical forces is therefore essential for mimicking in vivo physiologies in organ-on-a-chip technology. Biomechanical inputs in organ-on-a-chip systems can range from variations in extracellular matrix type and stiffness and applied shear stresses to active stretch/strain or compression forces using integrated flexible membranes. The main advantages of these organ-on-a-chip systems are therefore (a) the control over spatiotemporal organization of in vivo-like tissue architectures, (b) the ability to precisely control the amount, duration and intensity of the biomechanical stimuli, and (c) the capability of monitoring in real time the effects of applied mechanical forces on cell, tissue and organ functions. Consequently, over the last decade a variety of microfluidic devices have been introduced to recreate physiological microenvironments that also account for the influence of physical forces on biological functions. In this review we present recent advances in mechanobiological lab-on-a-chip systems and report on lessons learned from these current mechanobiological models. Additionally, future developments needed to engineer next-generation physiological and pathological organ-on-a-chip models are discussed.
Collapse
Affiliation(s)
- Ece Ergir
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria
| | - Barbara Bachmann
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Kompetenzzentrum für MechanoBiologie (INTERREG V-A Austria – Czech Republic Programme, ATCZ133), Vienna, Austria
| | - Heinz Redl
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Kompetenzzentrum für MechanoBiologie (INTERREG V-A Austria – Czech Republic Programme, ATCZ133), Vienna, Austria
| | - Giancarlo Forte
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- Competence Center for Mechanobiology (INTERREG V-A Austria – Czech Republic Programme, ATCZ133), Brno, Czechia
- Department of Biomaterials Science, Institute of Dentistry, University of Turku, Turku, Finland
| | - Peter Ertl
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Kompetenzzentrum für MechanoBiologie (INTERREG V-A Austria – Czech Republic Programme, ATCZ133), Vienna, Austria
| |
Collapse
|
17
|
Li J, Li Y, Gao B, Qin C, He Y, Xu F, Yang H, Lin M. Engineering mechanical microenvironment of macrophage and its biomedical applications. Nanomedicine (Lond) 2018; 13:555-576. [PMID: 29334336 DOI: 10.2217/nnm-2017-0324] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Macrophages are the most plastic cells in the hematopoietic system and can be widely found in almost all tissues. Recently studies have shown that mechanical cues (e.g., matrix stiffness and stress/strain) can significantly affect macrophage behaviors. Although existing reviews on the physical and mechanical cues that regulate the macrophage's phenotype are available, engineering mechanical microenvironment of macrophages in vitro as well as a comprehensive overview and prospects for their biomedical applications (e.g., tissue engineering and immunotherapy) has yet to be summarized. Thus, this review provides an overview on the existing methods for engineering mechanical microenvironment of macrophages in vitro and then a section on their biomedical applications and further perspectives are presented.
Collapse
Affiliation(s)
- Jing Li
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China.,Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China.,Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,Key Laboratory on Space Physics and Chemistry of Ministry of Education and Key Laboratory on Macromolecular Science & Technology of Shanxi Province, Department of Applied Chemistry, School of Science, Northwestern Polytechnical University, 710072, P.R China
| | - Yuhui Li
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,The Key Library of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Bin Gao
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,The Key Library of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China.,Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, P.R. China
| | - Chuanguang Qin
- Key Laboratory on Space Physics and Chemistry of Ministry of Education and Key Laboratory on Macromolecular Science & Technology of Shanxi Province, Department of Applied Chemistry, School of Science, Northwestern Polytechnical University, 710072, P.R China
| | - Yining He
- College of Food Science and Engineering, Northwest A & F University Yangling Shaanxi 712100 China
| | - Feng Xu
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,The Key Library of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China.,Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China
| | - Min Lin
- Bioinspired Engineering & Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.,The Key Library of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P.R. China
| |
Collapse
|
18
|
Zhang W, Huang G, Ng K, Ji Y, Gao B, Huang L, Zhou J, Lu TJ, Xu F. Engineering ellipsoidal cap-like hydrogel particles as building blocks or sacrificial templates for three-dimensional cell culture. Biomater Sci 2018; 6:885-892. [DOI: 10.1039/c7bm01186e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
AAO template-assisted fabrication of ellipsoidal cap-like hydrogel particles as building blocks or sacrificial templates for 3D cell culture.
