201
|
Zhu X, Wang Z, Teng F. A review of regulated self-organizing approaches for tissue regeneration. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 167:63-78. [PMID: 34293337 DOI: 10.1016/j.pbiomolbio.2021.07.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/06/2021] [Accepted: 07/15/2021] [Indexed: 12/13/2022]
Abstract
Tissue and organ regeneration is the dynamic process by which a population of cells rearranges into a specific form with specific functions. Traditional tissue regeneration utilizes tissue grafting, cell implantation, and structured scaffolds to achieve clinical efficacy. However, tissue grafting methods face a shortage of donor tissue, while cell implantation may involve leakage of the implanted cells without a supportive 3D matrix. Cell migration, proliferation, and differentiation in structured scaffolds may disorganize and frustrate the artificially pre-designed structures, and sometimes involve immunogenic reactions. To overcome this limitation, the self-organizing properties and innate regenerative capability of tissue/organism formation in the absence of guidance by structured scaffolds has been investigated. This review emphasizes the growing subfield of the regulated self-organizing approach for neotissue formation and describes advances in the subfield using diverse, cutting-edge, inter-disciplinarity technologies. We cohesively summarize the directed self-organization of cells in the micro-engineered cell-ECM system and 3D/4D cell printing. Mathematical modeling of cellular self-organization is also discussed for providing rational guidance to intractable problems in tissue regeneration. It is envisioned that future self-organization approaches integrating biomathematics, micro-nano engineering, and gene circuits developed from synthetic biology will continue to work in concert with self-organizing morphogenesis to enhance rational control during self-organizing in tissue and organ regeneration.
Collapse
Affiliation(s)
- Xiaolu Zhu
- College of Mechanical & Electrical Engineering, Hohai University, Changzhou, Jiangsu, 213022, China; Changzhou Key Laboratory of Digital Manufacture Technology, Hohai University, Changzhou, Jiangsu, 213022, China; Jiangsu Key Laboratory of Special Robot Technology, Hohai University, Changzhou, Jiangsu, 213022, China.
| | - Zheng Wang
- College of Mechanical & Electrical Engineering, Hohai University, Changzhou, Jiangsu, 213022, China
| | - Fang Teng
- Department of Gynaecology and Obstetrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu, 210004, China.
| |
Collapse
|
202
|
Zou R, Wu S, Wang Y, Kang X, Zhao S, Shi H, Zheng D, Gao B, Ma S, Niu L, Gao Y. Role of integrin‑linked kinase in static compressive stress‑induced autophagy via phosphatidylinositol 3 kinase in human periodontal ligament cells. Int J Mol Med 2021; 48:167. [PMID: 34278436 PMCID: PMC8285053 DOI: 10.3892/ijmm.2021.5000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Orthodontic tooth movement (OTM) is achieved by using mechanical stimuli, which lead to the remodeling of periodontal tissues. Previous findings have demonstrated that autophagy may be one of the cell responses to mechanical stress. As a key structure in the integrin pathway, integrin linked‑kinase (ILK) may play a role in the transmission of these mechanical signals. In addition, ILK is an important upstream molecule that regulates autophagy, under the influence of phosphatidylinositol 3 kinase (PI3K). Therefore, exploring the effect of mechanical stress on autophagy and the associated role of ILK/PI3K is of utmost significance to understanding the mechanism behind OTM. In the present study, human periodontal ligament cells (hPDLCs) were embedded into a collagen‑alginate complex hydrogel for three‑dimensional (3D) culturing. Static compressive stress (2.5 g/cm2) was loaded using the uniform weight method for 5, 15, 30, and 60 min. The autophagy of hPDLCs was detected by the expression of Beclin‑1 (BECN1) and ATG‑5 using RT‑qPCR and LC3, respectively, using immunofluorescence. The results showed that the level of autophagy and gene expression of ILK increased significantly under static compressive stress. In ILK‑silenced cells, static compressive stress could also upregulate ILK expression and increase the levels of autophagy. After PI3K inhibition, the increase in the autophagy level and the upregulation of ILK expression disappeared. These findings suggest that static compressive stress can induce autophagy in hPDLCs in a rapid, transient process, regulated by ILK and PI3K. Moreover, this static stress can upregulate ILK expression in a PI3K‑dependent manner.
Collapse
Affiliation(s)
- Rui Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shiyang Wu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yijie Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xueping Kang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shuyang Zhao
- College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Haoyu Shi
- College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Danqing Zheng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Bei Gao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Shuyu Ma
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Lin Niu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yunan Gao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
203
|
Kärki T, Tojkander S. TRPV Protein Family-From Mechanosensing to Cancer Invasion. Biomolecules 2021; 11:1019. [PMID: 34356643 PMCID: PMC8301805 DOI: 10.3390/biom11071019] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023] Open
Abstract
Biophysical cues from the cellular microenvironment are detected by mechanosensitive machineries that translate physical signals into biochemical signaling cascades. At the crossroads of extracellular space and cell interior are located several ion channel families, including TRP family proteins, that are triggered by mechanical stimuli and drive intracellular signaling pathways through spatio-temporally controlled Ca2+-influx. Mechanosensitive Ca2+-channels, therefore, act as critical components in the rapid transmission of physical signals into biologically compatible information to impact crucial processes during development, morphogenesis and regeneration. Given the mechanosensitive nature of many of the TRP family channels, they must also respond to the biophysical changes along the development of several pathophysiological conditions and have also been linked to cancer progression. In this review, we will focus on the TRPV, vanilloid family of TRP proteins, and their connection to cancer progression through their mechanosensitive nature.
Collapse
Affiliation(s)
- Tytti Kärki
- Department of Applied Physics, School of Science, Aalto University, 00076 Espoo, Finland;
| | - Sari Tojkander
- Department of Veterinary Biosciences, Section of Pathology, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
204
|
Torrino S, Grasset EM, Audebert S, Belhadj I, Lacoux C, Haynes M, Pisano S, Abélanet S, Brau F, Chan SY, Mari B, Oldham WM, Ewald AJ, Bertero T. Mechano-induced cell metabolism promotes microtubule glutamylation to force metastasis. Cell Metab 2021; 33:1342-1357.e10. [PMID: 34102109 DOI: 10.1016/j.cmet.2021.05.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/31/2021] [Accepted: 05/07/2021] [Indexed: 01/11/2023]
Abstract
Mechanical signals from the tumor microenvironment modulate cell mechanics and influence cell metabolism to promote cancer aggressiveness. Cells withstand external forces by adjusting the stiffness of their cytoskeleton. Microtubules (MTs) act as compression-bearing elements. Yet how cancer cells regulate MT dynamic in response to the locally constrained environment has remained unclear. Using breast cancer as a model of a disease in which mechanical signaling promotes disease progression, we show that matrix stiffening rewires glutamine metabolism to promote MT glutamylation and force MT stabilization, thereby promoting cell invasion. Pharmacologic inhibition of glutamine metabolism decreased MT glutamylation and affected their mechanical stabilization. Similarly, decreased MT glutamylation by overexpressing tubulin mutants lacking glutamylation site(s) decreased MT stability, thereby hampering cancer aggressiveness in vitro and in vivo. Together, our results decipher part of the enigmatic tubulin code that coordinates the fine-tunable properties of MT and link cell metabolism to MT dynamics and cancer aggressiveness.
Collapse
Affiliation(s)
| | - Eloise M Grasset
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephane Audebert
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Ilyes Belhadj
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | | | - Meagan Haynes
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sabrina Pisano
- Université Côte d'Azur, CNRS, INSERM, IRCAN, Nice, France
| | | | - Frederic Brau
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bernard Mari
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | - William M Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew J Ewald
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
205
|
Inman A, Smutny M. Feeling the force: Multiscale force sensing and transduction at the cell-cell interface. Semin Cell Dev Biol 2021; 120:53-65. [PMID: 34238674 DOI: 10.1016/j.semcdb.2021.06.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022]
Abstract
A universal principle of all living cells is the ability to sense and respond to mechanical stimuli which is essential for many biological processes. Recent efforts have identified critical mechanosensitive molecules and response pathways involved in mechanotransduction during development and tissue homeostasis. Tissue-wide force transmission and local force sensing need to be spatiotemporally coordinated to precisely regulate essential processes during development such as tissue morphogenesis, patterning, cell migration and organogenesis. Understanding how cells identify and interpret extrinsic forces and integrate a specific response on cell and tissue level remains a major challenge. In this review we consider important cellular and physical factors in control of cell-cell mechanotransduction and discuss their significance for cell and developmental processes. We further highlight mechanosensitive macromolecules that are known to respond to external forces and present examples of how force responses can be integrated into cell and developmental programs.
Collapse
Affiliation(s)
- Angus Inman
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV47AL, UK
| | - Michael Smutny
- Centre for Mechanochemical Cell Biology and Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV47AL, UK.
| |
Collapse
|
206
|
Tsai MC, Daniels JT. The impact of biomechanics on corneal endothelium tissue engineering. Exp Eye Res 2021; 209:108690. [PMID: 34216616 DOI: 10.1016/j.exer.2021.108690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/03/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022]
Abstract
The integrity of innermost layer of the cornea, the corneal endothelium, is key to sustaining corneal transparency. Therefore, disease or injury causing loss or damage to the corneal endothelial cell population may threaten vision. Transplantation of corneal tissue is the standard treatment used to replace malfunctioning corneal endothelial cells. However, this surgery is dependent upon donor tissue, which is limited in supply. Hence, tissue engineers have attempted to construct alternative transplantable tissues or cell therapies to alleviate this problem. Nevertheless, the intrinsic non-dividing nature of corneal endothelial cells continues to foil scientists in their attempts to yield large numbers of cells in the laboratory for use in such novel therapies. Interestingly, the contribution of the biomechanical properties of the underlying extracellular matrix (ECM) on cell division, tissue development and maintenance has been extensively investigated in other many cell types. However, the impact of biomechanics on corneal endothelial cell behaviour is relatively unexplored. Here, we describe contemporary tissue engineering solutions aimed at circumventing donor tissue scarcity. We review the ECM structure and biomechanical features of corneal endothelial cells. We discuss the alterations of ECM in endothelial disease development and progression and point out the role of ECM in developing a tissue-engineered corneal endothelium. We highlight the main biomechanical cues, including topographical and mechanical features, that impact cellular behaviors. Finally, we discuss the influence of biomechanical cues on cell and tissue development, and how corneal endothelial cells response to individual biomechanical stimuli in tissue engineering, which have implications for designing an engineered endothelium and maintaining cell function.
