1
|
Casarella S, Ferla F, Di Francesco D, Canciani E, Rizzi M, Boccafoschi F. Focal Adhesion's Role in Cardiomyocytes Function: From Cardiomyogenesis to Mechanotransduction. Cells 2024; 13:664. [PMID: 38667279 PMCID: PMC11049660 DOI: 10.3390/cells13080664] [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: 02/23/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Mechanotransduction refers to the ability of cells to sense mechanical stimuli and convert them into biochemical signals. In this context, the key players are focal adhesions (FAs): multiprotein complexes that link intracellular actin bundles and the extracellular matrix (ECM). FAs are involved in cellular adhesion, growth, differentiation, gene expression, migration, communication, force transmission, and contractility. Focal adhesion signaling molecules, including Focal Adhesion Kinase (FAK), integrins, vinculin, and paxillin, also play pivotal roles in cardiomyogenesis, impacting cell proliferation and heart tube looping. In fact, cardiomyocytes sense ECM stiffness through integrins, modulating signaling pathways like PI3K/AKT and Wnt/β-catenin. Moreover, FAK/Src complex activation mediates cardiac hypertrophic growth and survival signaling in response to mechanical loads. This review provides an overview of the molecular and mechanical mechanisms underlying the crosstalk between FAs and cardiac differentiation, as well as the role of FA-mediated mechanotransduction in guiding cardiac muscle responses to mechanical stimuli.
Collapse
Affiliation(s)
- Simona Casarella
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Federica Ferla
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Dalila Di Francesco
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering, University Hospital Research Center, Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Elena Canciani
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Manuela Rizzi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Francesca Boccafoschi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| |
Collapse
|
2
|
Mezher M, Dumbali S, Fenn I, Lamb C, Miller C, Sharmin S, Cabe JI, Bejar-Padilla V, Conway D, Maruthamuthu V. Vinculin is essential for sustaining normal levels of endogenous forces at cell-cell contacts. Biophys J 2023; 122:4518-4527. [PMID: 38350000 PMCID: PMC10719050 DOI: 10.1016/j.bpj.2023.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/11/2023] [Accepted: 10/25/2023] [Indexed: 02/15/2024] Open
Abstract
Transmission of cell-generated (i.e., endogenous) tension at cell-cell contacts is crucial for tissue shape changes during morphogenesis and adult tissue repair in tissues such as epithelia. E-cadherin-based adhesions at cell-cell contacts are the primary means by which endogenous tension is transmitted between cells. The E-cadherin-β-catenin-α-catenin complex mechanically couples to the actin cytoskeleton (and thereby the cell's contractile machinery) both directly and indirectly. However, the key adhesion constituents required for substantial endogenous force transmission at these adhesions in cell-cell contacts are unclear. Due to the role of α-catenin as a mechanotransducer that recruits vinculin at cell-cell contacts, we expected α-catenin to be essential for sustaining normal levels of force transmission. Instead, using the traction force imbalance method to determine the inter-cellular force at a single cell-cell contact between cell pairs, we found that it is vinculin that is essential for sustaining normal levels of endogenous force transmission, with absence of vinculin decreasing the inter-cellular tension by over 50%. Our results constrain the potential mechanical pathways of force transmission at cell-cell contacts and suggest that vinculin can transmit forces at E-cadherin adhesions independent of α-catenin, possibly through β-catenin. Furthermore, we tested the ability of lateral cell-cell contacts to withstand external stretch and found that both vinculin and α-catenin are essential to maintain cell-cell contact stability under external forces.
Collapse
Affiliation(s)
- Mazen Mezher
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Sandeep Dumbali
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Ian Fenn
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Carter Lamb
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Conrad Miller
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Saika Sharmin
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia
| | - Jolene I Cabe
- Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Vidal Bejar-Padilla
- Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Daniel Conway
- Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Venkat Maruthamuthu
- Mechanical & Aerospace Engineering, Old Dominion University, Norfolk, Virginia.
| |
Collapse
|
3
|
Bakhshandeh B, Sorboni SG, Ranjbar N, Deyhimfar R, Abtahi MS, Izady M, Kazemi N, Noori A, Pennisi CP. Mechanotransduction in tissue engineering: Insights into the interaction of stem cells with biomechanical cues. Exp Cell Res 2023; 431:113766. [PMID: 37678504 DOI: 10.1016/j.yexcr.2023.113766] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Stem cells in their natural microenvironment are exposed to biochemical and biophysical cues emerging from the extracellular matrix (ECM) and neighboring cells. In particular, biomechanical forces modulate stem cell behavior, biological fate, and early developmental processes by sensing, interpreting, and responding through a series of biological processes known as mechanotransduction. Local structural changes in the ECM and mechanics are driven by reciprocal activation of the cell and the ECM itself, as the initial deposition of matrix proteins sequentially affects neighboring cells. Recent studies on stem cell mechanoregulation have provided insight into the importance of biomechanical signals on proper tissue regeneration and function and have shown that precise spatiotemporal control of these signals exists in stem cell niches. Against this background, the aim of this work is to review the current understanding of the molecular basis of mechanotransduction by analyzing how biomechanical forces are converted into biological responses via cellular signaling pathways. In addition, this work provides an overview of advanced strategies using stem cells and biomaterial scaffolds that enable precise spatial and temporal control of mechanical signals and offer great potential for the fields of tissue engineering and regenerative medicine will be presented.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran.
| | | | - Nika Ranjbar
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Roham Deyhimfar
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Maryam Sadat Abtahi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mehrnaz Izady
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Navid Kazemi
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Atefeh Noori
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and Technology, Aalborg University, Denmark.
| |
Collapse
|
4
|
Goldshmid R, Simaan-Yameen H, Ifergan L, Loebel C, Burdick JA, Seliktar D. Modulus-dependent effects on neurogenic, myogenic, and chondrogenic differentiation of human mesenchymal stem cells in three-dimensional hydrogel cultures. J Biomed Mater Res A 2023; 111:1441-1458. [PMID: 37066837 DOI: 10.1002/jbm.a.37545] [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: 09/21/2022] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/18/2023]
Abstract
Human mesenchymal stromal cells (hMSCs) are of significant interest as a renewable source of therapeutically useful cells. In tissue engineering, hMSCs are implanted within a scaffold to provide enhanced capacity for tissue repair. The present study evaluates how mechanical properties of that scaffold can alter the phenotype and genotype of the cells, with the aim of augmenting hMSC differentiation along the myogenic, neurogenic or chondrogenic linages. The hMSCs were grown three-dimensionally (3D) in a hydrogel comprised of poly(ethylene glycol) (PEG)-conjugated to fibrinogen. The hydrogel's shear storage modulus (G'), which was controlled by increasing the amount of PEG-diacrylate cross-linker in the matrix, was varied in the range of 100-2000 Pascal (Pa). The differentiation into each lineage was initiated by a defined culture medium, and the hMSCs grown in the different modulus hydrogels were characterized using gene and protein expression. Materials having lower storage moduli (G' = 100 Pa) exhibited more hMSCs differentiating to neurogenic lineages. Myogenesis was favored in materials having intermediate modulus values (G' = 500 Pa), whereas chondrogenesis was favored in materials with a higher modulus (G' = 1000 Pa). Enhancing the differentiation pathway of hMSCs in 3D hydrogel scaffolds using simple modifications to mechanical properties represents an important achievement toward the effective application of these cells in tissue engineering.
Collapse
Affiliation(s)
- Revital Goldshmid
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Haneen Simaan-Yameen
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Liaura Ifergan
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Claudia Loebel
- Materials Science & Engineering Department, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, USA
| | - Dror Seliktar
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
5
|
Atcha H, Choi YS, Chaudhuri O, Engler AJ. Getting physical: Material mechanics is an intrinsic cell cue. Cell Stem Cell 2023; 30:750-765. [PMID: 37267912 PMCID: PMC10247187 DOI: 10.1016/j.stem.2023.05.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/30/2023] [Accepted: 05/02/2023] [Indexed: 06/04/2023]
Abstract
Advances in biomaterial science have allowed for unprecedented insight into the ability of material cues to influence stem cell function. These material approaches better recapitulate the microenvironment, providing a more realistic ex vivo model of the cell niche. However, recent advances in our ability to measure and manipulate niche properties in vivo have led to novel mechanobiological studies in model organisms. Thus, in this review, we will discuss the importance of material cues within the cell niche, highlight the key mechanotransduction pathways involved, and conclude with recent evidence that material cues regulate tissue function in vivo.
Collapse
Affiliation(s)
- Hamza Atcha
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Yu Suk Choi
- School of Human Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.
| |
Collapse
|
6
|
Zhang S, Chong LH, Woon JYX, Chua TX, Cheruba E, Yip AK, Li HY, Chiam KH, Koh CG. Zyxin regulates embryonic stem cell fate by modulating mechanical and biochemical signaling interface. Commun Biol 2023; 6:62. [PMID: 36653484 PMCID: PMC9849324 DOI: 10.1038/s42003-023-04421-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Biochemical signaling and mechano-transduction are both critical in regulating stem cell fate. How crosstalk between mechanical and biochemical cues influences embryonic development, however, is not extensively investigated. Using a comparative study of focal adhesion constituents between mouse embryonic stem cell (mESC) and their differentiated counterparts, we find while zyxin is lowly expressed in mESCs, its levels increase dramatically during early differentiation. Interestingly, overexpression of zyxin in mESCs suppresses Oct4 and Nanog. Using an integrative biochemical and biophysical approach, we demonstrate involvement of zyxin in regulating pluripotency through actin stress fibres and focal adhesions which are known to modulate cellular traction stress and facilitate substrate rigidity-sensing. YAP signaling is identified as an important biochemical effector of zyxin-induced mechanotransduction. These results provide insights into the role of zyxin in the integration of mechanical and biochemical cues for the regulation of embryonic stem cell fate.
Collapse
Affiliation(s)
- Songjing Zhang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lor Huai Chong
- Bioinformatics Institute A*STAR, Singapore, Singapore.,School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jessie Yong Xing Woon
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Theng Xuan Chua
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Ai Kia Yip
- Bioinformatics Institute A*STAR, Singapore, Singapore
| | - Hoi-Yeung Li
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Cheng-Gee Koh
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
7
|
A maladaptive feedback mechanism between the extracellular matrix and cytoskeleton contributes to hypertrophic cardiomyopathy pathophysiology. Commun Biol 2023; 6:4. [PMID: 36596888 PMCID: PMC9810744 DOI: 10.1038/s42003-022-04278-9] [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: 02/22/2022] [Accepted: 11/17/2022] [Indexed: 01/04/2023] Open
Abstract
Hypertrophic cardiomyopathy is an inherited disorder due to mutations in contractile proteins that results in a stiff, hypercontractile myocardium. To understand the role of cardiac stiffness in disease progression, here we create an in vitro model of hypertrophic cardiomyopathy utilizing hydrogel technology. Culturing wild-type cardiac myocytes on hydrogels with a Young's Moduli (stiffness) mimicking hypertrophic cardiomyopathy myocardium is sufficient to induce a hypermetabolic mitochondrial state versus myocytes plated on hydrogels simulating healthy myocardium. Significantly, these data mirror that of myocytes isolated from a murine model of human hypertrophic cardiomyopathy (cTnI-G203S). Conversely, cTnI-G203S myocyte mitochondrial function is completely restored when plated on hydrogels mimicking healthy myocardium. We identify a mechanosensing feedback mechanism between the extracellular matrix and cytoskeletal network that regulates mitochondrial function under healthy conditions, but participates in the progression of hypertrophic cardiomyopathy pathophysiology resulting from sarcomeric gene mutations. Importantly, we pinpoint key 'linker' sites in this schema that may represent potential therapeutic targets.
