1
|
Yang J, Wang P, Zhang Y, Zhang M, Sun Q, Chen H, Dong L, Chu Z, Xue B, Hoff WD, Zhao C, Wang W, Wei Q, Cao Y. Photo-tunable hydrogels reveal cellular sensing of rapid rigidity changes through the accumulation of mechanical signaling molecules. Cell Stem Cell 2025; 32:121-136.e6. [PMID: 39437791 DOI: 10.1016/j.stem.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 07/08/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Cells use traction forces to sense mechanical cues in their environment. While the molecular clutch model effectively explains how cells exert more forces on stiffer substrates, it falls short in addressing their adaptation to dynamic mechanical fluctuations prevalent in tissues and organs. Here, using hydrogel with photo-responsive rigidity, we show that cells' response to rigidity changes is frequency dependent. Strikingly, at certain frequencies, cellular traction forces exceed those on static substrates 4-fold stiffer, challenging the established molecular clutch model. We discover that the discrepancy between the rapid adaptation of traction forces and the slower deactivation of mechanotransduction signaling proteins results in their accumulation, thereby enhancing long-term cellular traction in dynamic settings. Consequently, we propose a new model that melds immediate mechanosensing with extended mechanical signaling. Our study underscores the significance of dynamic rigidity in the development of synthetic biomaterials, emphasizing the importance of considering both immediate and prolonged cellular responses.
Collapse
Affiliation(s)
- Jiapeng Yang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China; College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Peng Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Yu Zhang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Man Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu 610065, China
| | - Qian Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Huiyan Chen
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Liang Dong
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Zhiqin Chu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong 999077, China; Joint Appointment with School of Biomedical Sciences, The University of Hong Kong, Hong Kong 999077, China
| | - Bin Xue
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Wouter David Hoff
- Department of Physics, Oklahoma State University, Stillwater, OK 74078, USA
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Wei Wang
- Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China.
| | - Yi Cao
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan 250021, China; Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center at Nanjing University, Department of Physics, Nanjing University, Nanjing 210093, China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China.
| |
Collapse
|
2
|
Katsuta H, Sokabe M, Hirata H. From stress fiber to focal adhesion: a role of actin crosslinkers in force transmission. Front Cell Dev Biol 2024; 12:1444827. [PMID: 39193363 PMCID: PMC11347286 DOI: 10.3389/fcell.2024.1444827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
The contractile apparatus, stress fiber (SF), is connected to the cell adhesion machinery, focal adhesion (FA), at the termini of SF. The SF-FA complex is essential for various mechanical activities of cells, including cell adhesion to the extracellular matrix (ECM), ECM rigidity sensing, and cell migration. This mini-review highlights the importance of SF mechanics in these cellular activities. Actin-crosslinking proteins solidify SFs by attenuating myosin-driven flows of actin and myosin filaments within the SF. In the solidified SFs, viscous slippage between actin filaments in SFs and between the filaments and the surrounding cytosol is reduced, leading to efficient transmission of myosin-generated contractile force along the SFs. Hence, SF solidification via actin crosslinking ensures exertion of a large force to FAs, enabling FA maturation, ECM rigidity sensing and cell migration. We further discuss intracellular mechanisms for tuning crosslinker-modulated SF mechanics and the potential relationship between the aberrance of SF mechanics and pathology including cancer.
Collapse
Affiliation(s)
- Hiroki Katsuta
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Sokabe
- Human Information Systems Laboratories, Kanazawa Institute of Technology, Hakusan, Japan
| | - Hiroaki Hirata
- Department of Applied Bioscience, Kanazawa Institute of Technology, Hakusan, Japan
| |
Collapse
|
3
|
Fujimoto Y, Nakazawa N. The roles of FHL2 as a mechanotransducer for cellular functions in the mechanical environment. Front Cell Dev Biol 2024; 12:1431690. [PMID: 39129787 PMCID: PMC11310055 DOI: 10.3389/fcell.2024.1431690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/03/2024] [Indexed: 08/13/2024] Open
Abstract
The cell has multiple mechanisms for sensing and responding to dynamic changes in the mechanical environment. In the process, intracellular signaling is activated to modulate gene expression. Recent studies have shown that multifunctional signaling molecules that link intracellular force and gene expression are important for understanding cellular functions in the mechanical environment. This review discusses recent studies on one of the mechanotransducers, Four-and-a-half LIM domains 2 (FHL2), which localizes to focal adhesions (FAs), actin cytoskeleton, and nucleus. FHL2 localizes to FAs and the actin cytoskeleton in the cell on stiff substrate. In this situation, intracellular tension of F-actin by Myosin II is critical for FHL2 localization to FAs and actin stress fibers. In the case, a conserved phenylalanine in each LIM domain is responsible for its localization to F-actin. On the other hand, lower tension of F-actin in the cell on a soft substrate causes FHL2 to be released into the cytoplasm, resulting in its localization in the nucleus. At the molecular level, phosphorylation of specific tyrosine in FHL2 by FAK, non-receptor tyrosine kinase, is critical to nuclear localization. Finally, by binding to transcription factors, FHL2 modulates gene expression for cell proliferation as a transcriptional co-factor. Thus, FHL2 is involved in mechano-sensing and -transduction in the cell in a mechanical environment.
Collapse
Affiliation(s)
- Yukari Fujimoto
- Graduate School of Science and Engineering, Kindai University, Higashiosaka, Japan
| | - Naotaka Nakazawa
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Higashiosaka, Japan
| |
Collapse
|
4
|
Hu X, Jalal S, Yao M, Bakke O, Margadant F, Sheetz M. Differential Talin cleavage in transformed and non-transformed cells and its consequences. Front Cell Dev Biol 2024; 12:1430728. [PMID: 39086658 PMCID: PMC11289324 DOI: 10.3389/fcell.2024.1430728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024] Open
Abstract
This study investigates differences in focal adhesion (FA) morphology and Talin cleavage levels between transformed and non-transformed cell lines. Utilizing fluorescently tagged wild-type Talin and Talin mutants with calpain cleavage site mutations, FA structures were visualized. Mutations in different Talin cleavage sites showed varying impacts on FA morphology and distribution across melanoma cell lines (Meljuso, A375P, A2058) and a non-transformed cell line (HFF). Western blot analysis, ratiometric fluorescence intensity-based measurements, and FRAP experiments revealed higher Talin cleavage levels within FAs of transformed cell lines compared to non-transformed cells. Additionally, growth assays indicated that reducing calpain cleavage levels attenuated transformed cell growth. These findings suggest that Talin cleavage level is crucial for FA morphology and assembly, with higher levels observed in transformed cells, influencing their growth dynamics.
Collapse
Affiliation(s)
- Xian Hu
- Center for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Salma Jalal
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Mingxi Yao
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Oddmund Bakke
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Felix Margadant
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Molecular Mechanomedicine Program, Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, TX, United States
| | - Michael Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Molecular Mechanomedicine Program, Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
5
|
Ravn Berg S, Dikic A, Sharma A, Hagen L, Vågbø CB, Zatula A, Misund K, Waage A, Slupphaug G. Progression of monoclonal gammopathy of undetermined significance to multiple myeloma is associated with enhanced translational quality control and overall loss of surface antigens. J Transl Med 2024; 22:548. [PMID: 38849800 PMCID: PMC11162064 DOI: 10.1186/s12967-024-05345-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Despite significant advancements in treatment strategies, multiple myeloma remains incurable. Additionally, there is a distinct lack of reliable biomarkers that can guide initial treatment decisions and help determine suitable replacement or adjuvant therapies when relapse ensues due to acquired drug resistance. METHODS To define specific proteins and pathways involved in the progression of monoclonal gammopathy of undetermined significance (MGUS) to multiple myeloma (MM), we have applied super-SILAC quantitative proteomic analysis to CD138 + plasma cells from 9 individuals with MGUS and 37 with MM. RESULTS Unsupervised hierarchical clustering defined three groups: MGUS, MM, and MM with an MGUS-like proteome profile (ML) that may represent a group that has recently transformed to MM. Statistical analysis identified 866 differentially expressed proteins between MM and MGUS, and 189 between MM and ML, 177 of which were common between MGUS and ML. Progression from MGUS to MM is accompanied by upregulated EIF2 signaling, DNA repair, and proteins involved in translational quality control, whereas integrin- and actin cytoskeletal signaling and cell surface markers are downregulated. CONCLUSION Compared to the premalignant plasma cells in MGUS, malignant MM cells apparently have mobilized several pathways that collectively contribute to ensure translational fidelity and to avoid proteotoxic stress, especially in the ER. The overall reduced expression of immunoglobulins and surface antigens contribute to this and may additionally mediate evasion from recognition by the immune apparatus. Our analyses identified a range of novel biomarkers with potential prognostic and therapeutic value, which will undergo further evaluation to determine their clinical significance.
Collapse
Affiliation(s)
- Sigrid Ravn Berg
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs hospital, N-7491, Trondheim, Norway
| | - Aida Dikic
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs hospital, N-7491, Trondheim, Norway
| | - Animesh Sharma
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs hospital, N-7491, Trondheim, Norway
- PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, N-7491, Trondheim, Norway
| | - Lars Hagen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs hospital, N-7491, Trondheim, Norway
- PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, N-7491, Trondheim, Norway
| | - Cathrine Broberg Vågbø
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs hospital, N-7491, Trondheim, Norway
- PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, N-7491, Trondheim, Norway
| | - Alexey Zatula
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs hospital, N-7491, Trondheim, Norway
| | - Kristine Misund
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway
- Department of Medical Genetics, St Olavs hospital, N-7491, Trondheim, Norway
| | - Anders Waage
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway
- Department of Hematology, and Biobank1, St Olavs hospital, N-7491, Trondheim, Norway
| | - Geir Slupphaug
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NTNU, N-7491, Trondheim, Norway.
- Clinic of Laboratory Medicine, St. Olavs hospital, N-7491, Trondheim, Norway.
- PROMEC Core Facility for Proteomics and Modomics, Norwegian University of Science and Technology, NTNU, and the Central Norway Regional Health Authority Norway, N-7491, Trondheim, Norway.
| |
Collapse
|
6
|
Chen T, Giannone G. Single molecule imaging unveils cellular architecture, dynamics and mechanobiology. Curr Opin Cell Biol 2024; 88:102369. [PMID: 38759257 DOI: 10.1016/j.ceb.2024.102369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/19/2024]
Abstract
The biomechanical regulation of the cytoskeleton and cell adhesions underlies various essential cellular functions. Studying them requires visualizing their nanostructure and molecular dynamics with evermore precise spatio-temporal resolution. In this review we will focus on the recent advances in single molecule fluorescence imaging techniques and discuss how they improve our understanding of mechanically sensitive cellular structures such as adhesions and the cytoskeleton. We will also discuss future directions for research, emphasizing on the 3D nature of cellular structures and tissues, their mechanical regulation at the molecule level, as well as how super-resolution microscopy will enhance our knowledge on protein structure and conformational changes in the cellular context.