Collapse
Affiliation(s)
- Meng-Chen Tsai
- UCL Institute of Ophthalmology, University College London, London, UK.
| | - Julie T Daniels
- UCL Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
207
|
Emon MAB, Knoll S, Doha U, Ladehoff L, Lalonde L, Baietto D, Sivaguru M, Bhargava R, Saif MTA. Dose- independent threshold illumination for non-invasive time-lapse fluorescence imaging of live cells. EXTREME MECHANICS LETTERS 2021; 46:101249. [PMID: 34095408 PMCID: PMC8171180 DOI: 10.1016/j.eml.2021.101249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Fluorescent microscopy employs monochromatic light for excitation, which can adversely affect the cells being observed. We reported earlier that fibroblasts relax their contractile force in response to green light of typical intensity. Here we show that such effects are independent of extracellular matrix and cell lines. In addition, we establish a threshold intensity that elicits minimal or no adverse effect on cell contractility even for long-time exposure. This threshold intensity is wavelength dependent. We cultured fibroblasts on soft 2D elastic hydrogels embedded with fluorescent beads to trace substrate deformation and cell forces. The beads move towards cell center when cells contract, but they move away when cells relax. We use relaxation/contraction ratio (λ r), in addition to traction force, as measures of cell response to red (wavelength, λ=635-650 nm), green (λ=545-580 nm) and blue (λ=455-490 nm) lights with varying intensities. Our results suggest that intensities below 57, 31 and 3.5 W/m2 for red, green and blue lights, respectively, do not perturb force homeostasis. To our knowledge, these intensities are the lowest reported safe thresholds, implying that cell traction is a highly sensitive readout of the effect of light on cells. Most importantly, we find these threshold intensities to be dose-independent; i.e., safe regardless of the energy dosage or time of exposure. Conversely, higher intensities result in widespread force-relaxation in cells with λ r > 1. Furthermore, we present a photo-reaction based model that simulates photo-toxicity and predicts threshold intensity for different wavelengths within the visible spectra. In conclusion, we recommend employing illumination intensities below aforementioned wavelength-specific thresholds for time-lapse imaging of cells and tissues in order to avoid light-induced artifacts in experimental observations.
Collapse
Affiliation(s)
- M A Bashar Emon
- Dept. of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign
| | - Samantha Knoll
- Dept. of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign
| | - Umnia Doha
- Dept. of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign
| | - Lauren Ladehoff
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign
| | - Luke Lalonde
- Dept. of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign
| | - Danielle Baietto
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign
| | - Mayandi Sivaguru
- Carle Woese Institute of Genomic Biology, University of Illinois at Urbana-Champaign
| | - Rohit Bhargava
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign
| | - M Taher A Saif
- Dept. of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign
- Dept. of Bioengineering, University of Illinois at Urbana-Champaign
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign
- Corresponding author: M Taher A Saif, Gutgsell Professor, Associate Head for Graduate Programs and Research, Mechanical Science and Engineering, Research Professor, Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, 2101D Mechanical Engineering Laboratory, 105 S. Mathews Avenue, Urbana, IL 61801, USA, , Tel: 217-333-8552, Fax: 217-244-6534, http://saif.mechse.illinois.edu/
| |
Collapse
|
208
|
Yu LY, Tseng TJ, Lin HC, Hsu CL, Lu TX, Tsai CJ, Lin YC, Chu I, Peng CT, Chen HJ, Tsai FC. Synthetic dysmobility screen unveils an integrated STK40-YAP-MAPK system driving cell migration. SCIENCE ADVANCES 2021; 7:eabg2106. [PMID: 34321207 PMCID: PMC8318371 DOI: 10.1126/sciadv.abg2106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/10/2021] [Indexed: 05/05/2023]
Abstract
Integrating signals is essential for cell survival, leading to the concept of synthetic lethality. However, how signaling is integrated to control cell migration remains unclear. By conducting a "two-hit" screen, we revealed the synergistic reduction of cell migration when serine-threonine kinase 40 (STK40) and mitogen-activated protein kinase (MAPK) were simultaneously suppressed. Single-cell analyses showed that STK40 knockdown reduced cell motility and coordination by strengthening focal adhesion (FA) complexes. Furthermore, STK40 knockdown reduced the stability of yes-associated protein (YAP) and subsequently decreased YAP transported into the nucleus, while MAPK inhibition further weakened YAP activities in the nucleus to disturb FA remodeling. Together, we unveiled an integrated STK40-YAP-MAPK system regulating cell migration and introduced "synthetic dysmobility" as a novel strategy to collaboratively control cell migration.
Collapse
Affiliation(s)
- Ling-Yea Yu
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ting-Jen Tseng
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsuan-Chao Lin
- Department of Immunology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Lin Hsu
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ting-Xuan Lu
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
- Ph.D. Program in Biological Sciences, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Chia-Jung Tsai
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Yu-Chiao Lin
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - I Chu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chien-Tzu Peng
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hou-Jen Chen
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Feng-Chiao Tsai
- Department of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
209
|
Pocaterra A, Scattolin G, Romani P, Ament C, Ribback S, Chen X, Evert M, Calvisi DF, Dupont S. Fascin1 empowers YAP mechanotransduction and promotes cholangiocarcinoma development. Commun Biol 2021; 4:763. [PMID: 34155338 PMCID: PMC8217270 DOI: 10.1038/s42003-021-02286-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 06/01/2021] [Indexed: 12/14/2022] Open
Abstract
Mechanical forces control cell behavior, including cancer progression. Cells sense forces through actomyosin to activate YAP. However, the regulators of F-actin dynamics playing relevant roles during mechanostransduction in vitro and in vivo remain poorly characterized. Here we identify the Fascin1 F-actin bundling protein as a factor that sustains YAP activation in response to ECM mechanical cues. This is conserved in the mouse liver, where Fascin1 regulates YAP-dependent phenotypes, and in human cholangiocarcinoma cell lines. Moreover, this is relevant for liver tumorigenesis, because Fascin1 is required in the AKT/NICD cholangiocarcinogenesis model and it is sufficient, together with AKT, to induce cholangiocellular lesions in mice, recapitulating genetic YAP requirements. In support of these findings, Fascin1 expression in human intrahepatic cholangiocarcinomas strongly correlates with poor patient prognosis. We propose that Fascin1 represents a pro-oncogenic mechanism that can be exploited during intrahepatic cholangiocarcinoma development to overcome a mechanical tumor-suppressive environment.
Collapse
Affiliation(s)
- Arianna Pocaterra
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy
| | - Gloria Scattolin
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy
| | - Patrizia Romani
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy
| | - Cindy Ament
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California, USA
| | - Matthias Evert
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Diego F Calvisi
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy.
| |
Collapse
|
210
|
Miroshnikova YA, Manet S, Li X, Wickström SA, Faurobert E, Albiges-Rizo C. Calcium signaling mediates a biphasic mechanoadaptive response of endothelial cells to cyclic mechanical stretch. Mol Biol Cell 2021; 32:1724-1736. [PMID: 34081532 PMCID: PMC8684738 DOI: 10.1091/mbc.e21-03-0106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The vascular system is precisely regulated to adjust blood flow to organismal demand, thereby guaranteeing adequate perfusion under varying physiological conditions. Mechanical forces, such as cyclic circumferential stretch, are among the critical stimuli that dynamically adjust vessel distribution and diameter, but the precise mechanisms of adaptation to changing forces are unclear. We find that endothelial monolayers respond to cyclic stretch by transient remodeling of the vascular endothelial cadherin–based adherens junctions and the associated actomyosin cytoskeleton. Time-resolved proteomic profiling reveals that this remodeling is driven by calcium influx through the mechanosensitive Piezo1 channel, triggering Rho activation to increase actomyosin contraction. As the mechanical stimulus persists, calcium signaling is attenuated through transient down-regulation of Piezo1 protein. At the same time, filamins are phosphorylated to increase monolayer stiffness, allowing mechanoadaptation to restore junctional integrity despite continuing exposure to stretch. Collectively, this study identifies a biphasic response to cyclic stretch, consisting of an initial calcium-driven junctional mechanoresponse, followed by mechanoadaptation facilitated by monolayer stiffening.
Collapse
Affiliation(s)
- Yekaterina A Miroshnikova
- Université Grenoble Alpes, Institute for Advanced Biosciences, Grenoble 38042, France.,INSERM U1209, Institute for Advanced Biosciences, F-38700 La Tronche, France.,CNRS UMR 5039, Institute for Advanced Biosciences, F-38700 La Tronche, France.,Max Planck Institute for Biology of Ageing, D-50931 Cologne, Germany.,Helsinki Institute of Life Science, University of Helsinki, FI-00014 Helsinki, Finland.,Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland.,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FI-00014 Helsinki, Finland
| | - Sandra Manet
- Université Grenoble Alpes, Institute for Advanced Biosciences, Grenoble 38042, France.,INSERM U1209, Institute for Advanced Biosciences, F-38700 La Tronche, France.,CNRS UMR 5039, Institute for Advanced Biosciences, F-38700 La Tronche, France
| | - Xinping Li
- Max Planck Institute for Biology of Ageing, D-50931 Cologne, Germany
| | - Sara A Wickström
- Max Planck Institute for Biology of Ageing, D-50931 Cologne, Germany.,Helsinki Institute of Life Science, University of Helsinki, FI-00014 Helsinki, Finland.,Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, FI-00014 Helsinki, Finland.,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FI-00014 Helsinki, Finland
| | - Eva Faurobert
- Université Grenoble Alpes, Institute for Advanced Biosciences, Grenoble 38042, France.,INSERM U1209, Institute for Advanced Biosciences, F-38700 La Tronche, France.,CNRS UMR 5039, Institute for Advanced Biosciences, F-38700 La Tronche, France
| | - Corinne Albiges-Rizo
- Université Grenoble Alpes, Institute for Advanced Biosciences, Grenoble 38042, France.,INSERM U1209, Institute for Advanced Biosciences, F-38700 La Tronche, France.,CNRS UMR 5039, Institute for Advanced Biosciences, F-38700 La Tronche, France
| |
Collapse
|
211
|
Li H, Zhang C, Hu Y, Liu P, Sun F, Chen W, Zhang X, Ma J, Wang W, Wang L, Wu P, Liu Z. A reversible shearing DNA probe for visualizing mechanically strong receptors in living cells. Nat Cell Biol 2021; 23:642-651. [PMID: 34059812 DOI: 10.1038/s41556-021-00691-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/28/2021] [Indexed: 02/05/2023]
Abstract
In the last decade, DNA-based tension sensors have made significant contributions to the study of the importance of mechanical forces in many biological systems. Albeit successful, one shortcoming of these techniques is their inability to reversibly measure receptor forces in a higher regime (that is, >20 pN), which limits our understanding of the molecular details of mechanochemical transduction in living cells. Here, we developed a reversible shearing DNA-based tension probe (RSDTP) for probing molecular piconewton-scale forces between 4 and 60 pN transmitted by cells. Using these probes, we can easily distinguish the differences in force-bearing integrins without perturbing adhesion biology and reveal that a strong force-bearing integrin cluster can serve as a 'mechanical pivot' to maintain focal adhesion architecture and facilitate its maturation. The benefits of the RSDTP include a high dynamic range, reversibility and single-molecule sensitivity, all of which will facilitate a better understanding of the molecular mechanisms of mechanobiology.