Collapse
|
8
|
Bohere J, Eldridge-Thomas BL, Kolahgar G. Vinculin recruitment to α-catenin halts the differentiation and maturation of enterocyte progenitors to maintain homeostasis of the Drosophila intestine. eLife 2022; 11:e72836. [PMID: 36269226 PMCID: PMC9586559 DOI: 10.7554/elife.72836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/11/2022] [Indexed: 11/23/2022] Open
Abstract
Mechanisms communicating changes in tissue stiffness and size are particularly relevant in the intestine because it is subject to constant mechanical stresses caused by peristalsis of its variable content. Using the Drosophila intestinal epithelium, we investigate the role of vinculin, one of the best characterised mechanoeffectors, which functions in both cadherin and integrin adhesion complexes. We discovered that vinculin regulates cell fate decisions, by preventing precocious activation and differentiation of intestinal progenitors into absorptive cells. It achieves this in concert with α-catenin at sites of cadherin adhesion, rather than as part of integrin function. Following asymmetric division of the stem cell into a stem cell and an enteroblast (EB), the two cells initially remain connected by adherens junctions, where vinculin is required, only on the EB side, to maintain the EB in a quiescent state and inhibit further divisions of the stem cell. By manipulating cell tension, we show that vinculin recruitment to adherens junction regulates EB activation and numbers. Consequently, removing vinculin results in an enlarged gut with improved resistance to starvation. Thus, mechanical regulation at the contact between stem cells and their progeny is used to control tissue cell number.
Collapse
Affiliation(s)
- Jerome Bohere
- Department of Physiology, Development and Neuroscience, Downing St, University of CambridgeCambridgeUnited Kingdom
| | - Buffy L Eldridge-Thomas
- Department of Physiology, Development and Neuroscience, Downing St, University of CambridgeCambridgeUnited Kingdom
| | - Golnar Kolahgar
- Department of Physiology, Development and Neuroscience, Downing St, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
9
|
Lim R, Banerjee A, Biswas R, Chari AN, Raghavan S. Mechanotransduction through adhesion molecules: Emerging roles in regulating the stem cell niche. Front Cell Dev Biol 2022; 10:966662. [PMID: 36172276 PMCID: PMC9511051 DOI: 10.3389/fcell.2022.966662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Stem cells have been shown to play an important role in regenerative medicine due to their proliferative and differentiation potential. The challenge, however, lies in regulating and controlling their potential for this purpose. Stem cells are regulated by growth factors as well as an array of biochemical and mechanical signals. While the role of biochemical signals and growth factors in regulating stem cell homeostasis is well explored, the role of mechanical signals has only just started to be investigated. Stem cells interact with their niche or to other stem cells via adhesion molecules that eventually transduce mechanical cues to maintain their homeostatic function. Here, we present a comprehensive review on our current understanding of the influence of the forces perceived by cell adhesion molecules on the regulation of stem cells. Additionally, we provide insights on how this deeper understanding of mechanobiology of stem cells has translated toward therapeutics.
Collapse
Affiliation(s)
- Ryan Lim
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Avinanda Banerjee
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Ritusree Biswas
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
- Sastra University, Thanjavur, TN, India
| | - Anana Nandakumar Chari
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Srikala Raghavan
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
| |
Collapse
|
10
|
Hu D, Dong Z, Li B, Lu F, Li Y. Mechanical Force Directs Proliferation and Differentiation of Stem Cells. TISSUE ENGINEERING PART B: REVIEWS 2022; 29:141-150. [PMID: 35979892 DOI: 10.1089/ten.teb.2022.0052] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Stem cells have attracted much attention in the field of regeneration due to their unique ability to promote regeneration. Among the many approaches used to regulate directed proliferation and differentiation of stem cells, application of mechanical forces is safe, simple, and easy to implement, all of which are advantageous to practical applications. In this review, the mechanisms of mechanical regulation of stem cell proliferation and differentiation are summarized with emphasis on force transduction pathways from the extracellular matrix to the nucleus. Prospects for future clinical applications are also discussed. In conclusion, through specific signaling pathways, mechanical signals ultimately affect gene expression and thus guide cell fate. Mechanical factors can regulate proliferation and differentiation of stem cells through signaling pathways, a greater understanding of which will contribute to future research and applications of cell regeneration therapy. Impact statement Mechanical mechanics is vital for the regulation of cell fate; especially in the field of regenerative medicine, mechanical control has characteristics that are simple and comparable. Mechanically regulated pathways exist widely in cells and are distributed at various structural levels of cells. In this review, we categorized the mechanical regulatory pathways through the clue of the mechanical transmission. We tried to include some newly researched pathways, such as Piezo-related pathways, to show the recent vigorous development in this field.
Collapse
Affiliation(s)
- Delin Hu
- Southern Medical University Nanfang Hospital, Department of Plastic and Cosmetic Surgery, Guangzhou, Guangdong, China,
| | - Ziqing Dong
- Southern Medical University Nanfang Hospital, Department of Plastic and Cosmetic Surgery, Guangzhou, Guangdong, China,
| | - Bin Li
- Southern Medical University Nanfang Hospital, Department of Plastic and Cosmetic Surgery, Guangzhou, Guangdong, China,
| | - Feng Lu
- Southern Medical University Nanfang Hospital, Department of Plastic and Cosmetic Surgery, Guangzhou, Guangdong, China,
| | - Ye Li
- Southern Medical University Nanfang Hospital, Plastic and Cosmetic Surgery, guangzhou, Guangzhou, China, 510515,
| |
Collapse
|
11
|
Wang M, Ivanovska I, Vashisth M, Discher DE. Nuclear mechanoprotection: From tissue atlases as blueprints to distinctive regulation of nuclear lamins. APL Bioeng 2022; 6:021504. [PMID: 35719698 PMCID: PMC9203124 DOI: 10.1063/5.0080392] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/23/2022] [Indexed: 11/14/2022] Open
Abstract
Two meters of DNA in each of our cells must be protected against many types of damage. Mechanoprotection is increasingly understood to be conferred by the nuclear lamina of intermediate filament proteins, but very different patterns of expression and regulation between different cells and tissues remain a challenge to comprehend and translate into applications. We begin with a tutorial style presentation of "tissue blueprints" of lamin expression including single-cell RNA sequencing in major public datasets. Lamin-A, C profiles appear strikingly similar to those for the mechanosensitive factors Vinculin, Yap1, and Piezo1, whereas datasets for lamin-B1 align with and predict regulation by the cell cycle transcription factor, FOXM1, and further predict poor survival across multiple cancers. Various experiments support the distinction between the lamin types and add mechanistic insight into the mechano-regulation of lamin-A, C by both matrix elasticity and externally imposed tissue strain. Both A- and B-type lamins, nonetheless, protect the nucleus from rupture and damage. Ultimately, for mechanically active tissue constructs and organoids as well as cell therapies, lamin levels require particular attention as they help minimize nuclear damage and defects in a cell cycle.
Collapse
|
12
|
López-Jiménez C, Chiu LL, Waldman SD, Guilak F, Koch TG. TRPV4 activation enhances compressive properties and glycosaminoglycan deposition of equine neocartilage sheets. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100263. [DOI: 10.1016/j.ocarto.2022.100263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/10/2022] [Accepted: 04/11/2022] [Indexed: 11/24/2022] Open
|
13
|
Xu Y, Xin R, Sun H, Long D, Li Z, Liao H, Xue T, Zhang Z, Kang Y, Mao G. Long Non-coding RNAs LOC100126784 and POM121L9P Derived From Bone Marrow Mesenchymal Stem Cells Enhance Osteogenic Differentiation via the miR-503-5p/SORBS1 Axis. Front Cell Dev Biol 2021; 9:723759. [PMID: 34746123 PMCID: PMC8570085 DOI: 10.3389/fcell.2021.723759] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/30/2021] [Indexed: 12/22/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) play pivotal roles in mesenchymal stem cell differentiation. However, the mechanisms by which non-coding RNA (ncRNA) networks regulate osteogenic differentiation remain unclear. Therefore, our aim was to identify RNA-associated gene and transcript expression profiles during osteogenesis in bone marrow mesenchymal stem cells (BMSCs). Using transcriptome sequencing for differentially expressed ncRNAs and mRNAs between days 0 and 21 of osteogenic differentiation of BMSCs, we found that the microRNA (miRNA) miR-503-5p was significantly downregulated. However, the putative miR-503-5p target, sorbin and SH3 domain containing 1 (SORBS1), was significantly upregulated in osteogenesis. Moreover, through lncRNA-miRNA-mRNA interaction analyses and loss- and gain-of-function experiments, we discovered that the lncRNAs LOC100126784 and POM121L9P were abundant in the cytoplasm and enhanced BMSC osteogenesis by promoting SORBS1 expression. In contrast, miR-503-5p reversed this effect. Ago2 RNA-binding protein immunoprecipitation and dual-luciferase reporter assays further validated the direct binding of miR-503-5p to LOC100126784 and POM121L9P. Furthermore, SORBS1 knockdown suppressed early osteogenic differentiation in BMSCs, and co-transfection with SORBS1 small interfering RNAs counteracted the BMSCs’ osteogenic capacity promoted by LOC100126784- and POM121L9P-overexpressing lentivirus plasmids. Thus, the present study demonstrated that the lncRNAs LOC100126784 and POM121L9P facilitate the osteogenic differentiation of BMSCs via the miR-503-5p/SORBS1 axis, providing potential therapeutic targets for treating osteoporosis and bone defects.