Collapse
Affiliation(s)
- Weiwei Zhang
- Non-equilibrium Condensed Matter and Quantum Engineering Laboratory
- The Key Laboratory of Ministry of Education
- School of Science
- Xi'an Jiaotong University
- Xi'an 710049
| | - Guoyou Huang
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
| | - Kelvin Ng
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
- Department of Biomedical Engineering
| | - Yuan Ji
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
| | - Bin Gao
- Department of Endocrinology and Metabolism
- Xijing Hospital
- Fourth Military Medical University
- Xi'an 710054
- P.R. China
| | - Liqing Huang
- Non-equilibrium Condensed Matter and Quantum Engineering Laboratory
- The Key Laboratory of Ministry of Education
- School of Science
- Xi'an Jiaotong University
- Xi'an 710049
| | - Jinxiong Zhou
- State Key Laboratory for Strength and Vibration of Mechanical Structures
- School of Aerospace
- Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
- MOE Key Laboratory for Multifunctional Materials and Structures
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC)
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University
- Xi'an 710049
- P. R. China
| |
Collapse
|
19
|
Cheng B, Lin M, Huang G, Li Y, Ji B, Genin GM, Deshpande VS, Lu TJ, Xu F. Cellular mechanosensing of the biophysical microenvironment: A review of mathematical models of biophysical regulation of cell responses. Phys Life Rev 2017; 22-23:88-119. [PMID: 28688729 PMCID: PMC5712490 DOI: 10.1016/j.plrev.2017.06.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 06/14/2017] [Indexed: 12/11/2022]
Abstract
Cells in vivo reside within complex microenvironments composed of both biochemical and biophysical cues. The dynamic feedback between cells and their microenvironments hinges upon biophysical cues that regulate critical cellular behaviors. Understanding this regulation from sensing to reaction to feedback is therefore critical, and a large effort is afoot to identify and mathematically model the fundamental mechanobiological mechanisms underlying this regulation. This review provides a critical perspective on recent progress in mathematical models for the responses of cells to the biophysical cues in their microenvironments, including dynamic strain, osmotic shock, fluid shear stress, mechanical force, matrix rigidity, porosity, and matrix shape. The review highlights key successes and failings of existing models, and discusses future opportunities and challenges in the field.
Collapse
Affiliation(s)
- Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Guoyou Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuhui Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Baohua Ji
- Biomechanics and Biomaterials Laboratory, Department of Applied Mechanics, Beijing Institute of Technology, Beijing, China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; Department of Mechanical Engineering & Materials Science, and NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis 63130, MO, USA
| | - Vikram S Deshpande
- Department of Engineering, University of Cambridge, Cambridge CB2 1PZ, United Kingdom
| | - Tian Jian Lu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
20
|
Huang G, Li F, Zhao X, Ma Y, Li Y, Lin M, Jin G, Lu TJ, Genin GM, Xu F. Functional and Biomimetic Materials for Engineering of the Three-Dimensional Cell Microenvironment. Chem Rev 2017; 117:12764-12850. [PMID: 28991456 PMCID: PMC6494624 DOI: 10.1021/acs.chemrev.7b00094] [Citation(s) in RCA: 486] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cell microenvironment has emerged as a key determinant of cell behavior and function in development, physiology, and pathophysiology. The extracellular matrix (ECM) within the cell microenvironment serves not only as a structural foundation for cells but also as a source of three-dimensional (3D) biochemical and biophysical cues that trigger and regulate cell behaviors. Increasing evidence suggests that the 3D character of the microenvironment is required for development of many critical cell responses observed in vivo, fueling a surge in the development of functional and biomimetic materials for engineering the 3D cell microenvironment. Progress in the design of such materials has improved control of cell behaviors in 3D and advanced the fields of tissue regeneration, in vitro tissue models, large-scale cell differentiation, immunotherapy, and gene therapy. However, the field is still in its infancy, and discoveries about the nature of cell-microenvironment interactions continue to overturn much early progress in the field. Key challenges continue to be dissecting the roles of chemistry, structure, mechanics, and electrophysiology in the cell microenvironment, and understanding and harnessing the roles of periodicity and drift in these factors. This review encapsulates where recent advances appear to leave the ever-shifting state of the art, and it highlights areas in which substantial potential and uncertainty remain.