Collapse
Affiliation(s)
- Hongyun Li
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Chen Zhang
- College of Life Sciences, State Key Laboratory of Virology, Wuhan University, Wuhan, China
| | - Yuru Hu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Pengxiang Liu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Feng Sun
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Wei Chen
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Xinghua Zhang
- The Institute for Advanced Studies, Wuhan University, Wuhan, China.,College of Life Sciences, State Key Laboratory of Virology, Wuhan University, Wuhan, China
| | - Jie Ma
- School of Physics, Sun Yat-sen University, Guangzhou, China
| | - Wenxu Wang
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Liang Wang
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Piyu Wu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Zheng Liu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China.
| |
Collapse
|
212
|
Wasson EM, Dubbin K, Moya ML. Go with the flow: modeling unique biological flows in engineered in vitro platforms. LAB ON A CHIP 2021; 21:2095-2120. [PMID: 34008661 DOI: 10.1039/d1lc00014d] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Interest in recapitulating in vivo phenomena in vitro using organ-on-a-chip technology has grown rapidly and with it, attention to the types of fluid flow experienced in the body has followed suit. These platforms offer distinct advantages over in vivo models with regards to human relevance, cost, and control of inputs (e.g., controlled manipulation of biomechanical cues from fluid perfusion). Given the critical role biophysical forces play in several tissues and organs, it is therefore imperative that engineered in vitro platforms capture the complex, unique flow profiles experienced in the body that are intimately tied with organ function. In this review, we outline the complex and unique flow regimes experienced by three different organ systems: blood vasculature, lymphatic vasculature, and the intestinal system. We highlight current state-of-the-art platforms that strive to replicate physiological flows within engineered tissues while introducing potential limitations in current approaches.
Collapse
Affiliation(s)
- Elisa M Wasson
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Karen Dubbin
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| | - Monica L Moya
- Material Engineering Division, Lawrence Livermore National Laboratory, 7000 East Ave L-222, Livermore, CA 94551, USA.
| |
Collapse
|
213
|
Zbiral B, Weber A, Iturri J, Vivanco MDM, Toca-Herrera JL. Estrogen Modulates Epithelial Breast Cancer Cell Mechanics and Cell-to-Cell Contacts. MATERIALS 2021; 14:ma14112897. [PMID: 34071397 PMCID: PMC8198807 DOI: 10.3390/ma14112897] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023]
Abstract
Excessive estrogen exposure is connected with increased risk of breast cancer and has been shown to promote epithelial-mesenchymal-transition. Malignant cancer cells accumulate changes in cell mechanical and biochemical properties, often leading to cell softening. In this work we have employed atomic force microscopy to probe the influence of estrogen on the viscoelastic properties of MCF-7 breast cancer cells cultured either in normal or hormone free-medium. Estrogen led to a significant softening of the cells in all studied cases, while growing cells in hormone free medium led to an increase in the studied elastic and viscoelastic moduli. In addition, fluorescence microscopy shows that E-cadherin distribution is changed in cells when culturing them under estrogenic conditions. Furthermore, cell-cell contacts seemed to be weakened. These results were supported by AFM imaging showing changes in surfaces roughness, cell-cell contacts and cell height as result of estrogen treatment. This study therefore provides further evidence for the role of estrogen signaling in breast cancer.
Collapse
Affiliation(s)
- Barbara Zbiral
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (J.I.)
| | - Andreas Weber
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (J.I.)
- Correspondence: (A.W.); (J.L.T.-H.)
| | - Jagoba Iturri
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (J.I.)
| | - Maria d. M. Vivanco
- CIC bioGUNE, Basque Research and Technology Alliance, BRTA, Bizkaia Technology Park, 48160 Derio, Spain;
| | - José L. Toca-Herrera
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (J.I.)
- Correspondence: (A.W.); (J.L.T.-H.)
| |
Collapse
|
214
|
Sala S, Oakes PW. Stress fiber strain recognition by the LIM protein testin is cryptic and mediated by RhoA. Mol Biol Cell 2021; 32:1758-1771. [PMID: 34038160 PMCID: PMC8684727 DOI: 10.1091/mbc.e21-03-0156] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The actin cytoskeleton is a key regulator of mechanical processes in cells. The family of LIM domain proteins have recently emerged as important mechanoresponsive cytoskeletal elements capable of sensing strain in the actin cytoskeleton. The mechanisms regulating this mechanosensitive behavior, however, remain poorly understood. Here we show that the LIM domain protein testin is peculiar in that despite the full-length protein primarily appearing diffuse in the cytoplasm, the C-terminal LIM domains alone recognize focal adhesions and strained actin, while the N-terminal domains alone recognize stress fibers. Phosphorylation mutations in the dimerization regions of testin, however, reveal its mechanosensitivity and cause it to relocate to focal adhesions and sites of strain in the actin cytoskeleton. Finally, we demonstrate that activated RhoA causes testin to adorn stress fibers and become mechanosensitive. Together, our data show that testin’s mechanoresponse is regulated in cells and provide new insights into LIM domain protein recognition of the actin cytoskeleton’s mechanical state.
Collapse
Affiliation(s)
- Stefano Sala
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153
| | - Patrick W Oakes
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153
| |
Collapse
|
215
|
Macnamara CK. Biomechanical modelling of cancer: Agent‐based force‐based models of solid tumours within the context of the tumour microenvironment. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2021. [DOI: 10.1002/cso2.1018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Cicely K. Macnamara
- School of Mathematics and Statistics Mathematical Institute University of St Andrews St Andrews Fife UK
| |
Collapse
|
216
|
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: 33] [Impact Index Per Article: 11.0] [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
|
217
|
Li L, Wang H, Xiong C, Luo D, Chen H, Liu Y. Quantify the combined effects of temperature and force on the stability of DNA hairpin. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2021; 33:185102. [PMID: 33711825 DOI: 10.1088/1361-648x/abee38] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/12/2021] [Indexed: 06/12/2023]
Abstract
OxDNA, as a successful coarse-grain model, has been applied to reproduce the thermodynamic and mechanical properties of both single- and double-stranded DNA. In current simulation, oxDNA is extended to explore the combined effects of temperature and force on the stability of DNA hairpin and its free energy landscape. Simulations were carried out at different forces and temperatures, at each temperature, a 18-base-pair DNA hairpin dynamically transited between folded state and unfolded state, and the separation between two states is consistent with the full contour length of single-stranded DNA in the unfolded state. Two methods were used to identify the critical force of DNA hairpin at each temperature and the critical forces obtained from two methods were consistent with each other and gradually decreased with the increasing temperature from 300 K to 326 K. The critical force at 300 K is reasonably consistent with the single molecule result of DNA hairpin with the same stem length. The two-state free energy landscape can be elucidated from the probability distribution of DNA hairpin extension and its dependence on the force and temperature is totally different. The increasing temperature not only reduces the free energy barrier, but also alters the position of transition point along the extension coordinate, resulting in the reduction of folding distance and the extension of unfolding distance, but their sum is not obviously dependent on the temperature. Generally, an assumption that the location of transition state in two-state energy landscape is independent of the stretching force is used to analyze the data of the single molecule experiment, but current simulation results indicate that effects of stretching forces on the location of transition state in two-state energy landscape are dependent on temperature. At relatively high temperature, stretching force can also change the location of transition state in the free energy landscape.
Collapse
Affiliation(s)
- Lin Li
- College of Physics, Guizhou University, Guiyang 550025, People's Republic of China
| | - Hongchang Wang
- School of Physics and Electronic Science, Guizhou Normal University, Guiyang 550025, People's Republic of China
| | - Caiyun Xiong
- College of Physics, Guizhou University, Guiyang 550025, People's Republic of China
| | - Di Luo
- College of Physics, Guizhou University, Guiyang 550025, People's Republic of China
| | - Hu Chen
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Lab for Soft Functional Materials Research, Department of Physics, Xiamen University, Xiamen, People's Republic of China
| | - Yanhui Liu
- College of Physics, Guizhou University, Guiyang 550025, People's Republic of China
| |
Collapse
|
218
|
Martinez A, Buckley M, Scalise CB, Katre AA, Dholakia JJ, Crossman D, Birrer MJ, Berry JL, Arend RC. Understanding the effect of mechanical forces on ovarian cancer progression. Gynecol Oncol 2021; 162:154-162. [PMID: 33888338 DOI: 10.1016/j.ygyno.2021.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/05/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Mechanical forces including tension, compression, and shear stress are increasingly implicated in tumor progression and metastasis. Understanding the mechanisms behind epithelial ovarian cancer (EOC) progression and metastasis is critical, and this study aimed to elucidate the effect of oscillatory and constant tension on EOC. METHODS SKOV-3 and OVCAR-8 EOC cell lines were placed under oscillatory tension for 3 days and compared to cells placed under no tension. Cell proliferation, migration, and invasion were analyzed while RNAseq and Western Blots helped investigate the biological mechanisms underlying the increasingly aggressive state of the experimental cells. Finally, in vivo experiments using SCID mice assisted in confirming the in vitro results. RESULTS Oscillatory tension (OT) and constant tension (CT) significantly increased SKOV-3 proliferation, while OT caused a significant increase in proliferative genes, migration, and invasion in this cell line. CT did not cause significant increases in these areas. Neither OT nor CT increased proliferation or invasion in OVCAR-8 cells, while both tension types significantly increased cellular migration. Two proteins involved in metastasis, E-cadherin and Snail, were both significantly affected by OT in both cell lines, with E-cadherin levels decreasing and Snail levels increasing. In vivo, tumor growth and weight for both cell types were significantly increased, and ascites development was significantly higher in the experimental OVCAR-8 group than in the control group. CONCLUSIONS This study found that mechanical forces are influential in EOC progression and metastasis. Further analysis of downstream mechanisms involved in EOC metastasis will be critical for improvements in EOC treatment.