Collapse
Affiliation(s)
- Yiyang Xu
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China.,Department of Orthopedics, Shengli Clinical Medical College, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, China
| | - Ruobing Xin
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Hong Sun
- Department of Orthopaedics, Affiliated Hospital of Guizhou Medical University Guiyang, Guizhou, China
| | - Dianbo Long
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Zhiwen Li
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Hongyi Liao
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Ting Xue
- Fujian Provincial Hospital South Branch, Center of Health Management, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Ziji Zhang
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Yan Kang
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Guping Mao
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| |
Collapse
|
14
|
Wang DY, Melero C, Albaraky A, Atherton P, Jansen KA, Dimitracopoulos A, Dajas-Bailador F, Reid A, Franze K, Ballestrem C. Vinculin is required for neuronal mechanosensing but not for axon outgrowth. Exp Cell Res 2021; 407:112805. [PMID: 34487728 DOI: 10.1016/j.yexcr.2021.112805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/19/2021] [Accepted: 08/21/2021] [Indexed: 11/29/2022]
Abstract
Integrin receptors are transmembrane proteins that bind to the extracellular matrix (ECM). In most animal cell types integrins cluster together with adaptor proteins at focal adhesions that sense and respond to external mechanical signals. In the central nervous system (CNS), ECM proteins are sparsely distributed, the tissue is comparatively soft and neurons do not form focal adhesions. Thus, how neurons sense tissue stiffness is currently poorly understood. Here, we found that integrins and the integrin-associated proteins talin and focal adhesion kinase (FAK) are required for the outgrowth of neuronal processes. Vinculin, however, whilst not required for neurite outgrowth was a key regulator of integrin-mediated mechanosensing of neurons. During growth, growth cones of axons of CNS derived cells exerted dynamic stresses of around 10-12 Pa on their environment, and axons grew significantly longer on soft (0.4 kPa) compared to stiff (8 kPa) substrates. Depletion of vinculin blocked this ability of growth cones to distinguish between soft and stiff substrates. These data suggest that vinculin in neurons acts as a key mechanosensor, involved in the regulation of growth cone motility.
Collapse
Affiliation(s)
- De-Yao Wang
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health. The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Cristina Melero
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health. The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Ashwaq Albaraky
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health. The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Paul Atherton
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health. The University of Manchester, Oxford Road, Manchester, M13 9PT, UK; Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre. Manchester, M13 9PT, UK
| | - Karin A Jansen
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health. The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Andrea Dimitracopoulos
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | | | - Adam Reid
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre. Manchester, M13 9PT, UK; Department of Plastic Surgery & Nurns, Wythenshawe Hospital, Manchester University NHS Foundation Trust. Manchester Academic Health Science Centre, Manchester, M23 9LT, UK
| | - Kristian Franze
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK; Institute of Medical Physics, Friedrich-Alexander University Erlangen-Nuremberg, 91052, Erlangen, Germany; Max-Planck-Zentrum für Physik und Medizin, 91054, Erlangen, Germany
| | - Christoph Ballestrem
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health. The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
15
|
Kocsis Á, Pasztorek M, Rossmanith E, Djinovic Z, Mayr T, Spitz S, Zirath H, Ertl P, Fischer MB. Dependence of mitochondrial function on the filamentous actin cytoskeleton in cultured mesenchymal stem cells treated with cytochalasin B. J Biosci Bioeng 2021; 132:310-320. [PMID: 34175199 DOI: 10.1016/j.jbiosc.2021.05.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 12/28/2022]
Abstract
Owing to their self-renewal and multi-lineage differentiation capability, mesenchymal stem cells (MSCs) hold enormous potential in regenerative medicine. A prerequisite for a successful MSC therapy is the rigorous investigation of their function after in vitro cultivation. Damages introduced to mitochondria during cultivation adversely affect MSCs function and can determine their fate. While it has been shown that microtubules and vimentin intermediate filaments are important for mitochondrial dynamics and active mitochondrial transport within the cytoplasm of MSCs, the role of filamentous actin in this process has not been fully understood yet. To gain a deeper understanding of the interdependence between mitochondrial function and the cytoskeleton, we applied cytochalasin B to disturb the filamentous actin-based cytoskeleton of MSCs. In this study we combined conventional functional assays with a state-of-the-art oxygen sensor-integrated microfluidic device to investigate mitochondrial function. We demonstrated that cytochalasin B treatment at a dose of 16 μM led to a decrease in cell viability with high mitochondrial membrane potential, increased oxygen consumption rate, disturbed fusion and fission balance, nuclear extrusion and perinuclear accumulation of mitochondria. Treatment of MSCs for 48 h ultimately led to nuclear fragmentation, and activation of the intrinsic pathway of apoptotic cell death. Importantly, we could show that mitochondrial function of MSCs can efficiently recover from the damage to the filamentous actin-based cytoskeleton over a period of 24 h. As a result of our study, a causative connection between the filamentous actin-based cytoskeleton and mitochondrial dynamics was demonstrated.
Collapse
Affiliation(s)
- Ágnes Kocsis
- Department for Biomedical Research, Center of Experimental Medicine, Danube University Krems, Dr.-Karl-Dorrek-Straße 30, Krems an der Donau 3500, Austria
| | - Markus Pasztorek
- Department for Biomedical Research, Center of Experimental Medicine, Danube University Krems, Dr.-Karl-Dorrek-Straße 30, Krems an der Donau 3500, Austria
| | - Eva Rossmanith
- Department for Biomedical Research, Center of Experimental Medicine, Danube University Krems, Dr.-Karl-Dorrek-Straße 30, Krems an der Donau 3500, Austria
| | - Zoran Djinovic
- ACMIT Gmbh (Austrian Center for Medical Innovation and Technology), Viktor Kaplan-Straße 2/1, Wiener Neustadt 2700, Austria
| | - Torsten Mayr
- Institute of Analytical Chemistry and Food Chemistry, Graz University of Technology, Stremayrgasse 9 / II + III, Graz 8010, Austria
| | - Sarah Spitz
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163, Vienna 1060, Austria
| | - Helene Zirath
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163, Vienna 1060, Austria
| | - Peter Ertl
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163, Vienna 1060, Austria
| | - Michael B Fischer
- Department for Biomedical Research, Center of Experimental Medicine, Danube University Krems, Dr.-Karl-Dorrek-Straße 30, Krems an der Donau 3500, Austria; Clinic for Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Vienna 1090, Austria.
| |
Collapse
|
16
|
Warren D, Tomaskovic-Crook E, Wallace GG, Crook JM. Engineering in vitro human neural tissue analogs by 3D bioprinting and electrostimulation. APL Bioeng 2021; 5:020901. [PMID: 33834152 PMCID: PMC8019355 DOI: 10.1063/5.0032196] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/19/2021] [Indexed: 02/06/2023] Open
Abstract
There is a fundamental need for clinically relevant, reproducible, and standardized in vitro human neural tissue models, not least of all to study heterogenic and complex human-specific neurological (such as neuropsychiatric) disorders. Construction of three-dimensional (3D) bioprinted neural tissues from native human-derived stem cells (e.g., neural stem cells) and human pluripotent stem cells (e.g., induced pluripotent) in particular is appreciably impacting research and conceivably clinical translation. Given the ability to artificially and favorably regulate a cell's survival and behavior by manipulating its biophysical environment, careful consideration of the printing technique, supporting biomaterial and specific exogenously delivered stimuli, is both required and advantageous. By doing so, there exists an opportunity, more than ever before, to engineer advanced and precise tissue analogs that closely recapitulate the morphological and functional elements of natural tissues (healthy or diseased). Importantly, the application of electrical stimulation as a method of enhancing printed tissue development in vitro, including neuritogenesis, synaptogenesis, and cellular maturation, has the added advantage of modeling both traditional and new stimulation platforms, toward improved understanding of efficacy and innovative electroceutical development and application.
Collapse
Affiliation(s)
- Danielle Warren
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Fairy Meadow, NSW 2519 Australia
| | | | - Gordon G. Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Fairy Meadow, NSW 2519 Australia
| | - Jeremy M. Crook
- Author to whom correspondence should be addressed:. Tel.: +61 2 4221 3011
| |
Collapse
|
17
|
Dey K, Roca E, Ramorino G, Sartore L. Progress in the mechanical modulation of cell functions in tissue engineering. Biomater Sci 2021; 8:7033-7081. [PMID: 33150878 DOI: 10.1039/d0bm01255f] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In mammals, mechanics at multiple stages-nucleus to cell to ECM-underlie multiple physiological and pathological functions from its development to reproduction to death. Under this inspiration, substantial research has established the role of multiple aspects of mechanics in regulating fundamental cellular processes, including spreading, migration, growth, proliferation, and differentiation. However, our understanding of how these mechanical mechanisms are orchestrated or tuned at different stages to maintain or restore the healthy environment at the tissue or organ level remains largely a mystery. Over the past few decades, research in the mechanical manipulation of the surrounding environment-known as substrate or matrix or scaffold on which, or within which, cells are seeded-has been exceptionally enriched in the field of tissue engineering and regenerative medicine. To do so, traditional tissue engineering aims at recapitulating key mechanical milestones of native ECM into a substrate for guiding the cell fate and functions towards specific tissue regeneration. Despite tremendous progress, a big puzzle that remains is how the cells compute a host of mechanical cues, such as stiffness (elasticity), viscoelasticity, plasticity, non-linear elasticity, anisotropy, mechanical forces, and mechanical memory, into many biological functions in a cooperative, controlled, and safe manner. High throughput understanding of key cellular decisions as well as associated mechanosensitive downstream signaling pathway(s) for executing these decisions in response to mechanical cues, solo or combined, is essential to address this issue. While many reports have been made towards the progress and understanding of mechanical cues-particularly, substrate bulk stiffness and viscoelasticity-in regulating the cellular responses, a complete picture of mechanical cues is lacking. This review highlights a comprehensive view on the mechanical cues that are linked to modulate many cellular functions and consequent tissue functionality. For a very basic understanding, a brief discussion of the key mechanical players of ECM and the principle of mechanotransduction process is outlined. In addition, this review gathers together the most important data on the stiffness of various cells and ECM components as well as various tissues/organs and proposes an associated link from the mechanical perspective that is not yet reported. Finally, beyond addressing the challenges involved in tuning the interplaying mechanical cues in an independent manner, emerging advances in designing biomaterials for tissue engineering are also explored.
Collapse
Affiliation(s)
- Kamol Dey
- Department of Applied Chemistry and Chemical Engineering, Faculty of Science, University of Chittagong, Bangladesh
| | | | | | | |
Collapse
|
18
|
Kim H, Jeong JH, Fendereski M, Lee HS, Kang DY, Hur SS, Amirian J, Kim Y, Pham NT, Suh N, Hwang NSY, Ryu S, Yoon JK, Hwang Y. Heparin-Mimicking Polymer-Based In Vitro Platform Recapitulates In Vivo Muscle Atrophy Phenotypes. Int J Mol Sci 2021; 22:ijms22052488. [PMID: 33801235 PMCID: PMC7957884 DOI: 10.3390/ijms22052488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
The cell–cell/cell–matrix interactions between myoblasts and their extracellular microenvironment have been shown to play a crucial role in the regulation of in vitro myogenic differentiation and in vivo skeletal muscle regeneration. In this study, by harnessing the heparin-mimicking polymer, poly(sodium-4-styrenesulfonate) (PSS), which has a negatively charged surface, we engineered an in vitro cell culture platform for the purpose of recapitulating in vivo muscle atrophy-like phenotypes. Our initial findings showed that heparin-mimicking moieties inhibited the fusion of mononucleated myoblasts into multinucleated myotubes, as indicated by the decreased gene and protein expression levels of myogenic factors, myotube fusion-related markers, and focal adhesion kinase (FAK). We further elucidated the underlying molecular mechanism via transcriptome analyses, observing that the insulin/PI3K/mTOR and Wnt signaling pathways were significantly downregulated by heparin-mimicking moieties through the inhibition of FAK/Cav3. Taken together, the easy-to-adapt heparin-mimicking polymer-based in vitro cell culture platform could be an attractive platform for potential applications in drug screening, providing clear readouts of changes in insulin/PI3K/mTOR and Wnt signaling pathways.