Collapse
Affiliation(s)
- Tianchi Chen
- Interdisciplinary Institute for Neuroscience, Université Bordeaux, CNRS, UMR 5297, 33000 Bordeaux, France
| | - Grégory Giannone
- Interdisciplinary Institute for Neuroscience, Université Bordeaux, CNRS, UMR 5297, 33000 Bordeaux, France.
| |
Collapse
|
7
|
Kengyel A, Palarz PM, Krohn J, Marquardt A, Greve JN, Heiringhoff R, Jörns A, Manstein DJ. Motor properties of Myosin 5c are modulated by tropomyosin isoforms and inhibited by pentabromopseudilin. Front Physiol 2024; 15:1394040. [PMID: 38606007 PMCID: PMC11008601 DOI: 10.3389/fphys.2024.1394040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 04/13/2024] Open
Abstract
Myosin 5c (Myo5c) is a motor protein that is produced in epithelial and glandular tissues, where it plays an important role in secretory processes. Myo5c is composed of two heavy chains, each containing a generic motor domain, an elongated neck domain consisting of a single α-helix with six IQ motifs, each of which binds to a calmodulin (CaM) or a myosin light chain from the EF-hand protein family, a coiled-coil dimer-forming region and a carboxyl-terminal globular tail domain. Although Myo5c is a low duty cycle motor, when two or more Myo5c-heavy meromyosin (HMM) molecules are linked together, they move processively along actin filaments. We describe the purification and functional characterization of human Myo5c-HMM co-produced either with CaM alone or with CaM and the essential and regulatory light chains Myl6 and Myl12b. We describe the extent to which cofilaments of actin and Tpm1.6, Tpm1.8 or Tpm3.1 alter the maximum actin-activated ATPase and motile activity of the recombinant Myo5c constructs. The small allosteric effector pentabromopseudilin (PBP), which is predicted to bind in a groove close to the actin and nucleotide binding site with a calculated ΔG of -18.44 kcal/mol, inhibits the motor function of Myo5c with a half-maximal concentration of 280 nM. Using immunohistochemical staining, we determined the distribution and exact localization of Myo5c in endothelial and endocrine cells from rat and human tissue. Particular high levels of Myo5c were observed in insulin-producing β-cells located within the pancreatic islets of Langerhans.
Collapse
Affiliation(s)
- András Kengyel
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
- Department of Biophysics, University of Pécs Medical School, Pécs, Hungary
| | - Philip M. Palarz
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Jacqueline Krohn
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Anja Marquardt
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Johannes N. Greve
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Robin Heiringhoff
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Dietmar J. Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Shi L, Nadjar-Boger E, Jafarinia H, Carlier A, Wolfenson H. YAP mediates apoptosis through failed integrin adhesion reinforcement. Cell Rep 2024; 43:113811. [PMID: 38393944 DOI: 10.1016/j.celrep.2024.113811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 12/26/2023] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Extracellular matrix (ECM) rigidity is a major effector of cell fate decisions. Whereas cell proliferation on stiff matrices, wherein Yes-associated protein (YAP) plays a pivotal role, is well documented, activation of apoptosis in response to soft matrices is poorly understood. Here, we show that YAP drives the apoptotic decision as well. We find that in cells on soft matrices, YAP is recruited to small adhesions, phosphorylated at the Y357 residue, and translocated into the nucleus, ultimately leading to apoptosis. In contrast, Y357 phosphorylation levels are dramatically low in large adhesions on stiff matrices. Furthermore, mild attenuation of actomyosin contractility allows adhesion growth on soft matrices, leading to reduced Y357 phosphorylation levels and resulting in cell growth. These findings indicate that failed adhesion reinforcement drives rigidity-dependent apoptosis through YAP and that this decision is not determined solely by ECM rigidity but rather by the balance between cellular forces and ECM rigidity.
Collapse
Affiliation(s)
- Lidan Shi
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Elisabeth Nadjar-Boger
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Hamidreza Jafarinia
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, 6200 MD Maastricht, the Netherlands
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
9
|
Kureel SK, Blair B, Sheetz MP. Recent Advancement in Elimination Strategies and Potential Rejuvenation Targets of Senescence. Adv Biol (Weinh) 2024; 8:e2300461. [PMID: 37857532 DOI: 10.1002/adbi.202300461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Indexed: 10/21/2023]
Abstract
Cellular senescence is a state of exiting the cell cycle, resisting apoptosis, and changing phenotype. Senescent cells (SCs) can be identified by large, distorted morphology and irreversible inability to replicate. In early development, senescence has beneficial roles like tissue patterning and wound healing, where SCs are cleared by the immune system. However, there is a steep rise in SC number as organisms age. The issue with SC accumulation stems from the loss of cellular function, alterations of the microenvironment, and secretions of pro-inflammatory molecules, consisting of cytokines, chemokines, matrix metalloproteinases (MMPs), interleukins, and extracellular matrix (ECM)-associated molecules. This secreted cocktail is referred to as the senescence-associated secretory phenotype (SASP), a hallmark of cellular senescence. The SASP promotes inflammation and displays a bystander effect where paracrine signaling turns proliferating cells into senescent states. To alleviate age-associated diseases, researchers have developed novel methods and techniques to selectively eliminate SCs in aged individuals. Although studies demonstrated that selectively killing SCs improves age-related disorders, there are drawbacks to SC removal. Considering favorable aspects of senescence in the body, this paper reviews recent advancements in elimination strategies and potential rejuvenation targets of senescence to bring researchers in the field up to date.
Collapse
Affiliation(s)
- Sanjay Kumar Kureel
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Brandon Blair
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Michael P Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
10
|
Murugan NJ, Cariba S, Abeygunawardena S, Rouleau N, Payne SL. Biophysical control of plasticity and patterning in regeneration and cancer. Cell Mol Life Sci 2023; 81:9. [PMID: 38099951 PMCID: PMC10724343 DOI: 10.1007/s00018-023-05054-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/12/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023]
Abstract
Cells and tissues display a remarkable range of plasticity and tissue-patterning activities that are emergent of complex signaling dynamics within their microenvironments. These properties, which when operating normally guide embryogenesis and regeneration, become highly disordered in diseases such as cancer. While morphogens and other molecular factors help determine the shapes of tissues and their patterned cellular organization, the parallel contributions of biophysical control mechanisms must be considered to accurately predict and model important processes such as growth, maturation, injury, repair, and senescence. We now know that mechanical, optical, electric, and electromagnetic signals are integral to cellular plasticity and tissue patterning. Because biophysical modalities underly interactions between cells and their extracellular matrices, including cell cycle, metabolism, migration, and differentiation, their applications as tuning dials for regenerative and anti-cancer therapies are being rapidly exploited. Despite this, the importance of cellular communication through biophysical signaling remains disproportionately underrepresented in the literature. Here, we provide a review of biophysical signaling modalities and known mechanisms that initiate, modulate, or inhibit plasticity and tissue patterning in models of regeneration and cancer. We also discuss current approaches in biomedical engineering that harness biophysical control mechanisms to model, characterize, diagnose, and treat disease states.
Collapse
Affiliation(s)
- Nirosha J Murugan
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada.
- Allen Discovery Center, Tufts University, Medford, MA, USA.
| | - Solsa Cariba
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Nicolas Rouleau
- Department of Health Sciences, Wilfrid Laurier University, Waterloo, ON, Canada
- Allen Discovery Center, Tufts University, Medford, MA, USA
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Samantha L Payne
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
11
|
Jain K, Lim KYE, Sheetz MP, Kanchanawong P, Changede R. Intrinsic self-organization of integrin nanoclusters within focal adhesions is required for cellular mechanotransduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567975. [PMID: 38045378 PMCID: PMC10690202 DOI: 10.1101/2023.11.20.567975] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Upon interaction with the extracellular matrix, the integrin receptors form nanoclusters as a first biochemical response to ligand binding. Here, we uncover a critical biodesign principle where these nanoclusters are spatially self-organized, facilitating effective mechanotransduction. Mouse Embryonic Fibroblasts (MEFs) with integrin β3 nanoclusters organized themselves with an intercluster distance of ∼550 nm on uniformly coated fibronectin substrates, leading to larger focal adhesions. We determined that this spatial organization was driven by cell-intrinsic factors since there was no pre-existing pattern on the substrates. Altering this spatial organization using cyclo-RGD functionalized Titanium nanodiscs (of 100 nm, corroborating to the integrin nanocluster size) spaced at intervals of 300 nm (almost half), 600 nm (normal) or 1000 nm (almost double) resulted in abrogation in mechanotransduction, indicating that a new parameter i.e., an optimal intercluster distance is necessary for downstream function. Overexpression of α-actinin, which induces a kink in the integrin tail, disrupted the establishment of the optimal intercluster distance, while simultaneous co-overexpression of talin head with α-actinin rescued it, indicating a concentration-dependent competition, and that cytoplasmic activation of integrin by talin head is required for the optimal intercluster organization. Additionally, talin head-mediated recruitment of FHOD1 that facilitates local actin polymerization at nanoclusters, and actomyosin contractility were also crucial for establishing the optimal intercluster distance and a robust mechanotransduction response. These findings demonstrate that cell-intrinsic machinery plays a vital role in organizing integrin receptor nanoclusters within focal adhesions, encoding essential information for downstream mechanotransduction signalling.
Collapse
|
12
|
Meng Y, Huang K, Shi M, Huo Y, Han L, Liu B, Li Y. Research Advances in the Role of the Tropomyosin Family in Cancer. Int J Mol Sci 2023; 24:13295. [PMID: 37686101 PMCID: PMC10488083 DOI: 10.3390/ijms241713295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Cancer is one of the most difficult diseases for human beings to overcome. Its development is closely related to a variety of factors, and its specific mechanisms have been a hot research topic in the field of scientific research. The tropomyosin family (Tpm) is a group of proteins closely related to the cytoskeleton and actin, and recent studies have shown that they play an important role in various cancers, participating in a variety of biological activities, including cell proliferation, invasion, and migration, and have been used as biomarkers for various cancers. The purpose of this review is to explore the research progress of the Tpm family in tumorigenesis development, focusing on the molecular pathways associated with them and their relevant activities involved in tumors. PubMed and Web of Science databases were searched for relevant studies on the role of Tpms in tumorigenesis and development and the activities of Tpms involved in tumors. Data from the literature suggest that the Tpm family is involved in tumor cell proliferation and growth, tumor cell invasion and migration, tumor angiogenesis, tumor cell apoptosis, and immune infiltration of the tumor microenvironment, among other correlations. It can be used as a potential biomarker for early diagnosis, follow-up, and therapeutic response of some tumors. The Tpm family is involved in cancer in a close relationship with miRNAs and LncRNAs. Tpms are involved in tumor tissue invasion and migration as a key link. On this basis, TPM is frequently used as a biomarker for various cancers. However, the specific molecular mechanism of its involvement in cancer progression has not been explained clearly, which remains an important direction for future research.