Collapse
Affiliation(s)
- A Martinez
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - M Buckley
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - C B Scalise
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - A A Katre
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - J J Dholakia
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - D Crossman
- University of Alabama at Birmingham, Department of Genetics, Birmingham, AL 35294, USA
| | - M J Birrer
- University of Arkansas for Medical Sciences, Winthrop P. Rockefeller Cancer Institute, Little Rock, AR 72205, USA
| | - J L Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - R C Arend
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| |
Collapse
|
219
|
Carnicer-Lombarte A, Chen ST, Malliaras GG, Barone DG. Foreign Body Reaction to Implanted Biomaterials and Its Impact in Nerve Neuroprosthetics. Front Bioeng Biotechnol 2021; 9:622524. [PMID: 33937212 PMCID: PMC8081831 DOI: 10.3389/fbioe.2021.622524] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/19/2021] [Indexed: 12/04/2022] Open
Abstract
The implantation of any foreign material into the body leads to the development of an inflammatory and fibrotic process-the foreign body reaction (FBR). Upon implantation into a tissue, cells of the immune system become attracted to the foreign material and attempt to degrade it. If this degradation fails, fibroblasts envelop the material and form a physical barrier to isolate it from the rest of the body. Long-term implantation of medical devices faces a great challenge presented by FBR, as the cellular response disrupts the interface between implant and its target tissue. This is particularly true for nerve neuroprosthetic implants-devices implanted into nerves to address conditions such as sensory loss, muscle paralysis, chronic pain, and epilepsy. Nerve neuroprosthetics rely on tight interfacing between nerve tissue and electrodes to detect the tiny electrical signals carried by axons, and/or electrically stimulate small subsets of axons within a nerve. Moreover, as advances in microfabrication drive the field to increasingly miniaturized nerve implants, the need for a stable, intimate implant-tissue interface is likely to quickly become a limiting factor for the development of new neuroprosthetic implant technologies. Here, we provide an overview of the material-cell interactions leading to the development of FBR. We review current nerve neuroprosthetic technologies (cuff, penetrating, and regenerative interfaces) and how long-term function of these is limited by FBR. Finally, we discuss how material properties (such as stiffness and size), pharmacological therapies, or use of biodegradable materials may be exploited to minimize FBR to nerve neuroprosthetic implants and improve their long-term stability.
Collapse
Affiliation(s)
- Alejandro Carnicer-Lombarte
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Shao-Tuan Chen
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - George G. Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Damiano G. Barone
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
220
|
Sun B, Qu R, Fan T, Yang Y, Jiang X, Khan AU, Zhou Z, Zhang J, Wei K, Ouyang J, Dai J. Actin polymerization state regulates osteogenic differentiation in human adipose-derived stem cells. Cell Mol Biol Lett 2021; 26:15. [PMID: 33858321 PMCID: PMC8048231 DOI: 10.1186/s11658-021-00259-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/03/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Actin is an essential cellular protein that assembles into microfilaments and regulates numerous processes such as cell migration, maintenance of cell shape, and material transport. METHODS In this study, we explored the effect of actin polymerization state on the osteogenic differentiation of human adipose-derived stem cells (hASCs). The hASCs were treated for 7 days with different concentrations (0, 1, 5, 10, 20, and 50 nM) of jasplakinolide (JAS), a reagent that directly polymerizes F-actin. The effects of the actin polymerization state on cell proliferation, apoptosis, migration, and the maturity of focal adhesion-related proteins were assessed. In addition, western blotting and alizarin red staining assays were performed to assess osteogenic differentiation. RESULTS Cell proliferation and migration in the JAS (0, 1, 5, 10, and 20 nM) groups were higher than in the control group and the JAS (50 nM) group. The FAK, vinculin, paxillin, and talin protein expression levels were highest in the JAS (20 nM) group, while zyxin expression was highest in the JAS (50 nM) group. Western blotting showed that osteogenic differentiation in the JAS (0, 1, 5, 10, 20, and 50 nM) group was enhanced compared with that in the control group, and was strongest in the JAS (50 nM) group. CONCLUSIONS In summary, our data suggest that the actin polymerization state may promote the osteogenic differentiation of hASCs by regulating the protein expression of focal adhesion-associated proteins in a concentration-dependent manner. Our findings provide valuable information for exploring the mechanism of osteogenic differentiation in hASCs.
Collapse
Affiliation(s)
- Bing Sun
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yuchao Yang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Xin Jiang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Zhitao Zhou
- Central Laboratory, Southern Medical University, Guangzhou, China
| | - Jingliao Zhang
- Department of Foot and Ankle Surgery, Henan Luoyang Orthopedic Hospital, Zhengzhou, China
| | - Kuanhai Wei
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Guangdong Provincial Key Laboratory of Bone and Cartilage Regeneration Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China.
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics and Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China.
| |
Collapse
|
221
|
Bose S, Dasbiswas K, Gopinath A. Matrix Stiffness Modulates Mechanical Interactions and Promotes Contact between Motile Cells. Biomedicines 2021; 9:biomedicines9040428. [PMID: 33920918 PMCID: PMC8077938 DOI: 10.3390/biomedicines9040428] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 02/07/2023] Open
Abstract
The mechanical micro-environment of cells and tissues influences key aspects of cell structure and function, including cell motility. For proper tissue development, cells need to migrate, interact, and form contacts. Cells are known to exert contractile forces on underlying soft substrates and sense deformations in them. Here, we propose and analyze a minimal biophysical model for cell migration and long-range cell–cell interactions through mutual mechanical deformations of the substrate. We compute key metrics of cell motile behavior, such as the number of cell-cell contacts over a given time, the dispersion of cell trajectories, and the probability of permanent cell contact, and analyze how these depend on a cell motility parameter and substrate stiffness. Our results elucidate how cells may sense each other mechanically and generate coordinated movements and provide an extensible framework to further address both mechanical and short-range biophysical interactions.
Collapse
Affiliation(s)
- Subhaya Bose
- Department of Physics, University of California Merced, Merced, CA 95343, USA; (S.B.); (K.D.)
| | - Kinjal Dasbiswas
- Department of Physics, University of California Merced, Merced, CA 95343, USA; (S.B.); (K.D.)
| | - Arvind Gopinath
- Department of Bioengineering, University of California Merced, Merced, CA 95343, USA
- Correspondence:
| |
Collapse
|
222
|
Pfannenstill V, Barbotin A, Colin-York H, Fritzsche M. Quantitative Methodologies to Dissect Immune Cell Mechanobiology. Cells 2021; 10:851. [PMID: 33918573 PMCID: PMC8069647 DOI: 10.3390/cells10040851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 12/25/2022] Open
Abstract
Mechanobiology seeks to understand how cells integrate their biomechanics into their function and behavior. Unravelling the mechanisms underlying these mechanobiological processes is particularly important for immune cells in the context of the dynamic and complex tissue microenvironment. However, it remains largely unknown how cellular mechanical force generation and mechanical properties are regulated and integrated by immune cells, primarily due to a profound lack of technologies with sufficient sensitivity to quantify immune cell mechanics. In this review, we discuss the biological significance of mechanics for immune cells across length and time scales, and highlight several experimental methodologies for quantifying the mechanics of immune cells. Finally, we discuss the importance of quantifying the appropriate mechanical readout to accelerate insights into the mechanobiology of the immune response.
Collapse
Affiliation(s)
- Veronika Pfannenstill
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK; (V.P.); (A.B.)
| | - Aurélien Barbotin
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK; (V.P.); (A.B.)
| | - Huw Colin-York
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK; (V.P.); (A.B.)
| | - Marco Fritzsche
- Kennedy Institute for Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7LF, UK; (V.P.); (A.B.)
- Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0FA, UK
| |
Collapse
|
223
|
Le MCN, Xu K, Wang Z, Beverung S, Steward RL, Florczyk SJ. Evaluation of the effect of 3D porous Chitosan-alginate scaffold stiffness on breast cancer proliferation and migration. J Biomed Mater Res A 2021; 109:1990-2000. [PMID: 33811775 DOI: 10.1002/jbm.a.37191] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/09/2021] [Accepted: 03/24/2021] [Indexed: 11/11/2022]
Abstract
Breast cancer (BCa) is one of the most common cancers for women and metastatic BCa causes the majority of deaths. The extracellular matrix (ECM) stiffens during cancer progression and provides biophysical signals to modulate proliferation, morphology, and metastasis. Cells utilize mechanotransduction and integrins to sense and respond to ECM stiffness. Chitosan-alginate (CA) scaffolds have been used for 3D culture, but lack integrin binding ligands, resulting in round cell morphology and limited cell-material interaction. In this study, 2, 4, and 6 wt% CA scaffolds were produced to mimic the stages of BCa progression and evaluate the BCa response to CA scaffold stiffness. All three CA scaffold compositions highly porous with interconnected pores and scaffold stiffness increased with increasing polymer concentration. MDA-MB-231 (231) cells were cultured in CA scaffolds and 2D cultures for 7 d. All CA scaffold cultures had similar cell numbers at 7 d and the 231 cells formed clusters that increased in size during the culture. The 2 wt% CA had the largest clusters throughout the 7 d culture compared with the 4 and 6 wt% CA. The 231 cell migration was evaluated on 2D surfaces after 7 d culture. The 6 wt% CA cultured cells had the greatest migration speed, followed by 4 wt% CA, 2D cultures, and 2 wt% CA. These results suggest that 231 cells sensed the stiffness of CA scaffolds without the presence of focal adhesions. This indicates that a non-integrin-based mechanism may explain the observed mechanotransduction response.
Collapse
Affiliation(s)
- Minh-Chau N Le
- Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA.,Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, Florida, USA
| | - Kailei Xu
- Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Zi Wang
- Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA
| | - Sean Beverung
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, Florida, USA
| | - Robert L Steward
- Department of Mechanical and Aerospace Engineering, University of Central Florida, Orlando, Florida, USA.,Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Stephanie J Florczyk
- Department of Materials Science and Engineering, University of Central Florida, Orlando, Florida, USA.,Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
224
|
Subramanian K, Petzold H, Seelbinder B, Hersemann L, Nüsslein I, Kreysing M. Optical plasticity of mammalian cells. JOURNAL OF BIOPHOTONICS 2021; 14:e202000457. [PMID: 33345429 DOI: 10.1002/jbio.202000457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 06/12/2023]
Abstract
Transparency is widespread in nature, ranging from transparent insect wings to ocular tissues that enable you to read this text, and transparent marine vertebrates. And yet, cells and tissue models in biology are usually strongly light scattering and optically opaque, precluding deep optical microscopy. Here we describe the directed evolution of cultured mammalian cells toward increased transparency. We find that mutations greatly diversify the optical phenotype of Chinese Hamster Ovary cells, a cultured mammalian cell line. Furthermore, only three rounds of high-throughput optical selection and competitive growth are required to yield fit cells with greatly improved transparency. Based on 15 monoclonal cell lines derived from this directed evolution experiment, we find that the evolved transparency frequently goes along with a reduction of nuclear granularity and physiological shifts in gene expression profiles. In the future this optical plasticity of mammalian cells may facilitate genetic clearance of living tissues for in vivo microscopy.