Collapse
Affiliation(s)
- Hyunbum Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea;
| | - Ji Hoon Jeong
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Mona Fendereski
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Hyo-Shin Lee
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Da Yeon Kang
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan-si 31538, Korea; (D.Y.K.); (N.S.)
| | - Sung Sik Hur
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
| | - Jhaleh Amirian
- Institute of Tissue Regeneration, Soonchunhyang University, Asan-si 31538, Korea;
| | - Yunhye Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Nghia Thi Pham
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Nayoung Suh
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan-si 31538, Korea; (D.Y.K.); (N.S.)
| | - Nathaniel Suk-Yeon Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Korea;
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
| | - Jeong Kyo Yoon
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
- Correspondence: (J.K.Y.); (Y.H.); Tel.: +82-41-413-5016 (J.K.Y.); +82-41-413-5017 (Y.H.)
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-si 31151, Korea; (H.K.); (J.H.J.); (M.F.); (H.-S.L.); (S.S.H.); (Y.K.); (N.T.P.); (S.R.)
- Department of Integrated Biomedical Science, Soonchunhyang University, Asan-si 31538, Korea
- Correspondence: (J.K.Y.); (Y.H.); Tel.: +82-41-413-5016 (J.K.Y.); +82-41-413-5017 (Y.H.)
| |
Collapse
|
19
|
Wang S, Hashemi S, Stratton S, Arinzeh TL. The Effect of Physical Cues of Biomaterial Scaffolds on Stem Cell Behavior. Adv Healthc Mater 2021; 10:e2001244. [PMID: 33274860 DOI: 10.1002/adhm.202001244] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Stem cells have been sought as a promising cell source in the tissue engineering field due to their proliferative capacity as well as differentiation potential. Biomaterials have been utilized to facilitate the delivery of stem cells in order to improve their engraftment and long-term viability upon implantation. Biomaterials also have been developed as scaffolds to promote stem cell induced tissue regeneration. This review focuses on the latter where the biomaterial scaffold is designed to provide physical cues to stem cells in order to promote their behavior for tissue formation. Recent work that explores the effect of scaffold physical properties, topography, mechanical properties and electrical properties, is discussed. Although still being elucidated, the biological mechanisms, including cell shape, focal adhesion distribution, and nuclear shape, are presented. This review also discusses emerging areas and challenges in clinical translation.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | - Sharareh Hashemi
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | - Scott Stratton
- Department of Biomedical Engineering New Jersey Institute of Technology Newark NJ 07102 USA
| | | |
Collapse
|
20
|
Yu L, Hou Y, Xie W, Cuellar-Camacho JL, Wei Q, Haag R. Self-Strengthening Adhesive Force Promotes Cell Mechanotransduction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2006986. [PMID: 33206452 DOI: 10.1002/adma.202006986] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Indexed: 06/11/2023]
Abstract
The extracellular matrix (ECM) undergoes dynamic remodeling and progressive stiffening during tissue regeneration and disease progression. However, most of the artificial ECMs and in vitro disease models are mechanically static. Here, a self-strengthening polymer coating mimicking the dynamic nature of native ECM is designed to study the cellular response to dynamic biophysical cues and promote cell mechanical sensitive response. Spiropyran (SP) is utilized as dynamic anchor group to regulate the strength of cell adhesive peptide ligands. Benefiting from spontaneous thermal merocyanine-to-spiropyran (MC-SP) isomerization, the resulting self-responsive coating displays dynamic self-strengthening of interfacial interactions. Comparing with the static and all of the previous dynamic artificial ECMs, cells on this self-responsive surface remodel the weakly bonded MC-based coatings to activate α5β1 integrin and Rac signaling in the early adhesion stage. The subsequent MC-to-SP conversion strengthens the ligand-integrin interaction to further activate αvβ3 integrin and RhoA/ROCK signaling in the latter stage. This sequential process enhances cellular mechanotransduction as well as the osteogenic differentiation of mesenchymal stem cells (MSCs). It is worth emphasizing that the self-strengthening occurs spontaneously in the absence of any stimulus, making it especially useful for implanted scaffolds in regenerative medicine.
Collapse
Affiliation(s)
- Leixiao Yu
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Yong Hou
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Wenyan Xie
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, Berlin, 14195, Germany
| | - Jose Luis Cuellar-Camacho
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, China
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| |
Collapse
|
21
|
Parodi V, Jacchetti E, Bresci A, Talone B, Valensise CM, Osellame R, Cerullo G, Polli D, Raimondi MT. Characterization of Mesenchymal Stem Cell Differentiation within Miniaturized 3D Scaffolds through Advanced Microscopy Techniques. Int J Mol Sci 2020; 21:E8498. [PMID: 33187392 PMCID: PMC7696107 DOI: 10.3390/ijms21228498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022] Open
Abstract
Three-dimensional culture systems and suitable substrates topographies demonstrated to drive stem cell fate in vitro by mechanical conditioning. For example, the Nichoid 3D scaffold remodels stem cells and shapes nuclei, thus promoting stem cell expansion and stemness maintenance. However, the mechanisms involved in force transmission and in biochemical signaling at the basis of fate determination are not yet clear. Among the available investigation systems, confocal fluorescence microscopy using fluorescent dyes enables the observation of cell function and shape at the subcellular scale in vital and fixed conditions. Contrarily, nonlinear optical microscopy techniques, which exploit multi-photon processes, allow to study cell behavior in vital and unlabeled conditions. We apply confocal fluorescence microscopy, coherent anti-Stokes Raman scattering (CARS), and second harmonic generation (SHG) microscopy to characterize the phenotypic expression of mesenchymal stem cells (MSCs) towards adipogenic and chondrogenic differentiation inside Nichoid scaffolds, in terms of nuclear morphology and specific phenotypic products, by comparing these techniques. We demonstrate that the Nichoid maintains a rounded nuclei during expansion and differentiation, promoting MSCs adipogenic differentiation while inhibiting chondrogenesis. We show that CARS and SHG techniques are suitable for specific estimation of the lipid and collagenous content, thus overcoming the limitations of using unspecific fluorescent probes.
Collapse
Affiliation(s)
- Valentina Parodi
- Department of Chemistry, Materials and Chemical Engineering «G. Natta», Politecnico di Milano, 20133 Milano, Italy; (E.J.); (A.B.); (M.T.R.)
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering «G. Natta», Politecnico di Milano, 20133 Milano, Italy; (E.J.); (A.B.); (M.T.R.)
| | - Arianna Bresci
- Department of Chemistry, Materials and Chemical Engineering «G. Natta», Politecnico di Milano, 20133 Milano, Italy; (E.J.); (A.B.); (M.T.R.)
- Department of Physics, Politecnico di Milano, 20133 Milano, Italy; (B.T.); (C.M.V.); (R.O.); (G.C.); (D.P.)
- Istituto di Fotonica e Nanotecnologie (IFN), Consiglio Nazionale delle Ricerche (CNR), 20133 Milano, Italy
| | - Benedetta Talone
- Department of Physics, Politecnico di Milano, 20133 Milano, Italy; (B.T.); (C.M.V.); (R.O.); (G.C.); (D.P.)
- Istituto di Fotonica e Nanotecnologie (IFN), Consiglio Nazionale delle Ricerche (CNR), 20133 Milano, Italy
| | - Carlo M. Valensise
- Department of Physics, Politecnico di Milano, 20133 Milano, Italy; (B.T.); (C.M.V.); (R.O.); (G.C.); (D.P.)
- Istituto di Fotonica e Nanotecnologie (IFN), Consiglio Nazionale delle Ricerche (CNR), 20133 Milano, Italy
| | - Roberto Osellame
- Department of Physics, Politecnico di Milano, 20133 Milano, Italy; (B.T.); (C.M.V.); (R.O.); (G.C.); (D.P.)
- Istituto di Fotonica e Nanotecnologie (IFN), Consiglio Nazionale delle Ricerche (CNR), 20133 Milano, Italy
| | - Giulio Cerullo
- Department of Physics, Politecnico di Milano, 20133 Milano, Italy; (B.T.); (C.M.V.); (R.O.); (G.C.); (D.P.)
- Istituto di Fotonica e Nanotecnologie (IFN), Consiglio Nazionale delle Ricerche (CNR), 20133 Milano, Italy
| | - Dario Polli
- Department of Physics, Politecnico di Milano, 20133 Milano, Italy; (B.T.); (C.M.V.); (R.O.); (G.C.); (D.P.)
- Istituto di Fotonica e Nanotecnologie (IFN), Consiglio Nazionale delle Ricerche (CNR), 20133 Milano, Italy
| | - Manuela T. Raimondi
- Department of Chemistry, Materials and Chemical Engineering «G. Natta», Politecnico di Milano, 20133 Milano, Italy; (E.J.); (A.B.); (M.T.R.)
| |
Collapse
|
22
|
In Vitro Evidences of Different Fibroblast Morpho-Functional Responses to Red, Near-Infrared and Violet-Blue Photobiomodulation: Clues for Addressing Wound Healing. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10217878] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although photobiomodulation (PBM) has proven promising to treat wounds, the lack of univocal guidelines and of a thorough understanding of light–tissue interactions hampers its mainstream adoption for wound healing promotion. This study compared murine and human fibroblast responses to PBM by red (635 ± 5 nm), near-infrared (NIR, 808 ± 1 nm), and violet-blue (405 ± 5 nm) light (0.4 J/cm2 energy density, 13 mW/cm2 power density). Cell viability was not altered by PBM treatments. Light and confocal laser scanning microscopy and biochemical analyses showed, in red PBM irradiated cells: F-actin assembly reduction, up-regulated expression of Ki67 proliferation marker and of vinculin in focal adhesions, type-1 collagen down-regulation, matrix metalloproteinase-2 and metalloproteinase-9 expression/functionality increase concomitant to their inhibitors (TIMP-1 and TIMP-2) decrease. Violet-blue and even more NIR PBM stimulated collagen expression/deposition and, likely, cell differentiation towards (proto)myofibroblast phenotype. Indeed, these cells exhibited a higher polygonal surface area, stress fiber-like structures, increased vinculin- and phospho-focal adhesion kinase-rich clusters and α-smooth muscle actin. This study may provide the experimental groundwork to support red, NIR, and violet-blue PBM as potential options to promote proliferative and matrix remodeling/maturation phases of wound healing, targeting fibroblasts, and to suggest the use of combined PBM treatments in the wound management setting.