Collapse
Affiliation(s)
- Guoyou Huang
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Chemistry, School of Science,
Xi’an Jiaotong University, Xi’an 710049, People’s Republic
of China
| | - Xin Zhao
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Interdisciplinary Division of Biomedical
Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong,
People’s Republic of China
| | - Yufei Ma
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Yuhui Li
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Min Lin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Guorui Jin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- MOE Key Laboratory for Multifunctional Materials
and Structures, Xi’an Jiaotong University, Xi’an 710049,
People’s Republic of China
| | - Guy M. Genin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Mechanical Engineering &
Materials Science, Washington University in St. Louis, St. Louis 63130, MO,
USA
- NSF Science and Technology Center for
Engineering MechanoBiology, Washington University in St. Louis, St. Louis 63130,
MO, USA
| | - Feng Xu
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| |
Collapse
|
21
|
Miao C, Schiffhauer ES, Okeke EI, Robinson DN, Luo T. Parallel Compression Is a Fast Low-Cost Assay for the High-Throughput Screening of Mechanosensory Cytoskeletal Proteins in Cells. ACS APPLIED MATERIALS & INTERFACES 2017; 9:28168-28179. [PMID: 28795554 PMCID: PMC5891216 DOI: 10.1021/acsami.7b04622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Cellular mechanosensing is critical for many biological processes, including cell differentiation, proliferation, migration, and tissue morphogenesis. The actin cytoskeletal proteins play important roles in cellular mechanosensing. Many techniques have been used to investigate the mechanosensory behaviors of these proteins. However, a fast, low-cost assay for the quantitative characterization of these proteins is still lacking. Here, we demonstrate that compression assay using agarose overlay is suitable for the high throughput screening of mechanosensory proteins in live cells while requiring minimal experimental setup. We used several well-studied myosin II mutants to assess the compression assay. On the basis of elasticity theories, we simulated the mechanosensory accumulation of myosin II's and quantitatively reproduced the experimentally observed protein dynamics. Combining the compression assay with confocal microscopy, we monitored the polarization of myosin II oligomers at the subcellular level. The polarization was dependent on the ratio of the two principal strains of the cellular deformations. Finally, we demonstrated that this technique could be used on the investigation of other mechanosensory proteins.
Collapse
Affiliation(s)
- Chunguang Miao
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230000, China
| | - Eric S. Schiffhauer
- Departments of Cell Biology, Pharmacology and Molecular Medicine, and Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Evelyn I. Okeke
- Departments of Cell Biology, Pharmacology and Molecular Medicine, and Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Douglas N. Robinson
- Departments of Cell Biology, Pharmacology and Molecular Medicine, and Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21211, United States
| | - Tianzhi Luo
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei 230000, China
- Departments of Cell Biology, Pharmacology and Molecular Medicine, and Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
- Corresponding Author:
| |
Collapse
|
22
|
Wu RX, Yin Y, He XT, Li X, Chen FM. Engineering a Cell Home for Stem Cell Homing and Accommodation. ACTA ACUST UNITED AC 2017; 1:e1700004. [PMID: 32646164 DOI: 10.1002/adbi.201700004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/27/2017] [Indexed: 12/14/2022]
Abstract
Distilling complexity to advance regenerative medicine from laboratory animals to humans, in situ regeneration will continue to evolve using biomaterial strategies to drive endogenous cells within the human body for therapeutic purposes; this approach avoids the need for delivering ex vivo-expanded cellular materials. Ensuring the recruitment of a significant number of reparative cells from an endogenous source to the site of interest is the first step toward achieving success. Subsequently, making the "cell home" cell-friendly by recapitulating the natural extracellular matrix (ECM) in terms of its chemistry, structure, dynamics, and function, and targeting specific aspects of the native stem cell niche (e.g., cell-ECM and cell-cell interactions) to program and steer the fates of those recruited stem cells play equally crucial roles in yielding a therapeutically regenerative solution. This review addresses the key aspects of material-guided cell homing and the engineering of novel biomaterials with desirable ECM composition, surface topography, biochemistry, and mechanical properties that can present both biochemical and physical cues required for in situ tissue regeneration. This growing body of knowledge will likely become a design basis for the development of regenerative biomaterials for, but not limited to, future in situ tissue engineering and regeneration.