Collapse
Affiliation(s)
- Yucheng Meng
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Ke Huang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China
| | - Mingxuan Shi
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Yifei Huo
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Liang Han
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| | - Yi Li
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (Y.M.); (K.H.); (M.S.); (Y.H.); (L.H.)
| |
Collapse
|
13
|
Logvinov AS, Nefedova VV, Yampolskaya DS, Kleymenov SY, Levitsky DI, Matyushenko AM. Structural and Functional Properties of Tropomyosin Isoforms Tpm4.1 and Tpm2.1. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:801-809. [PMID: 37748876 DOI: 10.1134/s0006297923060081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 09/27/2023]
Abstract
Tropomyosin (Tpm) is one of the most important partners of actin filament that largely determines its properties. In animal organisms, there are different isoforms of Tpm, which are believed to be involved in the regulation of various cellular functions. However, molecular mechanisms by which various Tpm cytoplasmic regulate of the functioning of actin filaments are still poorly understood. Here, we investigated the properties of Tpm2.1 and Tpm4.1 isoforms and compared them to each other and to more extensively studied Tpm isoforms. Tpm2.1 and Tpm4.1 were very similar in their affinity to F-actin, thermal stability, and resistance to limited proteolysis by trypsin, but differed markedly in the viscosity of their solutions and thermal stability of their complexes with F-actin. The main difference of Tpm2.1 and Tpm4.1 from other Tpm isoforms (e.g., Tpm1.6 and Tpm1.7) was their extremely low thermal stability as measured by the CD and DSC methods. We suggested the possible causes of this instability based on comparing the amino acid sequences of Tpm4.1 and Tpm2.1 with the sequences of Tpm1.6 and Tpm1.7 isoforms, respectively, that have similar exon structure.
Collapse
Affiliation(s)
- Andrey S Logvinov
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Victoria V Nefedova
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Daria S Yampolskaya
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | - Sergey Y Kleymenov
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Dmitrii I Levitsky
- Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, 119071, Russia
| | | |
Collapse
|
14
|
Luo M, Huang M, Yang N, Zhu Y, Huang P, Xu Z, Wang W, Cai L. Impairment of rigidity sensing caused by mutant TP53 gain of function in osteosarcoma. Bone Res 2023; 11:28. [PMID: 37246175 DOI: 10.1038/s41413-023-00265-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/23/2023] [Accepted: 04/18/2023] [Indexed: 05/30/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant pediatric bone tumor and is characterized by high heterogeneity. Studies have revealed a wide range of phenotypic differences among OS cell lines in terms of their in vivo tumorigenicity and in vitro colony-forming abilities. However, the underlying molecular mechanism of these discrepancies remains unclear. The potential role of mechanotransduction in tumorigenicity is of particular interest. To this end, we tested the tumorigenicity and anoikis resistance of OS cell lines both in vitro and in vivo. We utilized a sphere culture model, a soft agar assay, and soft and rigid hydrogel surface culture models to investigate the function of rigidity sensing in the tumorigenicity of OS cells. Additionally, we quantified the expression of sensor proteins, including four kinases and seven cytoskeletal proteins, in OS cell lines. The upstream core transcription factors of rigidity-sensing proteins were further investigated. We detected anoikis resistance in transformed OS cells. The mechanosensing function of transformed OS cells was also impaired, with general downregulation of rigidity-sensing components. We identified toggling between normal and transformed growth based on the expression pattern of rigidity-sensing proteins in OS cells. We further uncovered a novel TP53 mutation (R156P) in transformed OS cells, which acquired gain of function to inhibit rigidity sensing, thus sustaining transformed growth. Our findings suggest a fundamental role of rigidity-sensing components in OS tumorigenicity as mechanotransduction elements through which cells can sense their physical microenvironment. In addition, the gain of function of mutant TP53 appears to serve as an executor for such malignant programs.
Collapse
Affiliation(s)
- Ming Luo
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Mingyang Huang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ningning Yang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yufan Zhu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Peng Huang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhujun Xu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Wengang Wang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Lin Cai
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
15
|
Ketebo AA, Din SU, Lee G, Park S. Mechanobiological Analysis of Nanoparticle Toxicity. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:nano13101682. [PMID: 37242097 DOI: 10.3390/nano13101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/06/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023]
Abstract
Nanoparticles (NPs) are commonly used in healthcare and nanotherapy, but their toxicity at high concentrations is well-known. Recent research has shown that NPs can also cause toxicity at low concentrations, disrupting various cellular functions and leading to altered mechanobiological behavior. While researchers have used different methods to investigate the effects of NPs on cells, including gene expression and cell adhesion assays, the use of mechanobiological tools in this context has been underutilized. This review emphasizes the importance of further exploring the mechanobiological effects of NPs, which could reveal valuable insights into the mechanisms behind NP toxicity. To investigate these effects, different methods, including the use of polydimethylsiloxane (PDMS) pillars to study cell motility, traction force production, and rigidity sensing contractions, have been employed. Understanding how NPs affect cell cytoskeletal functions through mechanobiology could have significant implications, such as developing innovative drug delivery systems and tissue engineering techniques, and could improve the safety of NPs for biomedical applications. In summary, this review highlights the significance of incorporating mechanobiology into the study of NP toxicity and demonstrates the potential of this interdisciplinary field to advance our knowledge and practical use of NPs.
Collapse
Affiliation(s)
- Abdurazak Aman Ketebo
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16499, Republic of Korea
| | - Shahab Ud Din
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16499, Republic of Korea
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
16
|
Melamed S, Zaffryar-Eilot S, Nadjar-Boger E, Aviram R, Zhao H, Yaseen-Badarne W, Kalev-Altman R, Sela-Donenfeld D, Lewinson O, Astrof S, Hasson P, Wolfenson H. Initiation of fibronectin fibrillogenesis is an enzyme-dependent process. Cell Rep 2023; 42:112473. [PMID: 37148241 DOI: 10.1016/j.celrep.2023.112473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/16/2023] [Accepted: 04/19/2023] [Indexed: 05/08/2023] Open
Abstract
Fibronectin fibrillogenesis and mechanosensing both depend on integrin-mediated force transmission to the extracellular matrix. However, force transmission is in itself dependent on fibrillogenesis, and fibronectin fibrils are found in soft embryos where high forces cannot be applied, suggesting that force cannot be the sole initiator of fibrillogenesis. Here, we identify a nucleation step prior to force transmission, driven by fibronectin oxidation mediated by lysyl oxidase enzyme family members. This oxidation induces fibronectin clustering, which promotes early adhesion, alters cellular response to soft matrices, and enhances force transmission to the matrix. In contrast, absence of fibronectin oxidation abrogates fibrillogenesis, perturbs cell-matrix adhesion, and compromises mechanosensation. Moreover, fibronectin oxidation promotes cancer cell colony formation in soft agar as well as collective and single-cell migration. These results reveal a force-independent enzyme-dependent mechanism that initiates fibronectin fibrillogenesis, establishing a critical step in cell adhesion and mechanosensing.
Collapse
Affiliation(s)
- Shay Melamed
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Shelly Zaffryar-Eilot
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Elisabeth Nadjar-Boger
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Rohtem Aviram
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ 07103, USA
| | - Wesal Yaseen-Badarne
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Rotem Kalev-Altman
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, Faculty of Agriculture, Food and Environmental Sciences, The Hebrew University, Rehovot, Israel
| | - Oded Lewinson
- Department of Molecular Microbiology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ 07103, USA
| | - Peleg Hasson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
17
|
Bhattacharya S, Mukherjee A, Pisano S, Dimri S, Knaane E, Altshuler A, Nasser W, Dey S, Shi L, Mizrahi I, Blum N, Jokel O, Amitai-Lange A, Kaganovsky A, Mimouni M, Socea S, Midlij M, Tiosano B, Hasson P, Feral C, Wolfenson H, Shalom-Feuerstein R. The biophysical property of the limbal niche maintains stemness through YAP. Cell Death Differ 2023:10.1038/s41418-023-01156-7. [PMID: 37095157 DOI: 10.1038/s41418-023-01156-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/26/2023] Open
Abstract
The cell fate decisions of stem cells (SCs) largely depend on signals from their microenvironment (niche). However, very little is known about how biochemical niche cues control cell behavior in vivo. To address this question, we focused on the corneal epithelial SC model in which the SC niche, known as the limbus, is spatially segregated from the differentiation compartment. We report that the unique biomechanical property of the limbus supports the nuclear localization and function of Yes-associated protein (YAP), a putative mediator of the mechanotransduction pathway. Perturbation of tissue stiffness or YAP activity affects SC function as well as tissue integrity under homeostasis and significantly inhibited the regeneration of the SC population following SC depletion. In vitro experiments revealed that substrates with the rigidity of the corneal differentiation compartment inhibit nuclear YAP localization and induce differentiation, a mechanism that is mediated by the TGFβ-SMAD2/3 pathway. Taken together, these results indicate that SC sense biomechanical niche signals and that manipulation of mechano-sensory machinery or its downstream biochemical output may bear fruits in SC expansion for regenerative therapy.
Collapse
Affiliation(s)
- Swarnabh Bhattacharya
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel.
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Departments of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Abhishek Mukherjee
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Sabrina Pisano
- Université Côte d'Azur, INSERM, CNRS, IRCAN, 06107, Nice, France
| | - Shalini Dimri
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Eman Knaane
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Anna Altshuler
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Waseem Nasser
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Sunanda Dey
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Lidan Shi
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Ido Mizrahi
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Noam Blum
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Ophir Jokel
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Aya Amitai-Lange
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Anna Kaganovsky
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Michael Mimouni
- Department of Ophthalmology, Rambam Health Care Campus, 31096, Haifa, Israel
| | - Sergiu Socea
- Department of Ophthalmology, Rambam Health Care Campus, 31096, Haifa, Israel
| | - Mohamad Midlij
- Department of Ophthalmology, Hilel Yafe Medical Center, Hadera, Israel
| | - Beatrice Tiosano
- Department of Ophthalmology, Hilel Yafe Medical Center, Hadera, Israel
| | - Peleg Hasson
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel
| | - Chloe Feral
- Université Côte d'Azur, INSERM, CNRS, IRCAN, 06107, Nice, France
| | - Haguy Wolfenson
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel.
| | - Ruby Shalom-Feuerstein
- Department of Genetics & Developmental Biology, The Rappaport Faculty of Medicine & Research Institute, Technion Integrated Cancer Center, Technion - Israel Institute of Technology, 31096, Haifa, Israel.
| |
Collapse
|
18
|
Nunes Vicente F, Chen T, Rossier O, Giannone G. Novel imaging methods and force probes for molecular mechanobiology of cytoskeleton and adhesion. Trends Cell Biol 2023; 33:204-220. [PMID: 36055943 DOI: 10.1016/j.tcb.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 12/01/2022]
Abstract
Detection and conversion of mechanical forces into biochemical signals is known as mechanotransduction. From cells to tissues, mechanotransduction regulates migration, proliferation, and differentiation in processes such as immune responses, development, and cancer progression. Mechanosensitive structures such as integrin adhesions, the actin cortex, ion channels, caveolae, and the nucleus sense and transmit forces. In vitro approaches showed that mechanosensing is based on force-dependent protein deformations and reorganizations. However, the mechanisms in cells remained unclear since cell imaging techniques lacked molecular resolution. Thanks to recent developments in super-resolution microscopy (SRM) and molecular force sensors, it is possible to obtain molecular insight of mechanosensing in live cells. We discuss how understanding of molecular mechanotransduction was revolutionized by these innovative approaches, focusing on integrin adhesions, actin structures, and the plasma membrane.