Collapse
|
23
|
Yu L, Hou Y, Xie W, Camacho JLC, Cheng C, Holle A, Young J, Trappmann B, Zhao W, Melzig MF, Cavalcanti-Adam EA, Zhao C, Spatz JP, Wei Q, Haag R. Ligand Diffusion Enables Force-Independent Cell Adhesion via Activating α5β1 Integrin and Initiating Rac and RhoA Signaling. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002566. [PMID: 32537880 DOI: 10.1002/adma.202002566] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/12/2020] [Indexed: 05/12/2023]
Abstract
Cells reside in a dynamic microenvironment in which adhesive ligand availability, density, and diffusivity are key factors regulating cellular behavior. Here, the cellular response to integrin-binding ligand dynamics by directly controlling ligand diffusivity via tunable ligand-surface interactions is investigated. Interestingly, cell spread on the surfaces with fast ligand diffusion is independent of myosin-based force generation. Fast ligand diffusion enhances α5β1 but not αvβ3 integrin activation and initiates Rac and RhoA but not ROCK signaling, resulting in lamellipodium-based fast cell spreading. Meanwhile, on surfaces with immobile ligands, αvβ3 and α5β1 integrins synergistically initiate intracellular-force-based canonical mechanotransduction pathways to enhance cell adhesion and osteogenic differentiation of stem cells. These results indicate the presence of heretofore-unrecognized pathways, distinct from canonical actomyosin-driven mechanisms, that are capable of promoting cell adhesion.
Collapse
Affiliation(s)
- Leixiao Yu
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Yong Hou
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Wenyan Xie
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, Berlin, 14195, Germany
| | - Jose Luis Cuellar Camacho
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Chong Cheng
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Andrew Holle
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, Heidelberg, 69120, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, Heidelberg, 69120, Germany
| | - Jennifer Young
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, Heidelberg, 69120, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, Heidelberg, 69120, Germany
| | - Britta Trappmann
- Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, Münster, 48149, Germany
| | - Weifeng Zhao
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Matthias F Melzig
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, Berlin, 14195, Germany
| | - Elisabetta A Cavalcanti-Adam
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, Heidelberg, 69120, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, Heidelberg, 69120, Germany
- Central Scientific Facility "Cellular Biotechnology", Jahnstr. 29, Heidelberg, 69120, Germany
| | - Changsheng Zhao
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, Heidelberg, 69120, Germany
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, Heidelberg, 69120, Germany
| | - Qiang Wei
- State Key Laboratory of Polymer Materials and Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| |
Collapse
|
24
|
Zhao C, Song X, Lu X. Directional Osteo-Differentiation Effect of hADSCs on Nanotopographical Self-Assembled Polystyrene Nanopit Surfaces. Int J Nanomedicine 2020; 15:3281-3290. [PMID: 32440124 PMCID: PMC7217320 DOI: 10.2147/ijn.s240300] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/10/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Cells exhibit high sensitivity and a diverse response to the nanotopography of the extracellular matrix, thereby endowing materials with instructive performances formerly reserved for growth factors. This finding leads to opportunities for improvement. However, the interplay between the topographical surface and cell behaviors remains incompletely understood. Methods In the present study, we showed nanosurfaces with various dimensions of nanopits (200–750 nm) fabricated by self-assembling polystyrene (PS) nanospheres. Human adipose-derived stem cell behaviors, such as cell morphology, adhesion, cytoskeleton contractility, proliferation, and differentiation, were investigated on the prepared PS nanopit surface. Results The osteogenic differentiation can be enhanced by nanopits with a diameter of 300–400 nm. Discussion The present study provided exciting new avenues to investigate cellular responses to well-defined nanoscale topographic features, which could further guide bone tissue engineering and stem cell clinical research. The capability to control developing biomaterials mimicking nanotopographic surfaces promoted functional tissue engineering, such as artificial joint replacement, bone repair, and dental applications.
Collapse
Affiliation(s)
- Changhong Zhao
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, People's Republic of China
| | - Xuebin Song
- School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, People's Republic of China
| | - Xiaoyuan Lu
- College of Medical Engineering, Xinxiang Medical University, Henan 453003, People's Republic of China
| |
Collapse
|
25
|
The role of Piezo proteins and cellular mechanosensing in tuning the fate of transplanted stem cells. Cell Tissue Res 2020; 381:1-12. [DOI: 10.1007/s00441-020-03191-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 02/19/2020] [Indexed: 12/18/2022]
|
26
|
Kluger C, Braun L, Sedlak SM, Pippig DA, Bauer MS, Miller K, Milles LF, Gaub HE, Vogel V. Different Vinculin Binding Sites Use the Same Mechanism to Regulate Directional Force Transduction. Biophys J 2020; 118:1344-1356. [PMID: 32109366 PMCID: PMC7091509 DOI: 10.1016/j.bpj.2019.12.042] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/17/2019] [Accepted: 12/30/2019] [Indexed: 12/18/2022] Open
Abstract
Vinculin is a universal adaptor protein that transiently reinforces the mechanical stability of adhesion complexes. It stabilizes mechanical connections that cells establish between the actomyosin cytoskeleton and the extracellular matrix via integrins or to neighboring cells via cadherins, yet little is known regarding its mechanical design. Vinculin binding sites (VBSs) from different nonhomologous actin-binding proteins use conserved helical motifs to associate with the vinculin head domain. We studied the mechanical stability of such complexes by pulling VBS peptides derived from talin, α-actinin, and Shigella IpaA out of the vinculin head domain. Experimental data from atomic force microscopy single-molecule force spectroscopy and steered molecular dynamics (SMD) simulations both revealed greater mechanical stability of the complex for shear-like than for zipper-like pulling configurations. This suggests that reinforcement occurs along preferential force directions, thus stabilizing those cytoskeletal filament architectures that result in shear-like pulling geometries. Large force-induced conformational changes in the vinculin head domain, as well as protein-specific fine-tuning of the VBS sequence, including sequence inversion, allow for an even more nuanced force response.
Collapse
Affiliation(s)
- Carleen Kluger
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lukas Braun
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Steffen M Sedlak
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Diana A Pippig
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Magnus S Bauer
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ken Miller
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lukas F Milles
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hermann E Gaub
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
27
|
An S, Han SY, Cho SW. Hydrogel-integrated Microfluidic Systems for Advanced Stem Cell Engineering. BIOCHIP JOURNAL 2019. [DOI: 10.1007/s13206-019-3402-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
28
|
Emerging Concepts and Tools in Cell Mechanomemory. Ann Biomed Eng 2019; 48:2103-2112. [DOI: 10.1007/s10439-019-02412-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/11/2019] [Indexed: 12/25/2022]
|
29
|
Pasztorek M, Rossmanith E, Mayr C, Hauser F, Jacak J, Ebner A, Weber V, Fischer MB. Influence of Platelet Lysate on 2D and 3D Amniotic Mesenchymal Stem Cell Cultures. Front Bioeng Biotechnol 2019; 7:338. [PMID: 31803733 PMCID: PMC6873824 DOI: 10.3389/fbioe.2019.00338] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/30/2019] [Indexed: 01/16/2023] Open
Abstract
The mechanobiological behavior of mesenchymal stem cells (MSCs) in two- (2D) or three-dimensional (3D) cultures relies on the formation of actin filaments which occur as stress fibers and depends on mitochondrial dynamics involving vimentin intermediate filaments. Here we investigate whether human platelet lysate (HPL), that can potentially replace fetal bovine serum for clinical-scale expansion of functional cells, can modulate the stress fiber formation, alter mitochondrial morphology, change membrane elasticity and modulate immune regulatory molecules IDO and GARP in amnion derived MSCs. We can provide evidence that culture supplementation with HPL led to a reduction of stress fiber formation in 2D cultured MSCs compared to a conventional growth medium (MSCGM). 3D MSC cultures, in contrast, showed decreased actin concentrations independent of HPL supplementation. When stress fibers were further segregated by their binding to focal adhesions, a reduction in ventral stress fibers was observed in response to HPL in 2D cultured MSCs, while the length of the individual ventral stress fibers increased. Dorsal stress fibers or transverse arcs were not affected. Interestingly, ventral stress fiber formation did not correlate with membrane elasticity. 2D cultured MSCs did not show differences in the Young's modulus when propagated in the presence of HPL and further cultivation to passage 3 also had no effect on membrane elasticity. In addition, HPL reduced the mitochondrial mass of 2D cultured MSCs while the mitochondrial mass in 3D cultured MSCs was low initially. When mitochondria were segregated into punctuate, rods and networks, a cultivation-induced increase in punctuate and network mitochondria was observed in 2D cultured MSCs of passage 3. Finally, mRNA and protein expression of the immunomodulatory molecule IDO relied on stimulation of 2D culture MSCs with pro-inflammatory cytokines IFN-γ and TNF-α with no effect upon HPL supplementation. GARP mRNA and surface expression was constitutively expressed and did not respond to HPL supplementation or stimulation with IFN-γ and TNF-α. In conclusion, we can say that MSCs cultivated in 2D and 3D are sensitive to medium supplementation with HPL with changes in actin filament formation, mitochondrial dynamics and membrane elasticity that can have an impact on the immunomodulatory function of MSCs.
Collapse
Affiliation(s)
- Markus Pasztorek
- Department for Biomedical Research, Center of Experimental Medicine, Danube University Krems, Krems an der Donau, Austria
| | - Eva Rossmanith
- Department for Biomedical Research, Center of Experimental Medicine, Danube University Krems, Krems an der Donau, Austria
| | - Christoph Mayr
- Department of Applied Experimental Biophysics, Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Fabian Hauser
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Linz, Austria
| | - Jaroslaw Jacak
- School of Medical Engineering and Applied Social Sciences, University of Applied Sciences Upper Austria, Linz, Austria
| | - Andreas Ebner
- Department of Applied Experimental Biophysics, Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Viktoria Weber
- Department for Biomedical Research, Center of Experimental Medicine, Danube University Krems, Krems an der Donau, Austria
- Christian Doppler Laboratories, Department for Biomedical Research, Danube University Krems, Krems an der Donau, Austria
| | - Michael B. Fischer
- Department for Biomedical Research, Center of Experimental Medicine, Danube University Krems, Krems an der Donau, Austria
- Christian Doppler Laboratories, Department for Biomedical Research, Danube University Krems, Krems an der Donau, Austria
| |
Collapse
|
30
|
Wong L, Kumar A, Gabela-Zuniga B, Chua J, Singh G, Happe CL, Engler AJ, Fan Y, McCloskey KE. Substrate stiffness directs diverging vascular fates. Acta Biomater 2019; 96:321-329. [PMID: 31326665 DOI: 10.1016/j.actbio.2019.07.030] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/16/2019] [Accepted: 07/16/2019] [Indexed: 12/16/2022]
Abstract
Embryonic stem cells (ESC) are excellent cell culture systems for elucidating developmental signals that may be part of the stem cell niche. Although stem cells are traditionally induced using predominately soluble signals, the mechanical environment of the niche can also play a role in directing cells towards differential cell lineages. Interested in diverging vascular fates, we set out to examine to what extent mechanical signaling played a role in endothelial cell and/or smooth muscle fate. Using chemically-defined staged vascular differentiation methods, vascular progenitor cells (VPC) fate was examined on single stiffness polyacrylamide hydrogels of 10 kPa, 40 kPa and >0.1 GPa. Emergence of vascular cell populations aligned with corresponding hydrogel stiffness: EC-lineages favoring the softer material and SMC lineages favoring the stiffest material. Statistical significance was observed on both cell lines on almost all days. Transcriptome analysis indicated that the populations on the varying stiffness emerge in distinct categories. Lastly, blocking studies show that αvβ1, and not αvβ6, activation mediates stiffness-directed vascular differentiation. Overall, these studies indicate that softer materials direct VPCs into a more EC-like fate compared to stiffer materials. STATEMENT OF SIGNIFICANCE: Although stem cells are traditionally induced using predominately soluble signals, the mechanical environment of the niche also plays a role in directing cell fate. Several studies have examined the stiffness-induced cell fate from mesenchymal stem cells (MSCs) and undifferentiated embryonic stem cells (ESCs). This is the first study that rigorously examines the role of matrix stiffness in diverging vascular fates from a purified population of vascular progenitor cells (VPCs). We show that the emergence of endothelial cell (EC) versus smooth muscle cell (SMC) populations corresponds with hydrogel stiffness: EC-lineages favoring the softness material and SMC lineages favoring the stiffest material, and that αvβ1 activation mediates this stiffness-directed vascular differentiation.