Collapse
Affiliation(s)
- Kaushikaram Subramanian
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology Dresden, Dresden, Germany
| | - Heike Petzold
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Benjamin Seelbinder
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology Dresden, Dresden, Germany
| | - Lena Hersemann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology Dresden, Dresden, Germany
| | - Ina Nüsslein
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Moritz Kreysing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology Dresden, Dresden, Germany
- Cluster of Excellence, PoL | Physics of Life, Biotechnology Center of the TU Dresden, Dresden, Germany
| |
Collapse
|
225
|
Evers TMJ, Holt LJ, Alberti S, Mashaghi A. Reciprocal regulation of cellular mechanics and metabolism. Nat Metab 2021; 3:456-468. [PMID: 33875882 PMCID: PMC8863344 DOI: 10.1038/s42255-021-00384-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/12/2021] [Indexed: 12/12/2022]
Abstract
Metabolism and mechanics are intrinsically intertwined. External forces, sensed through the cytoskeleton or distortion of the cell and organelles, induce metabolic changes in the cell. The resulting changes in metabolism, in turn, feed back to regulate every level of cell biology, including the mechanical properties of cells and tissues. Here we examine the links between metabolism and mechanics, highlighting signalling pathways involved in the regulation and response to cellular mechanosensing. We consider how forces and metabolism regulate one another through nanoscale molecular sensors, micrometre-scale cytoskeletal networks, organelles and dynamic biomolecular condensates. Understanding this cross-talk will create diagnostic and therapeutic opportunities for metabolic disorders such as cancer, cardiovascular pathologies and obesity.
Collapse
Affiliation(s)
- Tom M J Evers
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Faculty of Science, Leiden University, Leiden, the Netherlands
| | - Liam J Holt
- Institute for Systems Genetics, New York University Langone Health, New York, NY, USA
| | - Simon Alberti
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Alireza Mashaghi
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Faculty of Science, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
226
|
Activated nanoscale actin-binding domain motion in the catenin-cadherin complex revealed by neutron spin echo spectroscopy. Proc Natl Acad Sci U S A 2021; 118:2025012118. [PMID: 33753508 DOI: 10.1073/pnas.2025012118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
As the core component of the adherens junction in cell-cell adhesion, the cadherin-catenin complex transduces mechanical tension between neighboring cells. Structural studies have shown that the cadherin-catenin complex exists as an ensemble of flexible conformations, with the actin-binding domain (ABD) of α-catenin adopting a variety of configurations. Here, we have determined the nanoscale protein domain dynamics of the cadherin-catenin complex using neutron spin echo spectroscopy (NSE), selective deuteration, and theoretical physics analyses. NSE reveals that, in the cadherin-catenin complex, the motion of the entire ABD becomes activated on nanosecond to submicrosecond timescales. By contrast, in the α-catenin homodimer, only the smaller disordered C-terminal tail of ABD is moving. Molecular dynamics (MD) simulations also show increased mobility of ABD in the cadherin-catenin complex, compared to the α-catenin homodimer. Biased MD simulations further reveal that the applied external forces promote the transition of ABD in the cadherin-catenin complex from an ensemble of diverse conformational states to specific states that resemble the actin-bound structure. The activated motion and an ensemble of flexible configurations of the mechanosensory ABD suggest the formation of an entropic trap in the cadherin-catenin complex, serving as negative allosteric regulation that impedes the complex from binding to actin under zero force. Mechanical tension facilitates the reduction in dynamics and narrows the conformational ensemble of ABD to specific configurations that are well suited to bind F-actin. Our results provide a protein dynamics and entropic explanation for the observed force-sensitive binding behavior of a mechanosensitive protein complex.
Collapse
|
227
|
A Fully Integrated Arduino-Based System for the Application of Stretching Stimuli to Living Cells and Their Time-Lapse Observation: A Do-It-Yourself Biology Approach. Ann Biomed Eng 2021; 49:2243-2259. [PMID: 33728867 DOI: 10.1007/s10439-021-02758-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/20/2021] [Indexed: 10/21/2022]
Abstract
Mechanobiology has nowadays acquired the status of a topic of fundamental importance in a degree in Biological Sciences. It is inherently a multidisciplinary topic where biology, physics and engineering competences are required. A course in mechanobiology should include lab experiences where students can appreciate how mechanical stimuli from outside affect living cell behaviour. Here we describe all the steps to build a cell stretcher inside an on-stage cell incubator. This device allows exposing living cells to a periodic mechanical stimulus similar to what happens in physiological conditions such as, for example, in the vascular system or in the lungs. The reaction of the cells to the periodic mechanical stretching represents a prototype of a mechanobiological signal integrated by living cells. We also provide the theoretical and experimental aspects related to the calibration of the stretcher apparatus at a level accessible to researchers not used to dealing with topics like continuum mechanics and analysis of deformations. We tested our device by stretching cells of two different lines, U87-MG and Balb-3T3 cells, and we analysed and discussed the effect of the periodic stimulus on both cell reorientation and migration. We also discuss the basic aspects related to the quantitative analysis of the reorientation process and of cell migration. We think that the device we propose can be easily reproduced at low-cost within a project-oriented course in the fields of biology, biotechnology and medical engineering.
Collapse
|
228
|
Greenberg MJ, Tardiff JC. Complexity in genetic cardiomyopathies and new approaches for mechanism-based precision medicine. J Gen Physiol 2021; 153:e202012662. [PMID: 33512404 PMCID: PMC7852459 DOI: 10.1085/jgp.202012662] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022] Open
Abstract
Genetic cardiomyopathies have been studied for decades, and it has become increasingly clear that these progressive diseases are more complex than originally thought. These complexities can be seen both in the molecular etiologies of these disorders and in the clinical phenotypes observed in patients. While these disorders can be caused by mutations in cardiac genes, including ones encoding sarcomeric proteins, the disease presentation varies depending on the patient mutation, where mutations even within the same gene can cause divergent phenotypes. Moreover, it is challenging to connect the mutation-induced molecular insult that drives the disease pathogenesis with the various compensatory and maladaptive pathways that are activated during the course of the subsequent progressive, pathogenic cardiac remodeling. These inherent complexities have frustrated our ability to understand and develop broadly effective treatments for these disorders. It has been proposed that it might be possible to improve patient outcomes by adopting a precision medicine approach. Here, we lay out a practical framework for such an approach, where patient subpopulations are binned based on common underlying biophysical mechanisms that drive the molecular disease pathogenesis, and we propose that this function-based approach will enable the development of targeted therapeutics that ameliorate these effects. We highlight several mutations to illustrate the need for mechanistic molecular experiments that span organizational and temporal scales, and we describe recent advances in the development of novel therapeutics based on functional targets. Finally, we describe many of the outstanding questions for the field and how fundamental mechanistic studies, informed by our more nuanced understanding of the clinical disorders, will play a central role in realizing the potential of precision medicine for genetic cardiomyopathies.
Collapse
Affiliation(s)
- Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO
| | - Jil C. Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ
- Department of Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
229
|
Abstract
Biofilms are structured communities formed by a single or multiple microbial species. Within biofilms, bacteria are embedded into extracellular matrix, allowing them to build macroscopic objects. Biofilm structure can respond to environmental changes such as the presence of antibiotics or predators. By adjusting expression levels of surface and extracellular matrix components, bacteria tune cell-to-cell interactions. One major challenge in the field is the fact that these components are very diverse among different species. Deciphering how physical interactions within biofilms are affected by changes in gene expression is a promising approach to obtaining a more unified picture of how bacteria modulate biofilms. This review focuses on recent advances in characterizing attractive and repulsive forces between bacteria in correlation with biofilm structure, dynamics, and spreading. How bacteria control physical interactions to maximize their fitness is an emerging theme.
Collapse
Affiliation(s)
- Berenike Maier
- Institute for Biological Physics and Center for Molecular Medicine Cologne, University of Cologne, 50674 Cologne, Germany;
| |
Collapse
|
230
|
Hernández-Cáceres MP, Munoz L, Pradenas JM, Pena F, Lagos P, Aceiton P, Owen GI, Morselli E, Criollo A, Ravasio A, Bertocchi C. Mechanobiology of Autophagy: The Unexplored Side of Cancer. Front Oncol 2021; 11:632956. [PMID: 33718218 PMCID: PMC7952994 DOI: 10.3389/fonc.2021.632956] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Proper execution of cellular function, maintenance of cellular homeostasis and cell survival depend on functional integration of cellular processes and correct orchestration of cellular responses to stresses. Cancer transformation is a common negative consequence of mismanagement of coordinated response by the cell. In this scenario, by maintaining the balance among synthesis, degradation, and recycling of cytosolic components including proteins, lipids, and organelles the process of autophagy plays a central role. Several environmental stresses activate autophagy, among those hypoxia, DNA damage, inflammation, and metabolic challenges such as starvation. In addition to these chemical challenges, there is a requirement for cells to cope with mechanical stresses stemming from their microenvironment. Cells accomplish this task by activating an intrinsic mechanical response mediated by cytoskeleton active processes and through mechanosensitive protein complexes which interface the cells with their mechano-environment. Despite autophagy and cell mechanics being known to play crucial transforming roles during oncogenesis and malignant progression their interplay is largely overlooked. In this review, we highlight the role of physical forces in autophagy regulation and their potential implications in both physiological as well as pathological conditions. By taking a mechanical perspective, we wish to stimulate novel questions to further the investigation of the mechanical requirements of autophagy and appreciate the extent to which mechanical signals affect this process.
