1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Mechano-responsive hydrogel for direct stem cell manufacturing to therapy

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Bone marrow-derived mesenchymal stem cell (MSC) is one of the most actively studied cell types due to its regenerative potential and immunomodulatory properties. Conventional cell expansion methods using 2D tissue culture plates and 2.5D microcarriers in bioreactors can generate large cell numbers, but they compromise stem cell potency and lack mechanical preconditioning to prepare MSC for physiological loading expected in vivo. To overcome these challenges, in this work, we describe a 3D dynamic hydrogel using magneto-stimulation for direct MSC manufacturing to therapy. With our technology, we found that dynamic mechanical stimulation (DMS) enhanced matrix-integrin β1 interactions which induced MSCs spreading and proliferation. In addition, DMS could modulate MSC biofunctions including directing MSC differentiation into specific lineages and boosting paracrine activities (e.g., growth factor secretion) through YAP nuclear localization and FAK-ERK pathway. With our magnetic hydrogel, complex procedures from MSC manufacturing to final clinical use, can be integrated into one single platform, and we believe this ‘all-in-one’ technology could offer a paradigm shift to existing standards in MSC therapy.

          Graphical abstract

          Highlights

          • Dynamic mechanical stimulation (DMS) induced by wireless magnetic field enhances matrix-integrin β1 interactions.

          • 3D hydrogel platform optimizes mesenchymal stem cell (MSC) quantity and quality during manufacturing.

          • Mechanical pre-conditioning enhances MSC biofunctions including cell differentiation, expansion, and secretome.

          • ‘All-in-one’ strategy from MSC manufacturing to direct therapy/clinical use.

          Related collections

          Most cited references97

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Mesenchymal stem cell perspective: cell biology to clinical progress

          The terms MSC and MSCs have become the preferred acronym to describe a cell and a cell population of multipotential stem/progenitor cells commonly referred to as mesenchymal stem cells, multipotential stromal cells, mesenchymal stromal cells, and mesenchymal progenitor cells. The MSCs can differentiate to important lineages under defined conditions in vitro and in limited situations after implantation in vivo. MSCs were isolated and described about 30 years ago and now there are over 55,000 publications on MSCs readily available. Here, we have focused on human MSCs whenever possible. The MSCs have broad anti-inflammatory and immune-modulatory properties. At present, these provide the greatest focus of human MSCs in clinical testing; however, the properties of cultured MSCs in vitro suggest they can have broader applications. The medical utility of MSCs continues to be investigated in over 950 clinical trials. There has been much progress in understanding MSCs over the years, and there is a strong foundation for future scientific research and clinical applications, but also some important questions remain to be answered. Developing further methods to understand and unlock MSC potential through intracellular and intercellular signaling, biomedical engineering, delivery methods and patient selection should all provide substantial advancements in the coming years and greater clinical opportunities. The expansive and growing field of MSC research is teaching us basic human cell biology as well as how to use this type of cell for cellular therapy in a variety of clinical settings, and while much promise is evident, careful new work is still needed.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Mechanical forces direct stem cell behaviour in development and regeneration

            Stem cells and their local microenvironment, or niche, communicate through mechanical cues to regulate cell fate and cell behaviour and to guide developmental processes. During embryonic development, mechanical forces are involved in patterning and organogenesis. The physical environment of pluripotent stem cells regulates their self-renewal and differentiation. Mechanical and physical cues are also important in adult tissues, where adult stem cells require physical interactions with the extracellular matrix to maintain their potency. In vitro, synthetic models of the stem cell niche can be used to precisely control and manipulate the biophysical and biochemical properties of the stem cell microenvironment and to examine how the mode and magnitude of mechanical cues, such as matrix stiffness or applied forces, direct stem cell differentiation and function. Fundamental insights into the mechanobiology of stem cells also inform the design of artificial niches to support stem cells for regenerative therapies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Geometric cues for directing the differentiation of mesenchymal stem cells.

              Significant efforts have been directed to understanding the factors that influence the lineage commitment of stem cells. This paper demonstrates that cell shape, independent of soluble factors, has a strong influence on the differentiation of human mesenchymal stem cells (MSCs) from bone marrow. When exposed to competing soluble differentiation signals, cells cultured in rectangles with increasing aspect ratio and in shapes with pentagonal symmetry but with different subcellular curvature-and with each occupying the same area-display different adipogenesis and osteogenesis profiles. The results reveal that geometric features that increase actomyosin contractility promote osteogenesis and are consistent with in vivo characteristics of the microenvironment of the differentiated cells. Cytoskeletal-disrupting pharmacological agents modulate shape-based trends in lineage commitment verifying the critical role of focal adhesion and myosin-generated contractility during differentiation. Microarray analysis and pathway inhibition studies suggest that contractile cells promote osteogenesis by enhancing c-Jun N-terminal kinase (JNK) and extracellular related kinase (ERK1/2) activation in conjunction with elevated wingless-type (Wnt) signaling. Taken together, this work points to the role that geometric shape cues can play in orchestrating the mechanochemical signals and paracrine/autocrine factors that can direct MSCs to appropriate fates.
                Bookmark

                Author and article information

                Contributors
                Journal
                Bioact Mater
                Bioact Mater
                Bioactive Materials
                KeAi Publishing
                2452-199X
                02 January 2023
                June 2023
                02 January 2023
                : 24
                : 387-400
                Affiliations
                [a ]Department of Biomedical Engineering, National University of Singapore, 117583, Singapore
                [b ]Institute for Health Innovation & Technology, National University of Singapore, 117599, Singapore
                [c ]School of Civil Engineering and Architecture, Wuhan University of Technology, 430070, Wuhan, China
                [d ]NUS Tissue Engineering Program, National University of Singapore, 117510, Singapore
                [e ]Lee Kong Chian School of Medicine, Nanyang Technological University, 636921, Singapore
                [f ]School of Mechanical and Aerospace Engineering, Nanyang Technological University, 639798, Singapore
                [g ]Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 138648, Singapore
                [h ]Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288, Singapore
                [i ]Mechanobiology Institute, National University of Singapore, 117411, Singapore
                Author notes
                []Corresponding author. Department of Biomedical Engineering, National University of Singapore, 117583, Singapore. bietkpa@ 123456nus.edu.sg
                [1]

                Co-first author.

                Article
                S2452-199X(22)00507-2
                10.1016/j.bioactmat.2022.12.019
                9817177
                36632503
                3e6b73b7-3d6e-419b-933e-6efe619cbc6d
                © 2023 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 9 October 2022
                : 5 December 2022
                : 20 December 2022
                Categories
                Article

                mesenchymal stem cell,dynamic mechanical stimulation,magnetic hydrogel,stem cell manufacturing,cell therapy,msc, mesenchymal stem cell,dms, dynamic mechanical stimulation,mp, magnetic particle

                Comments

                Comment on this article