Collapse
|
31
|
Chin IL, Hool L, Choi YS. A Review of in vitro Platforms for Understanding Cardiomyocyte Mechanobiology. Front Bioeng Biotechnol 2019; 7:133. [PMID: 31231644 PMCID: PMC6560053 DOI: 10.3389/fbioe.2019.00133] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022] Open
Abstract
Mechanobiology—a cell's interaction with its physical environment—can influence a myriad of cellular processes including how cells migrate, differentiate and proliferate. In many diseases, remodeling of the extracellular matrix (ECM) is observed such as tissue stiffening in rigid scar formation after myocardial infarct. Utilizing knowledge of cell mechanobiology in relation to ECM remodeling during pathogenesis, elucidating the role of the ECM in the progression—and perhaps regression—of disease is a primary focus of the field. Although the importance of mechanical signaling in the cardiac cell is well-appreciated, our understanding of how these signals are sensed and transduced by cardiomyocytes is limited. To overcome this limitation, recently developed tools and resources have provided exciting opportunities to further our understandings by better recapitulating pathological spatiotemporal ECM stiffness changes in an in vitro setting. In this review, we provide an overview of a conventional model of mechanotransduction and present understandings of cardiomyocyte mechanobiology, followed by a review of emerging tools and resources that can be used to expand our knowledge of cardiomyocyte mechanobiology toward more clinically relevant applications.
Collapse
Affiliation(s)
- Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Livia Hool
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.,Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
32
|
Chen Y, Li Z, Ju LA. Tensile and compressive force regulation on cell mechanosensing. Biophys Rev 2019; 11:311-318. [PMID: 31073958 DOI: 10.1007/s12551-019-00536-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 04/25/2019] [Indexed: 12/14/2022] Open
Abstract
Receptor-mediated cell mechanosensing plays critical roles in cell spreading, migration, growth, and survival. Dynamic force spectroscopy (DFS) techniques have recently been advanced to visualize such processes, which allow the concurrent examination of molecular binding dynamics and cellular response to mechanical stimuli on single living cells. Notably, the live-cell DFS is able to manipulate the force "waveforms" such as tensile versus compressive, ramped versus clamped, static versus dynamic, and short versus long lasting forces, thereby deriving correlations of cellular responses with ligand binding kinetics and mechanical stimulation profiles. Here, by differentiating extracellular mechanical stimulations into two major categories, tensile force and compressive force, we review the latest findings on receptor-mediated mechanosensing mechanisms that are discovered by the state-of-the-art live-cell DFS technologies.
Collapse
Affiliation(s)
- Yunfeng Chen
- Department of Molecular Medicine, MERU-Roon Research Center on Vascular Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Zhiyong Li
- School of Chemistry, Physics and Mechanical Engineering, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Lining Arnold Ju
- Heart Research Institute, Sydney, Australia. .,School of Aerospace, Mechanical and Mechatronic Engineering, Darlington, Australia. .,Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia.
| |
Collapse
|
33
|
Vishnoi T, Singh A, Teotia AK, Kumar A. Chitosan-Gelatin-Polypyrrole Cryogel Matrix for Stem Cell Differentiation into Neural Lineage and Sciatic Nerve Regeneration in Peripheral Nerve Injury Model. ACS Biomater Sci Eng 2019; 5:3007-3021. [DOI: 10.1021/acsbiomaterials.9b00242] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
34
|
Hydrogels with enhanced protein conjugation efficiency reveal stiffness-induced YAP localization in stem cells depends on biochemical cues. Biomaterials 2019; 202:26-34. [PMID: 30826537 DOI: 10.1016/j.biomaterials.2019.02.021] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/24/2019] [Accepted: 02/21/2019] [Indexed: 11/20/2022]
Abstract
Polyacrylamide hydrogels have been widely used in stem cell mechanotransduction studies. Conventional conjugation methods of biochemical cues to polyacrylamide hydrogels suffer from low conjugation efficiency, which leads to poor attachment of human pluripotent stem cells (hPSCs) on soft substrates. In addition, while it is well-established that stiffness-dependent regulation of stem cell fate requires cytoskeletal tension, and is mediated through nuclear translocation of transcription regulator, Yes-associated protein (YAP), the role of biochemical cues in stiffness-dependent YAP regulation remains largely unknown. Here we report a method that enhances the conjugation efficiency of biochemical cues on polyacrylamide hydrogels compared to conventional methods. This modified method enables robust hPSC attachment, proliferation and maintenance of pluripotency across varying substrate stiffness (3 kPa-38 kPa). Using this hydrogel platform, we demonstrate that varying the types of biochemical cues (Matrigel, laminin, GAG-peptide) or density of Matrigel can alter stiffness-dependent YAP localization in hPSCs. In particular, we show that stiffness-dependent YAP localization is overridden at low or high density of Matrigel. Furthermore, human mesenchymal stem cells display stiffness-dependent YAP localization only at intermediate fibronectin density. The hydrogel platform with enhanced conjugation efficiency of biochemical cues provides a powerful tool for uncovering the role of biochemical cues in regulating mechanotransduction of various stem cell types.
Collapse
|
35
|
Xia S, Yim EKF, Kanchanawong P. Molecular Organization of Integrin-Based Adhesion Complexes in Mouse Embryonic Stem Cells. ACS Biomater Sci Eng 2019; 5:3828-3842. [PMID: 33438423 DOI: 10.1021/acsbiomaterials.8b01124] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mechanical microenvironment serves as an important factor influencing stem cell differentiation. Mechanobiological responses depend strongly on actomyosin contractility and integrin-based cell-extracellular matrix (ECM) interactions mediated by adhesive structures such as focal adhesions (FAs). While the roles of FAs in mechanobiology have been intensively studied in many mesenchymal and migratory cell types, recently it has been recognized that certain pluripotent stem cells (PSCs) exhibited significantly attenuated FA-mediated mechanobiological responses. FAs in such PSCs are sparsely distributed and much less prominent in comparison to "classical" FAs of typical adherent cells. Despite these differences, insights into how FAs in PSCs are structurally organized to perform their functions are still elusive. Using mouse embryonic stem cells (mESCs) to study PSC-ECM interactions, here we surveyed the molecular composition and nanostructural organization of FAs. We found that, despite being small in size, mESC FAs appeared to be compositionally mature, containing markers such as vinculin, zyxin, and α-actinin, and dependent on myosin II contractility. Using super-resolution microscopy, we revealed that mESC FAs were organized into a conserved multilayer nanoscale architecture. However, the nanodomain organization was compressed in mESCs, with the force transduction layer spanning ∼10 nm, significantly more compact than in FAs of other cell types. Furthermore, we found that the position and orientation of vinculin, a key mechanotransduction protein, were modulated in an ECM-dependent manner. Our analysis also revealed that while most core FA genes were expressed, the expression of LIM domain proteins was comparatively lower in PSCs. Altogether our results suggest that while core structural and mechanosensitive elements are operational in mESC FAs, their structural organization and regulatory aspects may diverge significantly from "classical" FAs, which may account for the attenuated mechanobiological responses of these cell types.
Collapse
Affiliation(s)
- Shumin Xia
- Mechanobiology Institute, Singapore, Republic of Singapore, 117411
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Pakorn Kanchanawong
- Mechanobiology Institute, Singapore, Republic of Singapore, 117411.,Department of Biomedical Engineering, National University of Singapore, Singapore, Republic of Singapore, 117411
| |
Collapse
|
36
|
Matte BF, Kumar A, Placone JK, Zanella VG, Martins MD, Engler AJ, Lamers ML. Matrix stiffness mechanically conditions EMT and migratory behavior of oral squamous cell carcinoma. J Cell Sci 2019; 132:jcs.224360. [PMID: 30559248 DOI: 10.1242/jcs.224360] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/09/2018] [Indexed: 12/29/2022] Open
Abstract
Tumors are composed of heterogeneous phenotypes, each having different sensitivities to the microenvironment. One microenvironment characteristic - matrix stiffness - helps to regulate malignant transformation and invasion in mammary tumors, but its influence on oral squamous cell carcinoma (OSCC) is unclear. We observed that, on stiff matrices, a highly invasive OSCC cell line (SCC25) comprising a low E-cad to N-cad ratio (InvH/E:NL; SCC25) had increased migration velocity and decreased adhesion strength compared to a less invasive OSCC cell line (Cal27) with high E-cad to N-cad ratio (InvL/E:NH; Cal27). However, InvL/E:NH cells acquire a mesenchymal signature and begin to migrate faster when exposed to prolonged time on a stiff niche, suggesting that cells can be mechanically conditioned. Owing to increased focal adhesion assembly, InvL/E:NH cells migrated faster, which could be reduced when increasing integrin affinity with high divalent cation concentrations. Mirroring these data in human patients, we observed that collagen organization, an indicator of matrix stiffness, was increased with advanced disease and correlated with early recurrence. Consistent with epithelial tumors, our data suggest that OSCC cells are mechanically sensitive and that their contribution to tumor progression is mediated in part by this sensitivity.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Bibiana F Matte
- Department of Oral Pathology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Department of Bioengineering, University of California, San Diego; La Jolla, CA 92093, USA
| | - Aditya Kumar
- Department of Bioengineering, University of California, San Diego; La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Jesse K Placone
- Department of Bioengineering, University of California, San Diego; La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Virgílio G Zanella
- Department of Oral Pathology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Head and Neck Surgery Department, Santa Rita Hospital, Santa Casa de Misericórdia de Porto Alegre, Porto Alegre, RS, Brazil
| | - Manoela D Martins
- Department of Oral Pathology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego; La Jolla, CA 92093, USA .,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Marcelo L Lamers
- Department of Oral Pathology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil .,Department of Morphological Sciences, Institute of Basic Health Sciences, Universidade Federal of Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
37
|
Abstract
It is increasingly clear that mechanotransduction pathways play important roles in regulating fundamental cellular functions. Of the basic mechanical functions, the determination of cellular morphology is critical. Cells typically use many mechanosensitive steps and different cell states to achieve a polarized shape through repeated testing of the microenvironment. Indeed, morphology is determined by the microenvironment through periodic activation of motility, mechanotesting, and mechanoresponse functions by hormones, internal clocks, and receptor tyrosine kinases. Patterned substrates and controlled environments with defined rigidities limit the range of cell behavior and influence cell state decisions and are thus very useful for studying these steps. The recently defined rigidity sensing process provides a good example of how cells repeatedly test their microenvironment and is also linked to cancer. In general, aberrant extracellular matrix mechanosensing is associated with numerous conditions, including cardiovascular disease, aging, and fibrosis, that correlate with changes in tissue morphology and matrix composition. Hence, detailed descriptions of the steps involved in sensing and responding to the microenvironment are needed to better understand both the mechanisms of tissue homeostasis and the pathomechanisms of human disease.