Collapse
Affiliation(s)
- Maria Paz Hernández-Cáceres
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Leslie Munoz
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Javiera M. Pradenas
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Pena
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Lagos
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Aceiton
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Gareth I. Owen
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
- Facultad De Odontología, Instituto De Investigación En Ciencias Odontológicas (ICOD), Universidad De Chile, Santiago, Chile
| | - Andrea Ravasio
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristina Bertocchi
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| |
Collapse
|
231
|
Münch J, Abdelilah-Seyfried S. Sensing and Responding of Cardiomyocytes to Changes of Tissue Stiffness in the Diseased Heart. Front Cell Dev Biol 2021; 9:642840. [PMID: 33718383 PMCID: PMC7952448 DOI: 10.3389/fcell.2021.642840] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiomyocytes are permanently exposed to mechanical stimulation due to cardiac contractility. Passive myocardial stiffness is a crucial factor, which defines the physiological ventricular compliance and volume of diastolic filling with blood. Heart diseases often present with increased myocardial stiffness, for instance when fibrotic changes modify the composition of the cardiac extracellular matrix (ECM). Consequently, the ventricle loses its compliance, and the diastolic blood volume is reduced. Recent advances in the field of cardiac mechanobiology revealed that disease-related environmental stiffness changes cause severe alterations in cardiomyocyte cellular behavior and function. Here, we review the molecular mechanotransduction pathways that enable cardiomyocytes to sense stiffness changes and translate those into an altered gene expression. We will also summarize current knowledge about when myocardial stiffness increases in the diseased heart. Sophisticated in vitro studies revealed functional changes, when cardiomyocytes faced a stiffer matrix. Finally, we will highlight recent studies that described modulations of cardiac stiffness and thus myocardial performance in vivo. Mechanobiology research is just at the cusp of systematic investigations related to mechanical changes in the diseased heart but what is known already makes way for new therapeutic approaches in regenerative biology.
Collapse
Affiliation(s)
- Juliane Münch
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
232
|
Gunasinghe SD, Peres NG, Goyette J, Gaus K. Biomechanics of T Cell Dysfunctions in Chronic Diseases. Front Immunol 2021; 12:600829. [PMID: 33717081 PMCID: PMC7948521 DOI: 10.3389/fimmu.2021.600829] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Understanding the mechanisms behind T cell dysfunctions during chronic diseases is critical in developing effective immunotherapies. As demonstrated by several animal models and human studies, T cell dysfunctions are induced during chronic diseases, spanning from infections to cancer. Although factors governing the onset and the extent of the functional impairment of T cells can differ during infections and cancer, most dysfunctional phenotypes share common phenotypic traits in their immune receptor and biophysical landscape. Through the latest developments in biophysical techniques applied to explore cell membrane and receptor-ligand dynamics, we are able to dissect and gain further insights into the driving mechanisms behind T cell dysfunctions. These insights may prove useful in developing immunotherapies aimed at reinvigorating our immune system to fight off infections and malignancies more effectively. The recent success with checkpoint inhibitors in treating cancer opens new avenues to develop more effective, targeted immunotherapies. Here, we highlight the studies focused on the transformation of the biophysical landscape during infections and cancer, and how T cell biomechanics shaped the immunopathology associated with chronic diseases.
Collapse
Affiliation(s)
- Sachith D Gunasinghe
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Newton G Peres
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Jesse Goyette
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, University of New South Wales, Sydney, NSW, Australia.,ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
233
|
Goult BT. The Mechanical Basis of Memory - the MeshCODE Theory. Front Mol Neurosci 2021; 14:592951. [PMID: 33716664 PMCID: PMC7947202 DOI: 10.3389/fnmol.2021.592951] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
One of the major unsolved mysteries of biological science concerns the question of where and in what form information is stored in the brain. I propose that memory is stored in the brain in a mechanically encoded binary format written into the conformations of proteins found in the cell-extracellular matrix (ECM) adhesions that organise each and every synapse. The MeshCODE framework outlined here represents a unifying theory of data storage in animals, providing read-write storage of both dynamic and persistent information in a binary format. Mechanosensitive proteins that contain force-dependent switches can store information persistently, which can be written or updated using small changes in mechanical force. These mechanosensitive proteins, such as talin, scaffold each synapse, creating a meshwork of switches that together form a code, the so-called MeshCODE. Large signalling complexes assemble on these scaffolds as a function of the switch patterns and these complexes would both stabilise the patterns and coordinate synaptic regulators to dynamically tune synaptic activity. Synaptic transmission and action potential spike trains would operate the cytoskeletal machinery to write and update the synaptic MeshCODEs, thereby propagating this coding throughout the organism. Based on established biophysical principles, such a mechanical basis for memory would provide a physical location for data storage in the brain, with the binary patterns, encoded in the information-storing mechanosensitive molecules in the synaptic scaffolds, and the complexes that form on them, representing the physical location of engrams. Furthermore, the conversion and storage of sensory and temporal inputs into a binary format would constitute an addressable read-write memory system, supporting the view of the mind as an organic supercomputer.
Collapse
Affiliation(s)
- Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
234
|
Pérez-Calixto D, Amat-Shapiro S, Zamarrón-Hernández D, Vázquez-Victorio G, Puech PH, Hautefeuille M. Determination by Relaxation Tests of the Mechanical Properties of Soft Polyacrylamide Gels Made for Mechanobiology Studies. Polymers (Basel) 2021; 13:629. [PMID: 33672475 PMCID: PMC7923444 DOI: 10.3390/polym13040629] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 02/04/2023] Open
Abstract
Following the general aim of recapitulating the native mechanical properties of tissues and organs in vitro, the field of materials science and engineering has benefited from recent progress in developing compliant substrates with physical and chemical properties similar to those of biological materials. In particular, in the field of mechanobiology, soft hydrogels can now reproduce the precise range of stiffnesses of healthy and pathological tissues to study the mechanisms behind cell responses to mechanics. However, it was shown that biological tissues are not only elastic but also relax at different timescales. Cells can, indeed, perceive this dissipation and actually need it because it is a critical signal integrated with other signals to define adhesion, spreading and even more complicated functions. The mechanical characterization of hydrogels used in mechanobiology is, however, commonly limited to the elastic stiffness (Young's modulus) and this value is known to depend greatly on the measurement conditions that are rarely reported in great detail. Here, we report that a simple relaxation test performed under well-defined conditions can provide all the necessary information for characterizing soft materials mechanically, by fitting the dissipation behavior with a generalized Maxwell model (GMM). The simple method was validated using soft polyacrylamide hydrogels and proved to be very useful to readily unveil precise mechanical properties of gels that cells can sense and offer a set of characteristic values that can be compared with what is typically reported from microindentation tests.
Collapse
Affiliation(s)
- Daniel Pérez-Calixto
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (D.P.-C.); (S.A.-S.); (D.Z.-H.); (G.V.-V.)
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
- Posgrado en Ciencia e Ingeniería de Materiales, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Samuel Amat-Shapiro
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (D.P.-C.); (S.A.-S.); (D.Z.-H.); (G.V.-V.)
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Diego Zamarrón-Hernández
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (D.P.-C.); (S.A.-S.); (D.Z.-H.); (G.V.-V.)
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Genaro Vázquez-Victorio
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (D.P.-C.); (S.A.-S.); (D.Z.-H.); (G.V.-V.)
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Pierre-Henri Puech
- Adhesion and Inflammation Lab (LAI), Aix Marseille University, LAI UM 61, Inserm, UMR_S 1067, CNRS, UMR 7333, F-13288 Marseille, France;
| | - Mathieu Hautefeuille
- Departamento de Física, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (D.P.-C.); (S.A.-S.); (D.Z.-H.); (G.V.-V.)
- Laboratorio Nacional de Soluciones Biomiméticas para Diagnóstico y Terapia, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
235
|
Lv Z, de-Carvalho J, Telley IA, Großhans J. Cytoskeletal mechanics and dynamics in the Drosophila syncytial embryo. J Cell Sci 2021; 134:134/4/jcs246496. [PMID: 33597155 DOI: 10.1242/jcs.246496] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cell and tissue functions rely on the genetic programmes and cascades of biochemical signals. It has become evident during the past decade that the physical properties of soft material that govern the mechanics of cells and tissues play an important role in cellular function and morphology. The biophysical properties of cells and tissues are determined by the cytoskeleton, consisting of dynamic networks of F-actin and microtubules, molecular motors, crosslinkers and other associated proteins, among other factors such as cell-cell interactions. The Drosophila syncytial embryo represents a simple pseudo-tissue, with its nuclei orderly embedded in a structured cytoskeletal matrix at the embryonic cortex with no physical separation by cellular membranes. Here, we review the stereotypic dynamics and regulation of the cytoskeleton in Drosophila syncytial embryos and how cytoskeletal dynamics underlies biophysical properties and the emergence of collective features. We highlight the specific features and processes of syncytial embryos and discuss the applicability of biophysical approaches.
Collapse
Affiliation(s)
- Zhiyi Lv
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
| | - Jorge de-Carvalho
- Instituto Gulbenkian de Ciência, Fundação Calouste Gulbenkian, 2780-156 Oeiras, Portugal
| | - Ivo A Telley
- Instituto Gulbenkian de Ciência, Fundação Calouste Gulbenkian, 2780-156 Oeiras, Portugal
| | - Jörg Großhans
- Fachbereich Biologie, Philipps-Universität Marburg, 35043 Marburg, Germany
| |
Collapse
|
236
|
Soto F, Guimarães CF, Reis RL, Franco W, Rizvi I, Demirci U. Emerging biofabrication approaches for gastrointestinal organoids towards patient specific cancer models. Cancer Lett 2021; 504:116-124. [PMID: 33577978 DOI: 10.1016/j.canlet.2021.01.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/14/2021] [Accepted: 01/23/2021] [Indexed: 01/12/2023]
Abstract
Tissue engineered organoids are simple biomodels that can emulate the structural and functional complexity of specific organs. Here, we review developments in three-dimensional (3D) artificial cell constructs to model gastrointestinal dynamics towards cancer diagnosis. We describe bottom-up approaches to fabricate close-packed cell aggregates, from the use of biochemical and physical cues to guide the self-assembly of organoids, to the use of engineering approaches, including 3D printing/additive manufacturing and external field-driven protocols. Finally, we outline the main challenges and possible risks regarding the potential translation of gastrointestinal organoids from laboratory settings to patient-specific models in clinical applications.