Collapse
Affiliation(s)
- Filipe Nunes Vicente
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Tianchi Chen
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Olivier Rossier
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France
| | - Grégory Giannone
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, Bordeaux, France.
| |
Collapse
|
19
|
Liu S, Li Y, Hong Y, Wang M, Zhang H, Ma J, Qu K, Huang G, Lu TJ. Mechanotherapy in oncology: Targeting nuclear mechanics and mechanotransduction. Adv Drug Deliv Rev 2023; 194:114722. [PMID: 36738968 DOI: 10.1016/j.addr.2023.114722] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 12/23/2022] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
Mechanotherapy is proposed as a new option for cancer treatment. Increasing evidence suggests that characteristic differences are present in the nuclear mechanics and mechanotransduction of cancer cells compared with those of normal cells. Recent advances in understanding nuclear mechanics and mechanotransduction provide not only further insights into the process of malignant transformation but also useful references for developing new therapeutic approaches. Herein, we present an overview of the alterations of nuclear mechanics and mechanotransduction in cancer cells and highlight their implications in cancer mechanotherapy.
Collapse
Affiliation(s)
- Shaobao Liu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics, Nanjing 210016, PR China
| | - Yuan Li
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuan Hong
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; National Science Foundation Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO 63130, USA
| | - Ming Wang
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Hao Zhang
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics, Nanjing 210016, PR China
| | - Jinlu Ma
- Department of Radiation Oncology, the First Affiliated Hospital, Xian Jiaotong University, Xi'an 710061, PR China
| | - Kai Qu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital, Xian Jiaotong University, Xi'an 710061, PR China
| | - Guoyou Huang
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan 430072, PR China.
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics, Nanjing 210016, PR China.
| |
Collapse
|
20
|
Wu Z, Ge L, Ma L, Lu M, Song Y, Deng S, Duan P, Du T, Wu Y, Zhang Z, Zhang S. TPM2 attenuates progression of prostate cancer by blocking PDLIM7-mediated nuclear translocation of YAP1. Cell Biosci 2023; 13:39. [PMID: 36823643 PMCID: PMC9948342 DOI: 10.1186/s13578-023-00993-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/18/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a common malignant tumor of the genitourinary system. Clinical intervention in advanced PCa remains challenging. Tropomyosins 2 (TPM2) are actin-binding proteins and have been found as a biomarker candidate for certain cancers. However, no studies have explored the role of TPM2 in PCa and its regulatory mechanism. METHODS TPM2 expression was assessed in Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) PCa patient dataset. The effect of TPM2 on PCa progression was assessed in vitro and in vivo by quantifying proliferation, migration, invasion and tumor growth assays, and the mechanism of TPM2 in PCa progression was gradually revealed by Western blotting, immunoprecipitation, and immunofluorescence staining arrays. RESULTS TPM2 was found to be severely downregulated in tumor tissues of PCa patients compared with tumor-adjacent normal tissues. In vitro experiments revealed that TPM2 overexpression inhibited PCa cell proliferation, invasion and androgen-independent proliferation. Moreover, TPM2 overexpression inhibited the growth of subcutaneous xenograft tumors in vivo. Mechanistically, this effect was noted to be dependent on PDZ-binding motif of TPM2. TPM2 competed with YAP1 for binding to PDLIM7 through the PDZ-binding motif. The binding of TPM2 to PDLIM7 subsequently inhibited the nuclear transport function of PDLIM7 for YAP1. YAP1 sequestered in the cytoplasm phosphorylated at S127, resulting in its inactivation or degradation which in turn inhibited the expression of YAP1 downstream target genes. CONCLUSIONS This study investigated the role of TPM2, PDLIM7, and YAP1 in PCa progression and castration resistance. TPM2 attenuates progression of PCa by blocking PDLIM7-mediated nuclear translocation of YAP1. Accordingly, targeting the expression or functional modulation of TPM2, PDLIM7, or YAP1 has the potential to be an effective therapeutic approach to reduce PCa proliferation and prevent the progression of castration-resistant prostate cancer (CRPC).
Collapse
Affiliation(s)
- Zonglong Wu
- grid.411642.40000 0004 0605 3760Department of Urology, Peking University Third Hospital, Beijing, 100191 People’s Republic of China
| | - Liyuan Ge
- grid.411642.40000 0004 0605 3760Department of Urology, Peking University Third Hospital, Beijing, 100191 People’s Republic of China
| | - Lulin Ma
- grid.411642.40000 0004 0605 3760Department of Urology, Peking University Third Hospital, Beijing, 100191 People’s Republic of China
| | - Min Lu
- grid.11135.370000 0001 2256 9319Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Yimeng Song
- grid.411642.40000 0004 0605 3760Department of Urology, Peking University Third Hospital, Beijing, 100191 People’s Republic of China
| | - Shaohui Deng
- grid.411642.40000 0004 0605 3760Department of Urology, Peking University Third Hospital, Beijing, 100191 People’s Republic of China
| | - Peichen Duan
- grid.411642.40000 0004 0605 3760Department of Urology, Peking University Third Hospital, Beijing, 100191 People’s Republic of China
| | - Tan Du
- grid.411642.40000 0004 0605 3760Department of Urology, Peking University Third Hospital, Beijing, 100191 People’s Republic of China
| | - Yaqian Wu
- grid.411642.40000 0004 0605 3760Department of Urology, Peking University Third Hospital, Beijing, 100191 People’s Republic of China
| | - Zhanyi Zhang
- grid.411642.40000 0004 0605 3760Department of Urology, Peking University Third Hospital, Beijing, 100191 People’s Republic of China
| | - Shudong Zhang
- Department of Urology, Peking University Third Hospital, Beijing, 100191, People's Republic of China.
| |
Collapse
|
21
|
Impact of baculoviral transduction of fluorescent actin on cellular forces. Eur J Cell Biol 2023; 102:151294. [PMID: 36791652 DOI: 10.1016/j.ejcb.2023.151294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 01/28/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Live staining of actin brings valuable information in the field of mechanobiology. Gene transfer of GFP-actin has been reported to disturb cell rheological properties while gene transfer of fluorescent actin binding proteins was not. However the influence of gene transfer on cellular forces in adhered cells has never been investigated. This would provide a more complete picture of mechanical disorders induced by actin live staining for mechanobiology studies. Indeed, most of these techniques were shown to alter cell morphology. Change in cell morphology may in itself be sufficient to perturb cellular forces. Here we focus on quantifying the alterations of cellular stresses that result from baculoviral transduction of GFP-actin in MDCK cell line. We report that GFP-actin transduction increases the proportion of cells with large intracellular or surface stresses, especially in epithelia with low cell density. We show that the enhancement of the mechanical stresses is accompanied by small perturbations of cell shape, but not by a significant change in cell size. We thus conclude that this live staining method enhances the cellular forces but only brings subtle shape alterations.
Collapse
|
22
|
Actin crosslinking by α-actinin averts viscous dissipation of myosin force transmission in stress fibers. iScience 2023; 26:106090. [PMID: 36852278 PMCID: PMC9958379 DOI: 10.1016/j.isci.2023.106090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Contractile force generated in actomyosin stress fibers (SFs) is transmitted along SFs to the extracellular matrix (ECM), which contributes to cell migration and sensing of ECM rigidity. In this study, we show that efficient force transmission along SFs relies on actin crosslinking by α-actinin. Upon reduction of α-actinin-mediated crosslinks, the myosin II activity induced flows of actin filaments and myosin II along SFs, leading to a decrease in traction force exertion to ECM. The fluidized SFs maintained their cable integrity probably through enhanced actin polymerization throughout SFs. A computational modeling analysis suggested that lowering the density of actin crosslinks caused viscous slippage of actin filaments in SFs and, thereby, dissipated myosin-generated force transmitting along SFs. As a cellular scale outcome, α-actinin depletion attenuated the ECM-rigidity-dependent difference in cell migration speed, which suggested that α-actinin-modulated SF mechanics is involved in the cellular response to ECM rigidity.
Collapse
|
23
|
Amer M, Wolfenson H. Measuring Cellular Traction Forces with Micropillar Arrays. Methods Mol Biol 2023; 2600:197-206. [PMID: 36587099 DOI: 10.1007/978-1-0716-2851-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Accurately evaluating cellular forces is critical for studying mechanosensing and mechanotransduction processes, and it necessitates sensitive measurements on the piconewton scale. Here we describe a specialized method that employs elastic polydimethylsiloxane (PDMS) micropillar arrays, which cells can adhere to and bend. The flexibility of the pillars correlates with their heights; the longer they are, the easier they are to bend. Thus, an array of taller pillars mimics a relatively soft substrate that readily yields in response to cellular forces. Tracking cell movements and pillar displacements using live-cell microscopy enables the calculation of cellular forces and the tracking of their dynamic features throughout early and late stages of cell spreading on the pillars. This technique offers the advantage of high spatial and temporal resolution analyses and constitutes a method to investigate the effect of substrate rigidities on cellular functions.
Collapse
Affiliation(s)
- Malak Amer
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
24
|
Sun Q, Pei F, Zhang M, Zhang B, Jin Y, Zhao Z, Wei Q. Curved Nanofiber Network Induces Cellular Bridge Formation to Promote Stem Cell Mechanotransduction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204479. [PMID: 36382560 PMCID: PMC9875655 DOI: 10.1002/advs.202204479] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Remarkable exertions are directed to reveal and understand topographic cues that induce cell mechanical sensitive responses including lineage determination. Extracellular matrix (ECM) is the sophisticated ensemble of diverse factors offering the complicated cellular microenvironment to regulate cell behaviors. However, the functions of only a few of these factors are revealed; most of them are still poorly understood. Herein, the focus is on understanding the curved structure in ECM network for regulating stem cell mechanotransduction. A curved nanofiber network mimicking the curved structure in ECM is fabricated by an improved electrospinning technology. Compared with the straight fibers, the curved fibers promote cell bridge formation because of the cytoskeleton tension. The actomyosin filaments are condensed near the curved edge of the non-adhesive bridge in the bridging cells, which generates higher myosin-II-based intracellular force. This force drives cell lineage commitment toward osteogenic differentiation. This study enriches and perfects the knowledge of the effects of topographic cues on cell behaviors and guides the development of novel biomaterials.