Collapse
Affiliation(s)
- Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel 31096;
| | - Bo Yang
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore;
| | - Michael P Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore; .,Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| |
Collapse
|
38
|
Rashad A, Mohamed-Ahmed S, Ojansivu M, Berstad K, Yassin MA, Kivijärvi T, Heggset EB, Syverud K, Mustafa K. Coating 3D Printed Polycaprolactone Scaffolds with Nanocellulose Promotes Growth and Differentiation of Mesenchymal Stem Cells. Biomacromolecules 2018; 19:4307-4319. [DOI: 10.1021/acs.biomac.8b01194] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Ahmad Rashad
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | | | - Miina Ojansivu
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
- Adult Stem Cell Research Group, Faculty of Medicine and Life Sciences and BioMediTech Institute, University of Tampere, Tampere, Finland
| | - Kaia Berstad
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Mohammed A. Yassin
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
- Department of Fiber and Polymer Technology, Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Tove Kivijärvi
- Department of Fiber and Polymer Technology, Royal Institute of Technology (KTH), Stockholm, Sweden
| | | | - Kristin Syverud
- RISE PFI, Trondheim, Norway
- Department of Chemical Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| |
Collapse
|
39
|
Jing G, Wang Z, Zhuang X, He X, Wu H, Wang Q, Cheng L, Liu Z, Wang S, Zhu R. Suspended graphene oxide nanosheets maintain the self-renewal of mouse embryonic stem cells via down-regulating the expression of Vinculin. Biomaterials 2018; 171:1-11. [PMID: 29677519 DOI: 10.1016/j.biomaterials.2018.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/05/2018] [Accepted: 04/10/2018] [Indexed: 12/17/2022]
Abstract
Graphene oxide (GO), with good hydrophilicity and biocompatibility, is widely explored as a carrier for various factors in the field of stem cell differentiation. However, its function of sustaining the stemness of mouse embryonic stem cells (mESCs) and the underlying mechanisms of this process remains undiscovered. Herein, we explored the biofunction of GO on mESCs and revealed the involved signaling pathways and key gene. The alkaline phosphatase activity detection, pluripotency genes quantification and the teratomas formation in vivo confirmed that GO nanosheets could sustain the self-renewal ability of mESCs instead of influencing its pluripotency. The underlying signaling pathways were uncovered by RNA-seq that integrin signaling pathway was involved in the biofunction of GO on mESCs and Vinculin turned to be a key gene for the effect of GO. Further experiments confirmed that the downregulation of Vinculin influenced the fate of mESCs through decreasing the expression of MEK1. Altogether, the study demonstrated for the first time that GOs hold the potential in sustaining the self-renewal of mESCs and clarified the mechanism of this function, which make it play a new role in stem cell research and regenerative medicine.
Collapse
Affiliation(s)
- Guoxin Jing
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, PR China
| | - Zhaojie Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, PR China
| | - Xizhen Zhuang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, PR China
| | - Xiaolie He
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, PR China
| | - Huijun Wu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, PR China
| | - Qingxiu Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, PR China
| | | | - Zhongmin Liu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, PR China
| | - Shilong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, PR China.
| | - Rongrong Zhu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, PR China.
| |
Collapse
|
40
|
Sphingosine 1-Phosphate Receptor 1 Is Required for MMP-2 Function in Bone Marrow Mesenchymal Stromal Cells: Implications for Cytoskeleton Assembly and Proliferation. Stem Cells Int 2018; 2018:5034679. [PMID: 29713350 PMCID: PMC5866864 DOI: 10.1155/2018/5034679] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/17/2017] [Accepted: 11/27/2017] [Indexed: 12/31/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cell- (BM-MSC-) based therapy is a promising option for regenerative medicine. An important role in the control of the processes influencing the BM-MSC therapeutic efficacy, namely, extracellular matrix remodelling and proliferation and secretion ability, is played by matrix metalloproteinase- (MMP-) 2. Therefore, the identification of paracrine/autocrine regulators of MMP-2 function may be of great relevance for improving BM-MSC therapeutic potential. We recently reported that BM-MSCs release the bioactive lipid sphingosine 1-phosphate (S1P) and, here, we demonstrated an impairment of MMP-2 expression/release when the S1P receptor subtype S1PR1 is blocked. Notably, active S1PR1/MMP-2 signalling is required for F-actin structure assembly (lamellipodia, microspikes, and stress fibers) and, in turn, cell proliferation. Moreover, in experimental conditions resembling the damaged/regenerating tissue microenvironment (hypoxia), S1P/S1PR1 system is also required for HIF-1α expression and vinculin reduction. Our findings demonstrate for the first time the trophic role of S1P/S1PR1 signalling in maintaining BM-MSCs' ability to modulate MMP-2 function, necessary for cytoskeleton reorganization and cell proliferation in both normoxia and hypoxia. Altogether, these data provide new perspectives for considering S1P/S1PR1 signalling a pharmacological target to preserve BM-MSC properties and to potentiate their beneficial potential in tissue repair.
Collapse
|
41
|
Holle AW, Young JL, Van Vliet KJ, Kamm RD, Discher D, Janmey P, Spatz JP, Saif T. Cell-Extracellular Matrix Mechanobiology: Forceful Tools and Emerging Needs for Basic and Translational Research. NANO LETTERS 2018; 18:1-8. [PMID: 29178811 PMCID: PMC5842374 DOI: 10.1021/acs.nanolett.7b04982] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Extracellular biophysical cues have a profound influence on a wide range of cell behaviors, including growth, motility, differentiation, apoptosis, gene expression, adhesion, and signal transduction. Cells not only respond to definitively mechanical cues from the extracellular matrix (ECM) but can also sometimes alter the mechanical properties of the matrix and hence influence subsequent matrix-based cues in both physiological and pathological processes. Interactions between cells and materials in vitro can modify cell phenotype and ECM structure, whether intentionally or inadvertently. Interactions between cell and matrix mechanics in vivo are of particular importance in a wide variety of disorders, including cancer, central nervous system injury, fibrotic diseases, and myocardial infarction. Both the in vitro and in vivo effects of this coupling between mechanics and biology hold important implications for clinical applications.
Collapse
Affiliation(s)
- Andrew W Holle
- Department of Cellular Biophysics, Max Planck Institute for Medical Research , Jahnstraße 29, 69120 Heidelberg, Germany
- Institute of Physical Chemistry, University of Heidelberg , 69117 Heidelberg, Germany
| | - Jennifer L Young
- Department of Cellular Biophysics, Max Planck Institute for Medical Research , Jahnstraße 29, 69120 Heidelberg, Germany
- Institute of Physical Chemistry, University of Heidelberg , 69117 Heidelberg, Germany
| | - Krystyn J Van Vliet
- BioSystems & Micromechanics IRG, Singapore-MIT Alliance in Research and Technology , Singapore
| | - Roger D Kamm
- BioSystems & Micromechanics IRG, Singapore-MIT Alliance in Research and Technology , Singapore
| | | | | | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research , Jahnstraße 29, 69120 Heidelberg, Germany
- Institute of Physical Chemistry, University of Heidelberg , 69117 Heidelberg, Germany
| | - Taher Saif
- Department of Mechanical Sciences and Engineering, University of Illinois at Urbana-Champaign , 1206 West Green Street, Urbana, Illinois 61801, United States
| |
Collapse
|
42
|
Photoresponsive Hydrogels with Photoswitchable Stiffness: Emerging Platforms to Study Temporal Aspects of Mesenchymal Stem Cell Responses to Extracellular Stiffness Regulation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1144:53-69. [PMID: 30456642 DOI: 10.1007/5584_2018_293] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An extensive number of cell-matrix interaction studies have identified matrix stiffness as a potent regulator of cellular properties and behaviours. Perhaps most notably, matrix stiffness has been demonstrated to regulate mesenchymal stem cell (MSC) phenotype and lineage commitment. Given the therapeutic potential for MSCs in regenerative medicine, significant efforts have been made to understand the molecular mechanisms involved in stiffness regulation. These efforts have predominantly focused on using stiffness-defined polyacrylamide (PA) hydrogels to culture cells in 2D and have enabled elucidation of a number of mechano-sensitive signalling pathways. However, despite proving to be a valuable tool, these stiffness-defined hydrogels do not reflect the dynamic nature of living tissues, which are subject to continuous remodelling during processes such as development, ageing, disease and regeneration. Therefore, in order to study temporal aspects of stiffness regulation, researchers have developed and exploited novel hydrogel substrates with in situ tuneable stiffness. In particular, photoresponsive hydrogels with photoswitchable stiffness are emerging as exciting platforms to study MSC stiffness regulation. This chapter provides an introduction to the use of PA hydrogel substrates, the molecular mechanisms of mechanotransduction currently under investigation and the development of these emerging photoresponsive hydrogel platforms.
Collapse
|
43
|
Kumar A, Placone JK, Engler AJ. Understanding the extracellular forces that determine cell fate and maintenance. Development 2017; 144:4261-4270. [PMID: 29183939 DOI: 10.1242/dev.158469] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cells interpret signals from their microenvironment while simultaneously modifying the niche through secreting factors and exerting mechanical forces. Many soluble stem cell cues have been determined over the past century, but in the past decade, our molecular understanding of mechanobiology has advanced to explain how passive and active forces induce similar signaling cascades that drive self-renewal, migration, differentiation or a combination of these outcomes. Improvements in stem cell culture methods, materials and biophysical tools that assess function have improved our understanding of these cascades. Here, we summarize these advances and offer perspective on ongoing challenges.