Collapse
Affiliation(s)
- Rui-Xin Wu
- State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P. R. China.,National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P. R. China.,National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Xiao-Tao He
- State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P. R. China.,National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Xuan Li
- State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P. R. China.,National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P. R. China.,National Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, P.R. China
| |
Collapse
|
23
|
Muhamed I, Chowdhury F, Maruthamuthu V. Biophysical Tools to Study Cellular Mechanotransduction. Bioengineering (Basel) 2017; 4:E12. [PMID: 28952491 PMCID: PMC5590431 DOI: 10.3390/bioengineering4010012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/30/2017] [Accepted: 02/02/2017] [Indexed: 01/25/2023] Open
Abstract
The cell membrane is the interface that volumetrically isolates cellular components from the cell's environment. Proteins embedded within and on the membrane have varied biological functions: reception of external biochemical signals, as membrane channels, amplification and regulation of chemical signals through secondary messenger molecules, controlled exocytosis, endocytosis, phagocytosis, organized recruitment and sequestration of cytosolic complex proteins, cell division processes, organization of the cytoskeleton and more. The membrane's bioelectrical role is enabled by the physiologically controlled release and accumulation of electrochemical potential modulating molecules across the membrane through specialized ion channels (e.g., Na⁺, Ca2+, K⁺ channels). The membrane's biomechanical functions include sensing external forces and/or the rigidity of the external environment through force transmission, specific conformational changes and/or signaling through mechanoreceptors (e.g., platelet endothelial cell adhesion molecule (PECAM), vascular endothelial (VE)-cadherin, epithelial (E)-cadherin, integrin) embedded in the membrane. Certain mechanical stimulations through specific receptor complexes induce electrical and/or chemical impulses in cells and propagate across cells and tissues. These biomechanical sensory and biochemical responses have profound implications in normal physiology and disease. Here, we discuss the tools that facilitate the understanding of mechanosensitive adhesion receptors. This article is structured to provide a broad biochemical and mechanobiology background to introduce a freshman mechano-biologist to the field of mechanotransduction, with deeper study enabled by many of the references cited herein.
Collapse
Affiliation(s)
- Ismaeel Muhamed
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA.
| | - Farhan Chowdhury
- Department of Mechanical Engineering and Energy Processes, Southern Illinois University Carbondale, Carbondale, IL 62901, USA.
| | - Venkat Maruthamuthu
- Department of Mechanical and Aerospace Engineering, Old Dominion University, Norfolk, VA 23529, USA.
| |
Collapse
|
24
|
Chen X, Bai S, Li B, Liu H, Wu G, Liu S, Zhao Y. Fabrication of gelatin methacrylate/nanohydroxyapatite microgel arrays for periodontal tissue regeneration. Int J Nanomedicine 2016; 11:4707-4718. [PMID: 27695327 PMCID: PMC5028089 DOI: 10.2147/ijn.s111701] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Periodontitis is a chronic infectious disease and is the major cause of tooth loss and other oral health issues around the world. Periodontal tissue regeneration has therefore always been the ultimate goal of dentists and researchers. Existing fabrication methods mainly focused on a top-down tissue engineering strategy in which several drawbacks remain, including low throughput and limited diffusion properties resulting from a large sample size. Gelatin methacrylate (GelMA) is a kind of photocrosslinkable and biocompatible hydrogel, with the capacities of enabling cell encapsulation and regeneration of functional tissues. Here, we developed a novel method to fabricate GelMA/nanohydroxylapatite (nHA) microgel arrays using a photocrosslinkable strategy. The viability, proliferation, and osteogenic differentiation and in vivo osteogenesis of human periodontal ligament stem cells (hPDLSCs) encapsulated in microgels were evaluated. The results suggested that such microgels provide great potential for periodontal tissue repair and regeneration. METHODS Microgel arrays were fabricated by blending different weight ratios of GelMA and nHA. hPDLSCs were encapsulated in GelMA/nHA microgels of various ratios for a systematic evaluation of cell viability, proliferation, and osteogenic differentiation. In vivo osteogenesis in nude mice was also studied. RESULTS The GelMA/nHA microgels exhibited appropriate microarchitecture, mechanical strength, and surface roughness, thus enabling cell adhesion and proliferation. Additionally, the GelMA/nHA microgels (10%/2% w/v) enhanced the osteogenic differentiation of hPDLSCs by elevating the expression levels of osteogenic biomarker genes, such as ALP, BSP, OCN, and RUNX2. In vivo ectopic transplantation results showed that GelMA/nHA microgels (10%/2% w/v) increased mineralized tissue formation with abundant vascularization, compared with the 1%, 3%, and the pure GelMA group. CONCLUSION The GelMA/nHA microgels (10%/2% w/v) facilitated hPDLSCs viability, proliferation, and osteogenic differentiation in vitro and further promoted new bone formation in vivo, suggesting that the GelMA/nHA microgels (10%/2% w/v) provide great potential for periodontal tissue regeneration.
Collapse
Affiliation(s)
- Xi Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics
| | - Shizhu Bai
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics
| | - Bei Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Oral Diseases, Center for Tissue Engineering, School of Stomatology
| | - Huan Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics
| | - Guofeng Wu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics
| | - Sha Liu
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, The Fourth Military Medical University, Shaanxi, People’s Republic of China
| | - Yimin Zhao
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Oral Diseases, Department of Prosthodontics
| |
Collapse
|