Collapse
Affiliation(s)
- Qian Sun
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065P. R. China
| | - Fang Pei
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065P. R. China
| | - Man Zhang
- College of Biomedical EngineeringSichuan UniversityChengdu610065P. R. China
| | - Bo Zhang
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Ying Jin
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Zhihe Zhao
- Department of OrthodonticsState Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Qiang Wei
- College of Polymer Science and EngineeringState Key Laboratory of Polymer Materials and EngineeringSichuan UniversityChengdu610065P. R. China
- College of Biomedical EngineeringSichuan UniversityChengdu610065P. R. China
| |
Collapse
|
25
|
Yao M, Tijore A, Cheng D, Li JV, Hariharan A, Martinac B, Tran Van Nhieu G, Cox CD, Sheetz M. Force- and cell state-dependent recruitment of Piezo1 drives focal adhesion dynamics and calcium entry. SCIENCE ADVANCES 2022; 8:eabo1461. [PMID: 36351022 PMCID: PMC9645726 DOI: 10.1126/sciadv.abo1461] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/21/2022] [Indexed: 11/11/2022]
Abstract
Mechanosensing is an integral part of many physiological processes including stem cell differentiation, fibrosis, and cancer progression. Two major mechanosensing systems-focal adhesions and mechanosensitive ion channels-can convert mechanical features of the microenvironment into biochemical signals. We report here unexpectedly that the mechanosensitive calcium-permeable channel Piezo1, previously perceived to be diffusive on plasma membranes, binds to matrix adhesions in a force-dependent manner, promoting cell spreading, adhesion dynamics, and calcium entry in normal but not in most cancer cells tested except some glioblastoma lines. A linker domain in Piezo1 is needed for binding to adhesions, and overexpression of the domain blocks Piezo1 binding to adhesions, decreasing adhesion size and cell spread area. Thus, we suggest that Piezo1 is a previously unidentified component of focal adhesions in nontransformed cells that catalyzes adhesion maturation and growth through force-dependent calcium signaling, but this function is absent in most cancer cells.
Collapse
Affiliation(s)
- Mingxi Yao
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen 518055, China
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Corresponding author. (M.Y); (C.C.); (M.S.)
| | - Ajay Tijore
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Center for Biosystems Science and Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Delfine Cheng
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
| | - Jinyuan Vero Li
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
| | - Anushya Hariharan
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
| | - Guy Tran Van Nhieu
- Ecole Normale Supérieure Paris-Saclay Gif-sur-Yvette, France
- Team Ca Signaling and Microbial Infections, Institute for Integrative Biology of the Cell–CNRS UMR9198–Inserm U1280, 1, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Charles D. Cox
- Victor Chang Cardiac Research Institute, Sydney NSW 2010, Australia
- Corresponding author. (M.Y); (C.C.); (M.S.)
| | - Michael Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411
- Department of Biological Sciences, National University of Singapore, Singapore 117558
- Molecular MechanoMedicine Program, Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
- Corresponding author. (M.Y); (C.C.); (M.S.)
| |
Collapse
|
26
|
Tijore A, Yang B, Sheetz M. Cancer cells can be killed mechanically or with combinations of cytoskeletal inhibitors. Front Pharmacol 2022; 13:955595. [PMID: 36299893 PMCID: PMC9589226 DOI: 10.3389/fphar.2022.955595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/12/2022] [Indexed: 12/24/2022] Open
Abstract
For over two centuries, clinicians have hypothesized that cancer developed preferentially at the sites of repeated damage, indicating that cancer is basically “continued healing.” Tumor cells can develop over time into other more malignant types in different environments. Interestingly, indefinite growth correlates with the depletion of a modular, early rigidity sensor, whereas restoring these sensors in tumor cells blocks tumor growth on soft surfaces and metastases. Importantly, normal and tumor cells from many different tissues exhibit transformed growth without the early rigidity sensor. When sensors are restored in tumor cells by replenishing depleted mechanosensory proteins that are often cytoskeletal, cells revert to normal rigidity-dependent growth. Surprisingly, transformed growth cells are sensitive to mechanical stretching or ultrasound which will cause apoptosis of transformed growth cells (Mechanoptosis). Mechanoptosis is driven by calcium entry through mechanosensitive Piezo1 channels that activate a calcium-induced calpain response commonly found in tumor cells. Since tumor cells from many different tissues are in a transformed growth state that is, characterized by increased growth, an altered cytoskeleton and mechanoptosis, it is possible to inhibit growth of many different tumors by mechanical activity and potentially by cytoskeletal inhibitors.
Collapse
Affiliation(s)
- Ajay Tijore
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
- *Correspondence: Ajay Tijore, ; Michael Sheetz,
| | - Bo Yang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Michael Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States
- *Correspondence: Ajay Tijore, ; Michael Sheetz,
| |
Collapse
|
27
|
Chighizola M, Dini T, Marcotti S, D'Urso M, Piazzoni C, Borghi F, Previdi A, Ceriani L, Folliero C, Stramer B, Lenardi C, Milani P, Podestà A, Schulte C. The glycocalyx affects the mechanotransductive perception of the topographical microenvironment. J Nanobiotechnology 2022; 20:418. [PMID: 36123687 PMCID: PMC9484177 DOI: 10.1186/s12951-022-01585-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/29/2022] [Indexed: 11/10/2022] Open
Abstract
The cell/microenvironment interface is the starting point of integrin-mediated mechanotransduction, but many details of mechanotransductive signal integration remain elusive due to the complexity of the involved (extra)cellular structures, such as the glycocalyx. We used nano-bio-interfaces reproducing the complex nanotopographical features of the extracellular matrix to analyse the glycocalyx impact on PC12 cell mechanosensing at the nanoscale (e.g., by force spectroscopy with functionalised probes). Our data demonstrates that the glycocalyx configuration affects spatio-temporal nanotopography-sensitive mechanotransductive events at the cell/microenvironment interface. Opposing effects of major glycocalyx removal were observed, when comparing flat and specific nanotopographical conditions. The excessive retrograde actin flow speed and force loading are strongly reduced on certain nanotopographies upon strong reduction of the native glycocalyx, while on the flat substrate we observe the opposite trend. Our results highlight the importance of the glycocalyx configuration in a molecular clutch force loading-dependent cellular mechanism for mechanosensing of microenvironmental nanotopographical features.
Collapse
Affiliation(s)
- Matteo Chighizola
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Tania Dini
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.,The FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Stefania Marcotti
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Mirko D'Urso
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.,Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Claudio Piazzoni
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Francesca Borghi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Anita Previdi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Laura Ceriani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Claudia Folliero
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.,The FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Brian Stramer
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Cristina Lenardi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Paolo Milani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Alessandro Podestà
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.
| | - Carsten Schulte
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.
| |
Collapse
|
28
|
Missirlis D, Heckmann L, Haraszti T, Spatz JP. Fibronectin anchoring to viscoelastic poly(dimethylsiloxane) elastomers controls fibroblast mechanosensing and directional motility. Biomaterials 2022; 287:121646. [PMID: 35785752 DOI: 10.1016/j.biomaterials.2022.121646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/24/2022] [Accepted: 06/22/2022] [Indexed: 11/19/2022]
Abstract
The established link between deregulated tissue mechanics and various pathological states calls for the elucidation of the processes through which cells interrogate and interpret the mechanical properties of their microenvironment. In this work, we demonstrate that changes in the presentation of the extracellular matrix protein fibronectin on the surface of viscoelastic silicone elastomers have an overarching effect on cell mechanosensing, that is independent of bulk mechanics. Reduction of surface hydrophilicity resulted in altered fibronectin adsorption strength as monitored using atomic force microscopy imaging and pulling experiments. Consequently, primary human fibroblasts were able to remodel the fibronectin coating, adopt a polarized phenotype and migrate directionally even on soft elastomers, that otherwise were not able to resist the applied traction forces. The findings presented here provide valuable insight on how cellular forces are regulated by ligand presentation and used by cells to probe their mechanical environment, and have implications on biomaterial design for cell guidance.
Collapse
Affiliation(s)
- Dimitris Missirlis
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Postal Address: Jahnstr. 29, D-69120, Heidelberg, Germany.
| | - Lara Heckmann
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Postal Address: Jahnstr. 29, D-69120, Heidelberg, Germany
| | - Tamás Haraszti
- DWI - Leibniz Institute for Interactive Materials, Postal Address: Forkenbeckstr. 50, D-52056, Aachen, Germany
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Postal Address: Jahnstr. 29, D-69120, Heidelberg, Germany; Department of Biophysical Chemistry, Physical Chemistry Institute, Heidelberg University, Postal Address: INF 253, D-69120, Heidelberg, Germany
| |
Collapse
|
29
|
Qin R, Melamed S, Yang B, Saxena M, Sheetz MP, Wolfenson H. Tumor Suppressor DAPK1 Catalyzes Adhesion Assembly on Rigid but Anoikis on Soft Matrices. Front Cell Dev Biol 2022; 10:959521. [PMID: 35927990 PMCID: PMC9343699 DOI: 10.3389/fcell.2022.959521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/24/2022] [Indexed: 01/05/2023] Open
Abstract
Cancer cells normally grow on soft surfaces due to impaired mechanosensing of the extracellular matrix rigidity. Upon restoration of proper mechanosensing, cancer cells undergo apoptosis on soft surfaces (anoikis) like most normal cells. However, the link between mechanosensing and activation of anoikis is not clear. Here we show that death associated protein kinase 1 (DAPK1), a tumor suppressor that activates cell death, is directly linked to anoikis activation through rigidity sensing. We find that when rigidity sensing is decreased through inhibition of DAPK1 activity, cells are transformed for growth on soft matrices. Further, DAPK1 catalyzes matrix adhesion assembly and is part of adhesions on rigid surfaces. This pathway involves DAPK1 phosphorylation of tropomyosin1.1, the talin1 head domain, and tyrosine phosphorylation of DAPK1 by Src. On soft surfaces, DAPK1 rapidly dissociates from the adhesion complexes and activates apoptosis as catalyzed by PTPN12 activity and talin1 head. Thus, DAPK1 is important for adhesion assembly on rigid surfaces and the activation of anoikis on soft surfaces through its binding to rigidity-sensing modules.
Collapse
Affiliation(s)
- Ruifang Qin
- Department of Biological Sciences, Columbia University, New York City, NY, United States
| | - Shay Melamed
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Bo Yang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Mayur Saxena
- Department of Biomedical Engineering, Columbia University, New York City, NY, United States
| | - Michael P. Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, United States
- *Correspondence: Haguy Wolfenson, ; Michael P. Sheetz,
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
- *Correspondence: Haguy Wolfenson, ; Michael P. Sheetz,
| |
Collapse
|
30
|
De Belly H, Paluch EK, Chalut KJ. Interplay between mechanics and signalling in regulating cell fate. Nat Rev Mol Cell Biol 2022; 23:465-480. [PMID: 35365816 DOI: 10.1038/s41580-022-00472-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/11/2022]
Abstract
Mechanical signalling affects multiple biological processes during development and in adult organisms, including cell fate transitions, cell migration, morphogenesis and immune responses. Here, we review recent insights into the mechanisms and functions of two main routes of mechanical signalling: outside-in mechanical signalling, such as mechanosensing of substrate properties or shear stresses; and mechanical signalling regulated by the physical properties of the cell surface itself. We discuss examples of how these two classes of mechanical signalling regulate stem cell function, as well as developmental processes in vivo. We also discuss how cell surface mechanics affects intracellular signalling and, in turn, how intracellular signalling controls cell surface mechanics, generating feedback into the regulation of mechanosensing. The cooperation between mechanosensing, intracellular signalling and cell surface mechanics has a profound impact on biological processes. We discuss here our understanding of how these three elements interact to regulate stem cell fate and development.