Collapse
Affiliation(s)
- Aditya Kumar
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Jesse K Placone
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA .,Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| |
Collapse
|
44
|
Kang P, Kumar S, Schaffer D. Novel biomaterials to study neural stem cell mechanobiology and improve cell-replacement therapies. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017; 4:13-20. [PMID: 29399646 PMCID: PMC5791915 DOI: 10.1016/j.cobme.2017.09.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neural stem cells (NSCs) are a valuable cell source for tissue engineering, regenerative medicine, disease modeling, and drug screening applications. Analogous to other stem cells, NSCs are tightly regulated by their microenvironmental niche, and prior work utilizing NSCs as a model system with engineered biomaterials has offered valuable insights into how biophysical inputs can regulate stem cell proliferation, differentiation, and maturation. In this review, we highlight recent exciting studies with innovative material platforms that enable narrow stiffness gradients, mechanical stretching, temporal stiffness switching, and three-dimensional culture to study NSCs. These studies have significantly advanced our knowledge of how stem cells respond to an array of different biophysical inputs and the underlying mechanosensitive mechanisms. In addition, we discuss efforts to utilize engineered material scaffolds to improve NSC-based translational efforts and the importance of mechanobiology in tissue engineering applications.
Collapse
Affiliation(s)
- Phillip Kang
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Lawrence Berkeley National Laboratory Physical Biosciences Division, 1 Cyclotron Rd, Berkeley, CA 94720, USA
| | - David Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
45
|
Jansen K, Atherton P, Ballestrem C. Mechanotransduction at the cell-matrix interface. Semin Cell Dev Biol 2017; 71:75-83. [DOI: 10.1016/j.semcdb.2017.07.027] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 01/09/2023]
|
46
|
Nagayama K, Inoue T, Hamada Y, Matsumoto T. A novel patterned magnetic micropillar array substrate for analysis of cellular mechanical responses. J Biomech 2017; 65:194-202. [PMID: 29126605 DOI: 10.1016/j.jbiomech.2017.10.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 10/16/2017] [Accepted: 10/17/2017] [Indexed: 02/02/2023]
Abstract
Traction forces generated at cellular focal adhesions (FAs) play an essential role in regulating various cellular functions. These forces (1-100 nN) can be measured by observing the local displacement of a flexible substrate upon which cells have been plated. Approaches employing this method include using microfabricated arrays of poly(dimethylsiloxane) (PDMS) micropillars that bend by cellular traction forces. A tool capable of applying a force to FAs independently, by actively moving the micropillars, should become a powerful tool to delineate the cellular mechanotransduction mechanisms. Here, we developed a patterned magnetic micropillar array PDMS substrate that can be used for the mechanical stimulation of cellular FAs and the measurement of associated traction forces. The diameter, length, and center-to-center spacing of the micropillars were 3, 9, and 9 µm, respectively. Iron particles were embedded into the micropillars, enabling the pillars to bend in response to an external magnetic field, which also controlled their location on the substrate. Applying a magnetic field of 0.3 T bent the pillars by ∼4 µm and allowed transfer of external forces to the actin cytoskeleton through FAs formed on the pillar top. Using this approach, we investigated the traction force changes in cultured aortic smooth muscle cells (SMCs) after local compressive stimuli to release cell pretension. The mechanical responses of SMCs were roughly classified into two types: almost a half of the cells showed a little decrease of traction force at each pillar following compressive stimulation, although cell area increased significantly; and the rest showed the opposite, with increased forces and a simultaneous decrease in area. The traction forces of SMCs fluctuated markedly during the local compression. The root mean square of traction forces significantly increased during the compression, and returned to the baseline level after its release. These results suggest that the fluctuation of forces may be caused by active reorganization of the actin cytoskeleton and/or its dynamic interaction with myosin molecules. Thus, our magnetic micropillar substrate would be useful in investigating the mechanotransduction mechanisms of cells.
Collapse
Affiliation(s)
- Kazuaki Nagayama
- Micro-Nano Biomechanics Laboratory, Department of Intelligent Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi 316-8511, Japan.
| | - Takuya Inoue
- Biomechanics Laboratory, Department of Mechanical Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Yasuhiro Hamada
- Biomechanics Laboratory, Department of Mechanical Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan
| | - Takeo Matsumoto
- Biomechanics Laboratory, Department of Mechanical Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan; Biomechanics Laboratory, Department of Mechanical Science and Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan.
| |
Collapse
|
47
|
Ichikawa T, Kita M, Matsui TS, Nagasato AI, Araki T, Chiang SH, Sezaki T, Kimura Y, Ueda K, Deguchi S, Saltiel AR, Kioka N. Vinexin family (SORBS) proteins play different roles in stiffness-sensing and contractile force generation. J Cell Sci 2017; 130:3517-3531. [PMID: 28864765 DOI: 10.1242/jcs.200691] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 08/10/2017] [Indexed: 12/17/2022] Open
Abstract
Vinexin, c-Cbl associated protein (CAP) and Arg-binding protein 2 (ArgBP2) constitute an adaptor protein family called the vinexin (SORBS) family that is targeted to focal adhesions (FAs). Although numerous studies have focused on each of the SORBS proteins and partially elucidated their involvement in mechanotransduction, a comparative analysis of their function has not been well addressed. Here, we established mouse embryonic fibroblasts that individually expressed SORBS proteins and analysed their functions in an identical cell context. Both vinexin-α and CAP co-localized with vinculin at FAs and promoted the appearance of vinculin-rich FAs, whereas ArgBP2 co-localized with α-actinin at the proximal end of FAs and punctate structures on actin stress fibers (SFs), and induced paxillin-rich FAs. Furthermore, both vinexin-α and CAP contributed to extracellular matrix stiffness-dependent vinculin behaviors, while ArgBP2 stabilized α-actinin on SFs and enhanced intracellular contractile forces. These results demonstrate the differential roles of SORBS proteins in mechanotransduction.
Collapse
Affiliation(s)
- Takafumi Ichikawa
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto 606-8507, Japan
| | - Masahiro Kita
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Tsubasa S Matsui
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Showa, Nagoya 466-8555, Japan.,Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Ayaka Ichikawa Nagasato
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Tomohiko Araki
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Showa, Nagoya 466-8555, Japan
| | - Shian-Huey Chiang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Takuhito Sezaki
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Yasuhisa Kimura
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Kazumitsu Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto 606-8507, Japan
| | - Shinji Deguchi
- Department of Nanopharmaceutical Sciences, Nagoya Institute of Technology, Showa, Nagoya 466-8555, Japan.,Division of Bioengineering, Graduate School of Engineering Science, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Alan R Saltiel
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Noriyuki Kioka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Sakyo, Kyoto 606-8502, Japan .,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Sakyo, Kyoto 606-8507, Japan
| |
Collapse
|
48
|
Garakani K, Shams H, Mofrad MRK. Mechanosensitive Conformation of Vinculin Regulates Its Binding to MAPK1. Biophys J 2017; 112:1885-1893. [PMID: 28494959 DOI: 10.1016/j.bpj.2017.03.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 03/14/2017] [Accepted: 03/22/2017] [Indexed: 11/17/2022] Open
Abstract
Extracellular matrix stiffness sensing by living cells is known to play a major role in a variety of cell mechanobiological processes, such as migration and differentiation. Various membrane and cytoplasmic proteins are involved in transmitting and transducing environmental signals to biochemical cascades. Protein kinases play a key role in regulating the activity of focal adhesion proteins. Recently, an interaction between mitogen-activated protein kinase (MAPK1) and vinculin was experimentally shown to mediate this process. Here, we adopt a molecular modeling approach to further investigate this interaction and its possible regulatory effects. Using a combination of data-driven flexible docking and molecular dynamics simulations guided by previous experimental studies, we predict the structure of the MAPK1-vinculin complex. Furthermore, by comparing the association of MAPK1 with open versus closed vinculin, we demonstrate that MAPK1 exhibits preferential binding toward the open conformation of vinculin, suggesting that the MAPK1-vinculin interaction is conformationally selective. Finally, we demonstrate that changes in the size of the D3-D4 cleft provide a structural basis for the conformational selectivity of the interaction.
Collapse
Affiliation(s)
- Kiavash Garakani
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California
| | - Hengameh Shams
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, California; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, California.
| |
Collapse
|
49
|
Nanoscale mechanobiology of cell adhesions. Semin Cell Dev Biol 2017; 71:53-67. [PMID: 28754443 DOI: 10.1016/j.semcdb.2017.07.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 12/16/2022]
Abstract
Proper physiological functions of cells and tissues depend upon their abilities to sense, transduce, integrate, and generate mechanical and biochemical signals. Although such mechanobiological phenomena are widely observed, the molecular mechanisms driving these outcomes are still not fully understood. Cell adhesions formed by integrins and cadherins receptors are key structures that process diverse sources of signals to elicit complex mechanobiological responses. Since the nanoscale is the length scale at which molecules interact to relay force and information, the understanding of cell adhesions at the nanoscale level is important for grasping the inner logics of cellular decision making. Until recently, the study of the biological nanoscale has been restricted by available molecular and imaging tools. Fortunately, rapid technological advances have increasingly opened up the nanoscale realm to systematic investigations. In this review, we discuss current insights and key open questions regarding the nanoscale structure and function relationship of cell adhesions, focusing on recent progresses in characterizing their composition, spatial organization, and cytomechanical operation.
Collapse
|
50
|
Atherton P, Lausecker F, Harrison A, Ballestrem C. Low-intensity pulsed ultrasound promotes cell motility through vinculin-controlled Rac1 GTPase activity. J Cell Sci 2017; 130:2277-2291. [PMID: 28576970 DOI: 10.1242/jcs.192781] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 05/29/2017] [Indexed: 12/16/2022] Open
Abstract
Low-intensity pulsed ultrasound (LIPUS) is a therapy used clinically to promote healing. Using live-cell imaging we show that LIPUS stimulation, acting through integrin-mediated cell-matrix adhesions, rapidly induces Rac1 activation associated with dramatic actin cytoskeleton rearrangements. Our study demonstrates that the mechanosensitive focal adhesion (FA) protein vinculin, and both focal adhesion kinase (FAK, also known as PTK2) and Rab5 (both the Rab5a and Rab5b isoforms) have key roles in regulating these effects. Inhibiting the link of vinculin to the actin-cytoskeleton abolished LIPUS sensing. We show that this vinculin-mediated link was not only critical for Rac1 induction and actin rearrangements, but was also important for the induction of a Rab5-dependent increase in the number of early endosomes. Expression of dominant-negative Rab5, or inhibition of endocytosis with dynasore, also blocked LIPUS-induced Rac1 signalling events. Taken together, our data show that LIPUS is sensed by cell matrix adhesions through vinculin, which in turn modulates a Rab5-Rac1 pathway to control ultrasound-mediated endocytosis and cell motility. Finally, we demonstrate that a similar FAK-Rab5-Rac1 pathway acts to control cell spreading upon fibronectin.
Collapse
Affiliation(s)
- Paul Atherton
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, USA
| | - Franziska Lausecker
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, USA
| | - Andrew Harrison
- Bioventus Cooperatief, Taurusavenue 31, 2132 LS Hoofddorp, The Netherlands
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, USA
| |
Collapse
|