Collapse
Affiliation(s)
- Fernando Soto
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA
| | - Carlos F Guimarães
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA; 3B's Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Walfre Franco
- Department of Biomedical Engineering, University of Massachusetts, Lowell, 01854, MA, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, 02114, MA, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA.
| |
Collapse
|
237
|
Almeida AF, Vinhas A, Gonçalves AI, Miranda MS, Rodrigues MT, Gomes ME. Magnetic triggers in biomedical applications - prospects for contact free cell sensing and guidance. J Mater Chem B 2021; 9:1259-1271. [PMID: 33410453 DOI: 10.1039/d0tb02474k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In recent years, the inputs from magnetically assisted strategies have been contributing to the development of more sensitive screening methods and precise means of diagnosis to overcome existing and emerging treatment challenges. The features of magnetic materials enabling in vivo traceability, specific targeting and space- and time-controlled delivery of nanomedicines have highlighted the resourcefulness of the magnetic toolbox for biomedical applications and theranostic strategies. The breakthroughs in magnetically assisted technologies for contact-free control of cell and tissue fate opens new perspectives to improve healing and instruct regeneration reaching a wide range of diseases and disorders. In this review, the contribution of magnetic nanoparticles (MNPs) will be explored as sophisticated and versatile nanotriggers, evidencing their unique cues to probe and control cell function. As cells detect and engage external magnetic features, these approaches will be overviewed considering molecular engineering and cell programming perspectives as well as cell and tissue targeting modalities. The therapeutic relevance of MNPs will be also emphasized as key components of nanostructured systems to control the release of nanomedicines and in the context of new therapy technologies.
Collapse
Affiliation(s)
- Ana F Almeida
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Adriana Vinhas
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana I Gonçalves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Margarida S Miranda
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Márcia T Rodrigues
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Manuela E Gomes
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal. and ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
238
|
Bretherton RC, DeForest CA. The Art of Engineering Biomimetic Cellular Microenvironments. ACS Biomater Sci Eng 2021; 7:3997-4008. [PMID: 33523625 DOI: 10.1021/acsbiomaterials.0c01549] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cells and their surrounding microenvironment exist in dynamic reciprocity, where bidirectional feedback and feedforward crosstalk drives essential processes in development, homeostasis, and disease. With the ongoing explosion of customizable biomaterial innovation for dynamic cell culture, an ever-expanding suite of user-programmable scaffolds now exists to probe cell fate in response to spatiotemporally controlled biophysical and biochemical cues. Here, we highlight emerging trends in these efforts, emphasizing strategies that offer tunability over complex network mechanics, present biomolecular cues anisotropically, and harness cells as physiochemical actuators of the pericellular niche. Altogether, these material advances will lead to breakthroughs in our basic understanding of how cells interact with, integrate signals from, and influence their surrounding microenvironment.
Collapse
Affiliation(s)
- Ross C Bretherton
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States.,Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Cole A DeForest
- Department of Bioengineering, University of Washington, Seattle, Washington 98105, United States.,Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, Washington 98109, United States.,Department of Chemical Engineering, University of Washington, Seattle, Washington 98195, United States.,Department of Chemistry, University of Washington, Seattle, Washington 98195, United States.,Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
239
|
Abstract
Bioimage analysis (BIA) has historically helped study how and why cells move; biological experiments evolved in intimate feedback with the most classical image processing techniques because they contribute objectivity and reproducibility to an eminently qualitative science. Cell segmentation, tracking, and morphology descriptors are all discussed here. Using ameboid motility as a case study, these methods help us illustrate how proper quantification can augment biological data, for example, by choosing mathematical representations that amplify initially subtle differences, by statistically uncovering general laws or by integrating physical insight. More recently, the non-invasive nature of quantitative imaging is fertilizing two blooming fields: mechanobiology, where many biophysical measurements remain inaccessible, and microenvironments, where the quest for physiological relevance has exploded data size. From relief to remedy, this trend indicates that BIA is to become a main vector of biological discovery as human visual analysis struggles against ever more complex data.
Collapse
Affiliation(s)
- Aleix Boquet-Pujadas
- Institut Pasteur, Bioimage Analysis Unit, 25 rue du Dr. Roux, Paris Cedex 15 75724, France
- Centre National de la Recherche Scientifique, CNRS UMR3691, Paris, France
- Sorbonne Université, Paris 75005, France
| | - Jean-Christophe Olivo-Marin
- Institut Pasteur, Bioimage Analysis Unit, 25 rue du Dr. Roux, Paris Cedex 15 75724, France
- Centre National de la Recherche Scientifique, CNRS UMR3691, Paris, France
| | - Nancy Guillén
- Institut Pasteur, Bioimage Analysis Unit, 25 rue du Dr. Roux, Paris Cedex 15 75724, France
- Centre National de la Recherche Scientifique, CNRS ERL9195, Paris, France
| |
Collapse
|
240
|
Özkale B, Sakar MS, Mooney DJ. Active biomaterials for mechanobiology. Biomaterials 2021; 267:120497. [PMID: 33129187 PMCID: PMC7719094 DOI: 10.1016/j.biomaterials.2020.120497] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 02/06/2023]
Abstract
Active biomaterials offer novel approaches to study mechanotransduction in mammalian cells. These material systems probe cellular responses by dynamically modulating their resistance to endogenous forces or applying exogenous forces on cells in a temporally controlled manner. Stimuli-responsive molecules, polymers, and nanoparticles embedded inside cytocompatible biopolymer networks transduce external signals such as light, heat, chemicals, and magnetic fields into changes in matrix elasticity (few kPa to tens of kPa) or forces (few pN to several μN) at the cell-material interface. The implementation of active biomaterials in mechanobiology has generated scientific knowledge and therapeutic potential relevant to a variety of conditions including but not limited to cancer metastasis, fibrosis, and tissue regeneration. We discuss the repertoire of cellular responses that can be studied using these platforms including receptor signaling as well as downstream events namely, cytoskeletal organization, nuclear shuttling of mechanosensitive transcriptional regulators, cell migration, and differentiation. We highlight recent advances in active biomaterials and comment on their future impact.
Collapse
Affiliation(s)
- Berna Özkale
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA, 02138, USA
| | - Mahmut Selman Sakar
- Institute of Mechanical Engineering and Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland.
| | - David J Mooney
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA, 02138, USA.
| |
Collapse
|
241
|
Jin X, Rosenbohm J, Kim E, Esfahani AM, Seiffert-Sinha K, Wahl JK, Lim JY, Sinha AA, Yang R. Modulation of Mechanical Stress Mitigates Anti-Dsg3 Antibody-Induced Dissociation of Cell-Cell Adhesion. Adv Biol (Weinh) 2021; 5:e2000159. [PMID: 33724731 PMCID: PMC7993752 DOI: 10.1002/adbi.202000159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/21/2020] [Indexed: 12/13/2022]
Abstract
It is becoming increasingly clear that mechanical stress in adhesive junctions plays a significant role in dictating the fate of cell-cell attachment under physiological conditions. Targeted disruption of cell-cell junctions leads to multiple pathological conditions, among them the life-threatening autoimmune blistering disease pemphigus vulgaris (PV). The dissociation of cell-cell junctions by autoantibodies is the hallmark of PV, however, the detailed mechanisms that result in tissue destruction remain unclear. Thus far, research and therapy in PV have focused primarily on immune mechanisms upstream of autoantibody binding, while the biophysical aspects of the cell-cell dissociation process leading to acantholysis are less well studied. In work aimed at illuminating the cellular consequences of autoantibody attachment, it is reported that externally applied mechanical stress mitigates antibody-induced monolayer fragmentation and inhibits p38 MAPK phosphorylation activated by anti-Dsg3 antibody. Further, it is demonstrated that mechanical stress applied externally to cell monolayers enhances cell contractility via RhoA activation and promotes the strengthening of cortical actin, which ultimately mitigates antibody-induced cell-cell dissociation. The study elevates understanding of the mechanism of acantholysis in PV and shifts the paradigm of PV disease development from a focus solely on immune pathways to highlight the key role of physical transformations at the target cell.
Collapse
Affiliation(s)
- Xiaowei Jin
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Jordan Rosenbohm
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Eunju Kim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Amir Monemian Esfahani
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | | | - James K Wahl
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, 68583, USA
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Animesh A Sinha
- Department of Dermatology, University at Buffalo, Buffalo, NY, 14203, USA
| | - Ruiguo Yang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
242
|
Blacutt J, Lan Z, Cosgriff-Hernandez EM, Gordon VD. Quantitative confocal microscopy and calibration for measuring differences in cyclic-di-GMP signalling by bacteria on biomedical hydrogels. ROYAL SOCIETY OPEN SCIENCE 2021; 8:201453. [PMID: 33614081 PMCID: PMC7890475 DOI: 10.1098/rsos.201453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/30/2020] [Indexed: 06/12/2023]
Abstract
The growth of bacterial biofilms on implanted medical devices causes harmful infections and device failure. Biofilm development initiates when bacteria attach to and sense a surface. For the common nosocomial pathogen Pseudomonas aeruginosa and many others, the transition to the biofilm phenotype is controlled by the intracellular signal and second messenger cyclic-di-GMP (c-di-GMP). It is not known how biomedical materials might be adjusted to impede c-di-GMP signalling, and there are few extant methods for conducting such studies. Here, we develop such a method. We allowed P. aeruginosa to attach to the surfaces of poly(ethylene glycol) diacrylate (PEGDA) hydrogels. These bacteria contained a plasmid for a green fluorescent protein (GFP) reporter for c-di-GMP. We used laser-scanning confocal microscopy to measure the dynamics of the GFP reporter for 3 h, beginning 1 h after introducing bacteria to the hydrogel. We controlled for the effects of changes in bacterial metabolism using a promoterless plasmid for GFP, and for the effects of light passing through different hydrogels being differently attenuated by using fluorescent plastic beads as 'standard candles' for calibration. We demonstrate that this method can measure statistically significant differences in c-di-GMP signalling associated with different PEGDA gel types and with the surface-exposed protein PilY1.
Collapse
Affiliation(s)
- Jacob Blacutt
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
- Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, TX, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | | | - Vernita D. Gordon
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
- Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, TX, USA
- Department of Physics, The University of Texas at Austin, Austin, TX, USA
- LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
243
|
Romani P, Valcarcel-Jimenez L, Frezza C, Dupont S. Crosstalk between mechanotransduction and metabolism. Nat Rev Mol Cell Biol 2021; 22:22-38. [PMID: 33188273 DOI: 10.1038/s41580-020-00306-w] [Citation(s) in RCA: 202] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2020] [Indexed: 12/22/2022]
Abstract
Mechanical forces shape cells and tissues during development and adult homeostasis. In addition, they also signal to cells via mechanotransduction pathways to control cell proliferation, differentiation and death. These processes require metabolism of nutrients for both energy generation and biosynthesis of macromolecules. However, how cellular mechanics and metabolism are connected is still poorly understood. Here, we discuss recent evidence indicating how the mechanical cues exerted by the extracellular matrix (ECM), cell-ECM and cell-cell adhesion complexes influence metabolic pathways. Moreover, we explore the energy and metabolic requirements associated with cell mechanics and ECM remodelling, implicating a reciprocal crosstalk between cell mechanics and metabolism.
Collapse
Affiliation(s)
- Patrizia Romani
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy
| | | | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge, UK.