Collapse
Affiliation(s)
- Henry De Belly
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Ewa K Paluch
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Kevin J Chalut
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Wellcome/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
31
|
Mukherjee A, Melamed S, Damouny-Khoury H, Amer M, Feld L, Nadjar-Boger E, Sheetz MP, Wolfenson H. α-Catenin links integrin adhesions to F-actin to regulate ECM mechanosensing and rigidity dependence. J Cell Biol 2022; 221:213257. [PMID: 35652786 PMCID: PMC9166284 DOI: 10.1083/jcb.202102121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 12/22/2021] [Accepted: 05/16/2022] [Indexed: 02/03/2023] Open
Abstract
Both cell-cell and cell-matrix adhesions are regulated by mechanical signals, but the mechanobiological processes that mediate the cross talk between these structures are poorly understood. Here we show that α-catenin, a mechanosensitive protein that is classically linked with cadherin-based adhesions, associates with and regulates integrin adhesions. α-Catenin is recruited to the edges of mesenchymal cells, where it interacts with F-actin. This is followed by mutual retrograde flow of α-catenin and F-actin from the cell edge, during which α-catenin interacts with vinculin within integrin adhesions. This interaction affects adhesion maturation, stress-fiber assembly, and force transmission to the matrix. In epithelial cells, α-catenin is present in cell-cell adhesions and absent from cell-matrix adhesions. However, when these cells undergo epithelial-to-mesenchymal transition, α-catenin transitions to the cell edge, where it facilitates proper mechanosensing. This is highlighted by the ability of α-catenin-depleted cells to grow on soft matrices. These results suggest a dual role of α-catenin in mechanosensing, through both cell-cell and cell-matrix adhesions.
Collapse
Affiliation(s)
- Abhishek Mukherjee
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Shay Melamed
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Hana Damouny-Khoury
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Malak Amer
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Lea Feld
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Elisabeth Nadjar-Boger
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Michael P. Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel,Correspondence to Haguy Wolfenson:
| |
Collapse
|
32
|
Shi Y, Sivarajan S, Crocker JC, Reich DH. Measuring Cytoskeletal Mechanical Fluctuations and Rheology with Active Micropost Arrays. Curr Protoc 2022; 2:e433. [PMID: 35612274 PMCID: PMC9321978 DOI: 10.1002/cpz1.433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The dynamics of the cellular actomyosin cytoskeleton are crucial to many aspects of cellular function. Here, we describe techniques that employ active micropost array detectors (AMPADs) to measure cytoskeletal rheology and mechanical force fluctuations. The AMPADS are arrays of flexible poly(dimethylsiloxane) (PDMS) microposts with magnetic nanowires embedded in a subset of microposts to enable actuation of those posts via an externally applied magnetic field. Techniques are described to track the magnetic microposts' motion with nanometer precision at up to 100 video frames per second to measure the local cellular rheology at well-defined positions. Application of these high-precision tracking techniques to the full array of microposts in contact with a cell also enables mapping of the cytoskeletal mechanical fluctuation dynamics with high spatial and temporal resolution. This article describes (1) the fabrication of magnetic micropost arrays, (2) measurement protocols for both local rheology and cytoskeletal force fluctuation mapping, and (3) special-purpose software routines to reduce and analyze these data. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Fabrication of magnetic micropost arrays Basic Protocol 2: Data acquisition for cellular force fluctuations on non-magnetic micropost arrays Basic Protocol 3: Data acquisition for local cellular rheology measurements with magnetic microposts Basic Protocol 4: Data reduction: determining microposts' motion Basic Protocol 5: Data analysis: determining local rheology from magnetic microposts Basic Protocol 6: Data analysis for force fluctuation measurements Support Protocol 1: Fabrication of magnetic Ni nanowires by electrodeposition Support Protocol 2: Configuring Streampix for magnetic rheology measurements.
Collapse
Affiliation(s)
- Yu Shi
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, Maryland.,Current address: Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina
| | - Shankar Sivarajan
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, Maryland
| | - John C Crocker
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel H Reich
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
33
|
Reindl T, Giese S, Greve JN, Reinke PY, Chizhov I, Latham SL, Mulvihill DP, Taft MH, Manstein DJ. Distinct actin–tropomyosin cofilament populations drive the functional diversification of cytoskeletal myosin motor complexes. iScience 2022; 25:104484. [PMID: 35720262 PMCID: PMC9204724 DOI: 10.1016/j.isci.2022.104484] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/02/2022] [Accepted: 05/24/2022] [Indexed: 11/02/2022] Open
Abstract
The effects of N-terminal acetylation of the high molecular weight tropomyosin isoforms Tpm1.6 and Tpm2.1 and the low molecular weight isoforms Tpm1.12, Tpm3.1, and Tpm4.2 on the actin affinity and the thermal stability of actin-tropomyosin cofilaments are described. Furthermore, we show how the exchange of cytoskeletal tropomyosin isoforms and their N-terminal acetylation affects the kinetic and chemomechanical properties of cytoskeletal actin-tropomyosin-myosin complexes. Our results reveal the extent to which the different actin-tropomyosin-myosin complexes differ in their kinetic and functional properties. The maximum sliding velocity of the actin filament as well as the optimal motor density for continuous unidirectional movement, parameters that were previously considered to be unique and invariant properties of each myosin isoform, are shown to be influenced by the exchange of the tropomyosin isoform and the N-terminal acetylation of tropomyosin. Tpm diversity is largely determined by sequences contributing to the overlap region Global sequence differences are of greater importance than variable exon 6 usage Tpm isoforms confer distinctly altered properties to cytoskeletal myosin motors Cytoskeletal myosins are differentially affected by N-terminal acetylation of Tpm
Collapse
|
34
|
Seetharaman S, Vianay B, Roca V, Farrugia AJ, De Pascalis C, Boëda B, Dingli F, Loew D, Vassilopoulos S, Bershadsky A, Théry M, Etienne-Manneville S. Microtubules tune mechanosensitive cell responses. NATURE MATERIALS 2022; 21:366-377. [PMID: 34663953 DOI: 10.1038/s41563-021-01108-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 08/20/2021] [Indexed: 05/05/2023]
Abstract
Mechanotransduction is a process by which cells sense the mechanical properties of their surrounding environment and adapt accordingly to perform cellular functions such as adhesion, migration and differentiation. Integrin-mediated focal adhesions are major sites of mechanotransduction and their connection with the actomyosin network is crucial for mechanosensing as well as for the generation and transmission of forces onto the substrate. Despite having emerged as major regulators of cell adhesion and migration, the contribution of microtubules to mechanotransduction still remains elusive. Here, we show that talin- and actomyosin-dependent mechanosensing of substrate rigidity controls microtubule acetylation (a tubulin post-translational modification) by promoting the recruitment of α-tubulin acetyltransferase 1 (αTAT1) to focal adhesions. Microtubule acetylation tunes the mechanosensitivity of focal adhesions and Yes-associated protein (YAP) translocation. Microtubule acetylation, in turn, promotes the release of the guanine nucleotide exchange factor GEF-H1 from microtubules to activate RhoA, actomyosin contractility and traction forces. Our results reveal a fundamental crosstalk between microtubules and actin in mechanotransduction that contributes to mechanosensitive cell adhesion and migration.
Collapse
Affiliation(s)
- Shailaja Seetharaman
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Université Paris Descartes, Paris, France
| | - Benoit Vianay
- Paris University, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, Paris, France
| | - Vanessa Roca
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Aaron J Farrugia
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Chiara De Pascalis
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Batiste Boëda
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Florent Dingli
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | - Damarys Loew
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | | | - Alexander Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Manuel Théry
- Paris University, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
35
|
Seetharaman S, Vianay B, Roca V, Farrugia AJ, De Pascalis C, Boëda B, Dingli F, Loew D, Vassilopoulos S, Bershadsky A, Théry M, Etienne-Manneville S. Microtubules tune mechanosensitive cell responses. NATURE MATERIALS 2022; 21:366-377. [PMID: 34663953 DOI: 10.1101/2020.07.22.205203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 08/20/2021] [Indexed: 05/24/2023]
Abstract
Mechanotransduction is a process by which cells sense the mechanical properties of their surrounding environment and adapt accordingly to perform cellular functions such as adhesion, migration and differentiation. Integrin-mediated focal adhesions are major sites of mechanotransduction and their connection with the actomyosin network is crucial for mechanosensing as well as for the generation and transmission of forces onto the substrate. Despite having emerged as major regulators of cell adhesion and migration, the contribution of microtubules to mechanotransduction still remains elusive. Here, we show that talin- and actomyosin-dependent mechanosensing of substrate rigidity controls microtubule acetylation (a tubulin post-translational modification) by promoting the recruitment of α-tubulin acetyltransferase 1 (αTAT1) to focal adhesions. Microtubule acetylation tunes the mechanosensitivity of focal adhesions and Yes-associated protein (YAP) translocation. Microtubule acetylation, in turn, promotes the release of the guanine nucleotide exchange factor GEF-H1 from microtubules to activate RhoA, actomyosin contractility and traction forces. Our results reveal a fundamental crosstalk between microtubules and actin in mechanotransduction that contributes to mechanosensitive cell adhesion and migration.
Collapse
Affiliation(s)
- Shailaja Seetharaman
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Université Paris Descartes, Paris, France
| | - Benoit Vianay
- Paris University, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, Paris, France
| | - Vanessa Roca
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Aaron J Farrugia
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Chiara De Pascalis
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Batiste Boëda
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Florent Dingli
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | - Damarys Loew
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | | | - Alexander Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Manuel Théry
- Paris University, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
36
|
Mgharbel A, Migdal C, Bouchonville N, Dupenloup P, Fuard D, Lopez-Soler E, Tomba C, Courçon M, Gulino-Debrac D, Delanoë-Ayari H, Nicolas A. Cells on Hydrogels with Micron-Scaled Stiffness Patterns Demonstrate Local Stiffness Sensing. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:648. [PMID: 35214978 PMCID: PMC8880377 DOI: 10.3390/nano12040648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 12/15/2022]
Abstract
Cell rigidity sensing-a basic cellular process allowing cells to adapt to mechanical cues-involves cell capabilities exerting force on the extracellular environment. In vivo, cells are exposed to multi-scaled heterogeneities in the mechanical properties of the surroundings. Here, we investigate whether cells are able to sense micron-scaled stiffness textures by measuring the forces they transmit to the extracellular matrix. To this end, we propose an efficient photochemistry of polyacrylamide hydrogels to design micron-scale stiffness patterns with kPa/µm gradients. Additionally, we propose an original protocol for the surface coating of adhesion proteins, which allows tuning the surface density from fully coupled to fully independent of the stiffness pattern. This evidences that cells pull on their surroundings by adjusting the level of stress to the micron-scaled stiffness. This conclusion was achieved through improvements in the traction force microscopy technique, e.g., adapting to substrates with a non-uniform stiffness and achieving a submicron resolution thanks to the implementation of a pyramidal optical flow algorithm. These developments provide tools for enhancing the current understanding of the contribution of stiffness alterations in many pathologies, including cancer.
Collapse
Affiliation(s)
- Abbas Mgharbel
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Camille Migdal
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Nicolas Bouchonville
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
| | - Paul Dupenloup
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
| | - David Fuard
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
| | - Eline Lopez-Soler
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Caterina Tomba
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
- University Grenoble Alps, CNRS, Grenoble INP, Institut Néel, 38000 Grenoble, France
| | - Marie Courçon
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Danielle Gulino-Debrac
- University Grenoble Alps, CEA, CNRS, Inserm, BIG-BCI, 38000 Grenoble, France; (M.C.); (D.G.-D.)
| | - Héléne Delanoë-Ayari
- Université de Lyon, University Claude Bernard Lyon1, CNRS, Institut Lumière Matière, 69622 Villeurbanne, France;
| | - Alice Nicolas
- University Grenoble Alps, CNRS, LTM, 38000 Grenoble, France; (A.M.); (C.M.); (N.B.); (P.D.); (D.F.); (E.L.-S.); (C.T.)
| |
Collapse
|
37
|
Yang YA, Nguyen E, Sankara Narayana GHN, Heuzé M, Fu C, Yu H, Mège RM, Ladoux B, Sheetz MP. Local contractions regulate E-cadherin rigidity sensing. SCIENCE ADVANCES 2022; 8:eabk0387. [PMID: 35089785 PMCID: PMC8797795 DOI: 10.1126/sciadv.abk0387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
E-cadherin is a major cell-cell adhesion molecule involved in mechanotransduction at cell-cell contacts in tissues. Because epithelial cells respond to rigidity and tension in tissue through E-cadherin, there must be active processes that test and respond to the mechanical properties of these adhesive contacts. Using submicrometer, E-cadherin-coated polydimethylsiloxane pillars, we find that cells generate local contractions between E-cadherin adhesions and pull to a constant distance for a constant duration, irrespective of pillar rigidity. These cadherin contractions require nonmuscle myosin IIB, tropomyosin 2.1, α-catenin, and binding of vinculin to α-catenin. Cells spread to different areas on soft and rigid surfaces with contractions, but spread equally on soft and rigid without. We further observe that cadherin contractions enable cells to test myosin IIA-mediated tension of neighboring cells and sort out myosin IIA-depleted cells. Thus, we suggest that epithelial cells test and respond to the mechanical characteristics of neighboring cells through cadherin contractions.
Collapse
Affiliation(s)
- Yi-An Yang
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Emmanuelle Nguyen
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | | | - Melina Heuzé
- Université de Paris, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Chaoyu Fu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Hanry Yu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Physiology, Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, Singapore 117593, Singapore
- Institute of Bioengineering and Bioimaging, A*STAR, Singapore 138669, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - René-Marc Mège
- Université de Paris, CNRS, Institut Jacques Monod, F-75013 Paris, France
| | - Benoit Ladoux
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Université de Paris, CNRS, Institut Jacques Monod, F-75013 Paris, France
- Corresponding author. (M.P.S.); (B.L.)
| | - Michael P. Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Corresponding author. (M.P.S.); (B.L.)
| |
Collapse
|
38
|
Cagigas ML, Bryce NS, Ariotti N, Brayford S, Gunning PW, Hardeman EC. Correlative cryo-ET identifies actin/tropomyosin filaments that mediate cell-substrate adhesion in cancer cells and mechanosensitivity of cell proliferation. NATURE MATERIALS 2022; 21:120-128. [PMID: 34518666 DOI: 10.1038/s41563-021-01087-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 07/23/2021] [Indexed: 05/26/2023]
Abstract
The actin cytoskeleton is the primary driver of cellular adhesion and mechanosensing due to its ability to generate force and sense the stiffness of the environment. At the cell's leading edge, severing of the protruding Arp2/3 actin network generates a specific actin/tropomyosin (Tpm) filament population that controls lamellipodial persistence. The interaction between these filaments and adhesion to the environment is unknown. Using cellular cryo-electron tomography we resolve the ultrastructure of the Tpm/actin copolymers and show that they specifically anchor to nascent adhesions and are essential for focal adhesion assembly. Re-expression of Tpm1.8/1.9 in transformed and cancer cells is sufficient to restore cell-substrate adhesions. We demonstrate that knock-out of Tpm1.8/1.9 disrupts the formation of dorsal actin bundles, hindering the recruitment of α-actinin and non-muscle myosin IIa, critical mechanosensors. This loss causes a force-generation and proliferation defect that is notably reversed when cells are grown on soft surfaces. We conclude that Tpm1.8/1.9 suppress the metastatic phenotype, which may explain why transformed cells naturally downregulate this Tpm subset during malignant transformation.
Collapse
Affiliation(s)
- Maria Lastra Cagigas
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Nicole S Bryce
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Nicholas Ariotti
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Electron Microscope Unit, Mark Wainwright Analytical Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Simon Brayford
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Sydney, New South Wales, Australia
| | - Peter W Gunning
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia.
| | - Edna C Hardeman
- School of Medical Sciences, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
39
|
Zhang J, Wong SHD, Wu X, Lei H, Qin M, Shi P, Wang W, Bian L, Cao Y. Engineering Photoresponsive Ligand Tethers for Mechanical Regulation of Stem Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2105765. [PMID: 34561928 DOI: 10.1002/adma.202105765] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/26/2021] [Indexed: 06/13/2023]
Abstract
Regulating stem cell functions by precisely controlling the nanoscale presentation of bioactive ligands has a substantial impact on tissue engineering and regenerative medicine but remains a major challenge. Here it is shown that bioactive ligands can become mechanically "invisible" by increasing their tether lengths to the substrate beyond a critical length, providing a way to regulate mechanotransduction without changing the biochemical conditions. Building on this finding, light switchable tethers are rationally designed, whose lengths can be modulated reversibly by switching a light-responsive protein, pdDronpa, in between monomer and dimer states. This allows the regulation of the adhesion, spreading, and differentiation of stem cells by light on substrates of well-defined biochemical and physical properties. Spatiotemporal regulation of differential cell fates on the same substrate is further demonstrated, which may represent an important step toward constructing complex organoids or mini tissues by spatially defining the mechanical cues of the cellular microenvironment with light.
Collapse
Affiliation(s)
- Junsheng Zhang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Siu Hong Dexter Wong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, 999077, China
| | - Xin Wu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Hai Lei
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| | - Meng Qin
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Peng Shi
- School of Biomedical Sciences and EngineeringSouth China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Liming Bian
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
- School of Biomedical Sciences and EngineeringSouth China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Department of Physics, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
40
|
Fang Y, Hu Y, Cheng F, Xin Y. Biomechanical model of cells probing the myosin-II-independent mechanosensing mechanism. Phys Rev E 2021; 104:064403. [PMID: 35030921 DOI: 10.1103/physreve.104.064403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 11/18/2021] [Indexed: 06/14/2023]
Abstract
Mechanosensing of cells to extracellular matrix (ECM) is highly active and plays a crucial role in various physiological processes. Growing numbers of studies provide evidence that cell sensitivity to ECM stiffness is a complex stress-strain feedback process. However, the mechanisms that rule this process are still not fully known. Here, an alternative mechanosensing scheme of cells, which is different from the previous myosin-II-based mechanisms, is proposed by employing the tension in cortical cytoskeletons (CSKs) as a force module to probe the substrate. The molecular mechanotransduction from cortical CSKs, through actin filaments and focal adhesions, and finally to the substrate, is mechanically modeled to scale the dynamic traction forces of cells. The developed model captures the characteristic spread of cells with respect to ECM stiffness whereby the spread is fully developed on a stiff substrate but not on a soft one. Furthermore, durotactic migration of cells on an elastic-gradient substrate is successfully modeled by the current method. The cells are concluded to migrate, actuated by the polarized traction forces from the stiffness gradient of the substrate and the stiffness matching between cells and substrate. Finally, the cells are proposed to actively target the preferred substrate by following a rule of mechanical matching between cells and substrate. This study provides a theoretical tool to advance our knowledge regarding the passive mechanical properties and the active sensing of cells, and further promotes the discovery of mechanosensing mechanisms as well as the material design for embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Yuqiang Fang
- Department of Mechanics, School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China
| | - Yanbing Hu
- Department of Ultrasound, the Second Hospital Affiliated to Jilin University, Changchun 130021, China
| | - Fei Cheng
- Department of Mechanics, School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China
| | - Yuanzhu Xin
- Department of Mechanics, School of Mechanical and Aerospace Engineering, Jilin University, Changchun 130025, China
| |
Collapse
|
41
|
Abstract
The contraction-relaxation cycle of the heart is one of the most robust mechanical systems in the body that adapts rapidly to the body's needs by changing mechanical parameters. In many respects, we can consider the cardiac system as a complex machine and can use engineering approaches to describe its function. The classical physiology of the heart also focused on understanding function but the new molecular level tools in light microscopy and nanoengineering now enable a deeper understanding of the physiology. The field of mechanobiology has emerged with a focus on how mechanical activity alters biological systems at the molecular level and how those systems in turn control mechanical parameters. In the case of mechanical activity, there are clearly benefits of exercise for the heart, for cancer patients, and for aging but we do not understand the links at a molecular level. Why does regular exercise benefit the heart? We have some preliminary clues at a molecular level about the benefits of physical activity in the cases of cancer and aging; however, there is less known about how exercise affects cardiovascular performance. Unlike the omics approaches which generally link proteins to processes, a mechanobiological understanding of a process explains how forces and mechanical activity will regulate the process through modifications of protein activities. In other words, mechanical activity is an essential component of most biological systems that is transduced into biochemical changes in protein activity. Further, it follows logically that if a mechanical parameter of the cardiac system is typically controlled, then cellular mechanosensing systems must be able to directly or indirectly measure that parameter. The challenge is to understand how changes in activity of the heart are controlled in the short term and then how the system adapts to the integrated level of activity over the longer term. By way of introduction to molecular mechanobiology, I will present examples of mechanosensing from the molecular to the cellular scale and how they may be integrated at the cell and tissue levels. An important element of Mechanobiology at the system level is the physiological state of the cell: i.e., the cell in a senescent state, a cancer state, or a normal cell state (Sheetz 2019). The background for the mechanobiological approach is discussed in "The Cell as a Machine" (Sheetz and Yu, Cambridge Univ Press, 2018), which considers cell states and the molecular systems underlying the important cellular functions. A major challenge in mechanobiology is the understanding of the transduction of mechanical activity into changes in cell function. Of particular relevance here is the benefit of exercise to cardiac performance. This has been seen in many cases and there are a variety of factors that contribute. Further, exercise will benefit cancer patients and will reverse some of the adverse effects of aging. Exercise will cause increased cardiac activity that will be sensed by many mechanosensory systems from a molecular to a cellular level both in the heart and in the vasculature. At a molecular level in cardiac systems, proteins are able to measure stress and strain and to generate appropriate signals of the magnitude of stress and strain that can regulate the cellular contractility and other parameters. The protein sensors are generally passive systems that give a transient measure of local parameters such as the stress at cell-cell junctions during contraction and the strain of the sarcomeres during relaxation. Large stresses at the junctions can activate signaling systems that can reduce contractility or over time activate remodeling of the junctions to better support larger stresses. The proteins involved and their sensory mechanisms are not known currently; however, the mechanosensitive channel, Piezo1, has been implicated in the transduction process in the vasculature (Beech 2018). In the case of strain sensors, large stretches of titin during relaxation can unfold more titin domains that can send signals to the cell. Two different mechanisms of strain sensing are likely in titin. The titin kinase domain is activated by strain but the substrates of the kinase are not know in vivo (Linke 2018). In the backbone of titin are many Ig domains that unfold at different forces and unfolding could cause the binding of proteins that would then activate enzymatic pathways to alter the contractile cycle to give the proper level of strain (Ait-Mou et al. 2017; Granzier et al. 2014; Granzier et al. 2009). The cell-matrix adhesion protein, talin, has eleven cryptic binding sites for another adhesion protein, vinculin, that are revealed by the unfolding of domains in the talin molecule (Yao et al. 2016). Since some domains unfold at lower forces than others, small strains will preferentially unfold those domains, making the system an excellent sensor of the extent of stretch as expected for titin. Because there is an ordered array of many titin molecules, the sensing of strain can be very sensitive to small changes in sarcomere length. Needless to say, titin is only one part of the regulatory system that controls sarcomere length. As one goes more deeply into the working of the system, it is evident that many additional mechanosensory elements are involved in maintaining a functioning cardiac system.