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padua Medical School, Padua, Italy.
| |
Collapse
|
244
|
D'Urso M, Kurniawan NA. Mechanical and Physical Regulation of Fibroblast-Myofibroblast Transition: From Cellular Mechanoresponse to Tissue Pathology. Front Bioeng Biotechnol 2020; 8:609653. [PMID: 33425874 PMCID: PMC7793682 DOI: 10.3389/fbioe.2020.609653] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Fibroblasts are cells present throughout the human body that are primarily responsible for the production and maintenance of the extracellular matrix (ECM) within the tissues. They have the capability to modify the mechanical properties of the ECM within the tissue and transition into myofibroblasts, a cell type that is associated with the development of fibrotic tissue through an acute increase of cell density and protein deposition. This transition from fibroblast to myofibroblast-a well-known cellular hallmark of the pathological state of tissues-and the environmental stimuli that can induce this transition have received a lot of attention, for example in the contexts of asthma and cardiac fibrosis. Recent efforts in understanding how cells sense their physical environment at the micro- and nano-scales have ushered in a new appreciation that the substrates on which the cells adhere provide not only passive influence, but also active stimulus that can affect fibroblast activation. These studies suggest that mechanical interactions at the cell-substrate interface play a key role in regulating this phenotype transition by changing the mechanical and morphological properties of the cells. Here, we briefly summarize the reported chemical and physical cues regulating fibroblast phenotype. We then argue that a better understanding of how cells mechanically interact with the substrate (mechanosensing) and how this influences cell behaviors (mechanotransduction) using well-defined platforms that decouple the physical stimuli from the chemical ones can provide a powerful tool to control the balance between physiological tissue regeneration and pathological fibrotic response.
Collapse
Affiliation(s)
- Mirko D'Urso
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
245
|
Unraveling the mechanobiology of immune cells. Curr Opin Biotechnol 2020; 66:236-245. [PMID: 33007634 PMCID: PMC7524653 DOI: 10.1016/j.copbio.2020.09.004] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/02/2020] [Accepted: 09/06/2020] [Indexed: 12/27/2022]
Abstract
Immune cells can sense and respond to biophysical cues - from dynamic forces to spatial features - during their development, activation, differentiation and expansion. These biophysical signals regulate a variety of immune cell functions such as leukocyte extravasation, macrophage polarization, T cell selection and T cell activation. Recent studies have advanced our understanding on immune responses to biophysical cues and the underlying mechanisms of mechanotransduction, which provides rational basis for the design and development of immune-modulatory therapeutics. This review discusses the recent progress in mechanosensing and mechanotransduction of immune cells, particularly monocytes/macrophages and T lymphocytes, and features new biomaterial designs and biomedical devices that translate these findings into biomedical applications.
Collapse
|
246
|
Kabigting JET, Toyama Y. Interplay between caspase, Yes-associated protein, and mechanics: A possible switch between life and death? Curr Opin Cell Biol 2020; 67:141-146. [PMID: 33189987 DOI: 10.1016/j.ceb.2020.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/05/2020] [Accepted: 10/13/2020] [Indexed: 01/05/2023]
Abstract
Organism development requires fine-tuning of the cell number by apoptosis and cell division, as well as proper cell fate specification. These processes are achieved through the integration of intracellular signals and intercellular interactions with neighboring cells as well as the extracellular environment. Apoptosis, a form of cell death typically associated with development and homeostasis, is mainly regulated by the caspase family of proteases. Although caspases are known to initiate and execute apoptosis, it is also known that low caspase levels have a broad spectrum of nonapoptotic functions, including differentiation and organ growth. These different roles of caspases raise intriguing questions: how are caspase levels regulated and what defines the balance between life and death? In this review, we focus on some recent findings that highlight how nonlethal levels of caspase activity, transcriptional coregulator Yes-associated protein (YAP), and mechanical factors influence each other in determining cell fate. We further discuss a possibility that the mechanical signals encountered by cells could regulate the level of caspase activity by mechanics through YAP and, in turn, how this determines whether a cell is susceptible or resistant to undergoing apoptosis in response to cell death stimuli.
Collapse
Affiliation(s)
| | - Yusuke Toyama
- Mechanobiology Institute, Level 5, T-lab Building, 5A Engineering Drive 1, Singapore, 117411; Department of Biological Sciences, National University of Singapore, 14 Science Drive 4, Singapore, 117543.
| |
Collapse
|
247
|
Del Pozo MA, Lolo FN, Echarri A. Caveolae: Mechanosensing and mechanotransduction devices linking membrane trafficking to mechanoadaptation. Curr Opin Cell Biol 2020; 68:113-123. [PMID: 33188985 DOI: 10.1016/j.ceb.2020.10.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023]
Abstract
Mechanical forces (extracellular matrix stiffness, vascular shear stress, and muscle stretching) reaching the plasma membrane (PM) determine cell behavior. Caveolae are PM-invaginated nanodomains with specific lipid and protein composition. Being highly abundant in mechanically challenged tissues (muscles, lungs, vessels, and adipose tissues), they protect cells from mechanical stress damage. Caveolae flatten upon increased PM tension, enabling both force sensing and accommodation, critical for cell mechanoprotection and homeostasis. Thus, caveolae are highly plastic, ranging in complexity from flattened membranes to vacuolar invaginations surrounded by caveolae-rosettes-which also contribute to mechanoprotection. Caveolar components crosstalk with mechanotransduction pathways and recent studies show that they translocate from the PM to the nucleus to convey stress information. Furthermore, caveolae components can regulate membrane traffic from/to the PM to adapt to environmental mechanical forces. The interdependence between lipids and caveolae starts to be understood, and the relevance of caveolae-dependent membrane trafficking linked to mechanoadaption to different physiopathological processes is emerging.
Collapse
Affiliation(s)
- Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| | - Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Asier Echarri
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| |
Collapse
|
248
|
Vasilevich AS, Vermeulen S, Kamphuis M, Roumans N, Eroumé S, Hebels DGAJ, van de Peppel J, Reihs R, Beijer NRM, Carlier A, Carpenter AE, Singh S, de Boer J. On the correlation between material-induced cell shape and phenotypical response of human mesenchymal stem cells. Sci Rep 2020; 10:18988. [PMID: 33149200 PMCID: PMC7642380 DOI: 10.1038/s41598-020-76019-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/13/2020] [Indexed: 12/21/2022] Open
Abstract
Learning rules by which cell shape impacts cell function would enable control of cell physiology and fate in medical applications, particularly, on the interface of cells and material of the implants. We defined the phenotypic response of human bone marrow-derived mesenchymal stem cells (hMSCs) to 2176 randomly generated surface topographies by probing basic functions such as migration, proliferation, protein synthesis, apoptosis, and differentiation using quantitative image analysis. Clustering the surfaces into 28 archetypical cell shapes, we found a very strict correlation between cell shape and physiological response and selected seven cell shapes to describe the molecular mechanism leading to phenotypic diversity. Transcriptomics analysis revealed a tight link between cell shape, molecular signatures, and phenotype. For instance, proliferation is strongly reduced in cells with limited spreading, resulting in down-regulation of genes involved in the G2/M cycle and subsequent quiescence, whereas cells with large filopodia are related to activation of early response genes and inhibition of the osteogenic process. In this paper we were aiming to identify a universal set of genes that regulate the material-induced phenotypical response of human mesenchymal stem cells. This will allow designing implants that can actively regulate cellular, molecular signalling through cell shape. Here we are proposing an approach to tackle this question.
Collapse
Affiliation(s)
- Aliaksei S Vasilevich
- BIS-Biointerface Science in Regenerative Medicine, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Steven Vermeulen
- BIS-Biointerface Science in Regenerative Medicine, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Marloes Kamphuis
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Nadia Roumans
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Said Eroumé
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Dennie G A J Hebels
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Rika Reihs
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Nick R M Beijer
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Anne E Carpenter
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jan de Boer
- BIS-Biointerface Science in Regenerative Medicine, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
249
|
Dawson MR, Xuan B, Hsu J, Ghosh D. Force balancing ACT-IN the tumor microenvironment: Cytoskeletal modifications in cancer and stromal cells to promote malignancy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 360:1-31. [PMID: 33962748 DOI: 10.1016/bs.ircmb.2020.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tumor microenvironment is a complex milieu that dictates the growth, invasion, and metastasis of cancer cells. Both cancer and stromal cells in the tumor tissue encounter and adapt to a variety of extracellular factors, and subsequently contribute and drive the progression of the disease to more advanced stages. As the disease progresses, a small population of cancer cells becomes more invasive through a complex process known as epithelial-mesenchymal transition, and nearby stromal cells assume a carcinoma associated fibroblast phenotype characterized by enhanced migration, cell contractility, and matrix secretion with the ability to reorganize extracellular matrices. As cells transition into more malignant phenotypes their biophysical properties, controlled by the organization of cytoskeletal proteins, are altered. Actin and its associated proteins are essential modulators and facilitators of these changes. As the cells respond to the cues in the microenvironment, actin driven mechanical forces inside and outside the cells also evolve. Recent advances in biophysical techniques have enabled us to probe these actin driven changes in cancer and stromal cells and demarcate their role in driving changes in the microenvironment. Understanding the underlying biophysical mechanisms that drive cancer progression could provide critical insight on novel therapeutic approaches in the fight against cancer.
Collapse
Affiliation(s)
- Michelle R Dawson
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States; Brown University, Center for Biomedical Engineering, Providence, RI, United States.
| | - Botai Xuan
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States
| | - Jeffrey Hsu
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States
| | - Deepraj Ghosh
- Department of Molecular Pharmacology, Physiology, and Biotechnology, Brown University, Providence, RI, United States
| |
Collapse
|
250
|
Mechanically tuning actin filaments to modulate the action of actin-binding proteins. Curr Opin Cell Biol 2020; 68:72-80. [PMID: 33160108 DOI: 10.1016/j.ceb.2020.09.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022]
Abstract
In cells, the actin cytoskeleton is regulated by an interplay between mechanics and biochemistry. A key mechanism, which has emerged based on converging indications from structural, cellular, and biophysical data, depicts the actin filament as a mechanically tunable substrate: mechanical stress applied to an actin filament induces conformational changes, which modify the binding and the regulatory action of actin-binding proteins. For a long time, however, direct evidence of this mechanotransductive mechanism was very scarce. This situation is changing rapidly, and recent in vitro single-filament studies using different techniques have revealed that several actin-binding proteins are able to sense tension, curvature, and/or torsion, applied to actin filaments. Here, we discuss these recent advances and their possible implications.
Collapse
|