Collapse
|
42
|
Swiatlowska P, Iskratsch T. Tools for studying and modulating (cardiac muscle) cell mechanics and mechanosensing across the scales. Biophys Rev 2021; 13:611-623. [PMID: 34765044 PMCID: PMC8553672 DOI: 10.1007/s12551-021-00837-2] [Citation(s) in RCA: 8] [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: 07/13/2021] [Accepted: 08/24/2021] [Indexed: 12/26/2022] Open
Abstract
Cardiomyocytes generate force for the contraction of the heart to pump blood into the lungs and body. At the same time, they are exquisitely tuned to the mechanical environment and react to e.g. changes in cell and extracellular matrix stiffness or altered stretching due to reduced ejection fraction in heart disease, by adapting their cytoskeleton, force generation and cell mechanics. Both mechanical sensing and cell mechanical adaptations are multiscale processes. Receptor interactions with the extracellular matrix at the nanoscale will lead to clustering of receptors and modification of the cytoskeleton. This in turn alters mechanosensing, force generation, cell and nuclear stiffness and viscoelasticity at the microscale. Further, this affects cell shape, orientation, maturation and tissue integration at the microscale to macroscale. A variety of tools have been developed and adapted to measure cardiomyocyte receptor-ligand interactions and forces or mechanics at the different ranges, resulting in a wealth of new information about cardiomyocyte mechanobiology. Here, we take stock at the different tools for exploring cardiomyocyte mechanosensing and cell mechanics at the different scales from the nanoscale to microscale and macroscale.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| |
Collapse
|
43
|
Amer M, Shi L, Wolfenson H. The 'Yin and Yang' of Cancer Cell Growth and Mechanosensing. Cancers (Basel) 2021; 13:4754. [PMID: 34638240 PMCID: PMC8507527 DOI: 10.3390/cancers13194754] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/07/2021] [Accepted: 09/16/2021] [Indexed: 01/06/2023] Open
Abstract
In cancer, two unique and seemingly contradictory behaviors are evident: on the one hand, tumors are typically stiffer than the tissues in which they grow, and this high stiffness promotes their malignant progression; on the other hand, cancer cells are anchorage-independent-namely, they can survive and grow in soft environments that do not support cell attachment. How can these two features be consolidated? Recent findings on the mechanisms by which cells test the mechanical properties of their environment provide insight into the role of aberrant mechanosensing in cancer progression. In this review article, we focus on the role of high stiffness on cancer progression, with particular emphasis on tumor growth; we discuss the mechanisms of mechanosensing and mechanotransduction, and their dysregulation in cancerous cells; and we propose that a 'yin and yang' type phenomenon exists in the mechanobiology of cancer, whereby a switch in the type of interaction with the extracellular matrix dictates the outcome of the cancer cells.
Collapse
Affiliation(s)
- Malak Amer
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Lidan Shi
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
44
|
Singh A, Tijore A, Margadant F, Simpson C, Chitkara D, Low BC, Sheetz M. Enhanced tumor cell killing by ultrasound after microtubule depolymerization. Bioeng Transl Med 2021; 6:e10233. [PMID: 34589605 PMCID: PMC8459596 DOI: 10.1002/btm2.10233] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 11/24/2022] Open
Abstract
Recent studies show that tumor cells are vulnerable to mechanical stresses and undergo calcium-dependent apoptosis (mechanoptosis) with mechanical perturbation by low-frequency ultrasound alone. To determine if tumor cells are particularly sensitive to mechanical stress in certain phases of the cell cycle, inhibitors of the cell-cycle phases are tested for effects on mechanoptosis. Most inhibitors show no significant effect, but inhibitors of mitosis that cause microtubule depolymerization increase the mechanoptosis. Surprisingly, ultrasound treatment also disrupts microtubules independent of inhibitors in tumor cells but not in normal cells. Ultrasound causes calcium entry through mechanosensitive Piezo1 channels that disrupts microtubules via calpain protease activation. Myosin IIA contractility is required for ultrasound-mediated mechanoptosis and microtubule disruption enhances myosin IIA contractility through activation of GEF-H1 and RhoA pathway. Further, ultrasound promotes contractility-dependent Piezo1 expression and localization to the peripheral adhesions where activated Piezo1 allows calcium entry to continue feedback loop. Thus, the synergistic action of ultrasound and nanomolar concentrations of microtubule depolymerizing agents can enhance tumor therapies.
Collapse
Affiliation(s)
- Aditi Singh
- Mechanobiology InstituteNational University of SingaporeSingapore
- Department of PharmacyBirla Institute of Technology and SciencePilaniIndia
| | - Ajay Tijore
- Mechanobiology InstituteNational University of SingaporeSingapore
| | - Felix Margadant
- Mechanobiology InstituteNational University of SingaporeSingapore
| | - Chloe Simpson
- Mechanobiology InstituteNational University of SingaporeSingapore
| | - Deepak Chitkara
- Department of PharmacyBirla Institute of Technology and SciencePilaniIndia
| | - Boon Chuan Low
- Mechanobiology InstituteNational University of SingaporeSingapore
| | - Michael Sheetz
- Mechanobiology InstituteNational University of SingaporeSingapore
- Biochemistry and Molecular Biology DepartmentUniversity of Texas Medical BranchGalvestonTexasUSA
| |
Collapse
|
45
|
Andreu I, Falcones B, Hurst S, Chahare N, Quiroga X, Le Roux AL, Kechagia Z, Beedle AEM, Elosegui-Artola A, Trepat X, Farré R, Betz T, Almendros I, Roca-Cusachs P. The force loading rate drives cell mechanosensing through both reinforcement and cytoskeletal softening. Nat Commun 2021; 12:4229. [PMID: 34244477 PMCID: PMC8270983 DOI: 10.1038/s41467-021-24383-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/15/2021] [Indexed: 01/08/2023] Open
Abstract
Cell response to force regulates essential processes in health and disease. However, the fundamental mechanical variables that cells sense and respond to remain unclear. Here we show that the rate of force application (loading rate) drives mechanosensing, as predicted by a molecular clutch model. By applying dynamic force regimes to cells through substrate stretching, optical tweezers, and atomic force microscopy, we find that increasing loading rates trigger talin-dependent mechanosensing, leading to adhesion growth and reinforcement, and YAP nuclear localization. However, above a given threshold the actin cytoskeleton softens, decreasing loading rates and preventing reinforcement. By stretching rat lungs in vivo, we show that a similar phenomenon may occur. Our results show that cell sensing of external forces and of passive mechanical parameters (like tissue stiffness) can be understood through the same mechanisms, driven by the properties under force of the mechanosensing molecules involved.
Collapse
Affiliation(s)
- Ion Andreu
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | | | - Sebastian Hurst
- Institute of Cell Biology, Center of Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Nimesh Chahare
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Universitat Politècnica de Catalunya (UPC), Campus Nord, Barcelona, Spain
| | - Xarxa Quiroga
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Anabel-Lise Le Roux
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Zanetta Kechagia
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Amy E M Beedle
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Department of Physics, King's College London, Strand, London, UK
| | - Alberto Elosegui-Artola
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Boston, MA, USA
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Passeig de Lluís Companys, Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Ramon Farré
- Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Timo Betz
- Institute of Cell Biology, Center of Molecular Biology of Inflammation (ZMBE), University of Münster, Münster, Germany
| | - Isaac Almendros
- Universitat de Barcelona, Barcelona, Spain.
- CIBER de Enfermedades Respiratorias, Madrid, Spain.
- Institut d'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain.
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain.
- Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
46
|
Ketebo AA, Park C, Kim J, Jun M, Park S. Probing mechanobiological role of filamin A in migration and invasion of human U87 glioblastoma cells using submicron soft pillars. NANO CONVERGENCE 2021; 8:19. [PMID: 34213679 PMCID: PMC8253861 DOI: 10.1186/s40580-021-00267-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/21/2021] [Indexed: 05/21/2023]
Abstract
Filamin A (FLNa) belongs to an actin-binding protein family in binding and cross-linking actin filaments into a three-dimensional structure. However, little attention has been given to its mechanobiological role in cancer cells. Here, we quantitatively investigated the role of FLNa by analyzing the following parameters in negative control (NC) and FLNa-knockdown (KD) U87 glioma cells using submicron pillars (900 nm diameter and 2 μm height): traction force (TF), rigidity sensing ability, cell aspect ratio, migration speed, and invasiveness. During the initial phase of cell adhesion (< 1 h), FLNa-KD cells polarized more slowly than did NC cells, which can be explained by the loss of rigidity sensing in FLNa-KD cells. The higher motility of FLNa-KD cells relative to NC cells can be explained by the high TF exerted by FLNa-KD cells when compared to NC cells, while the higher invasiveness of FLNa-KD cells relative to NC cells can be explained by a greater number of filopodia in FLNa-KD cells than in NC cells. Our results suggest that FLNa plays important roles in suppressing motility and invasiveness of U87 cells.
Collapse
Affiliation(s)
- Abdurazak Aman Ketebo
- Department of Mechanical Engineering, Sungkyunkwan University (SKKU), 16419, Suwon, Korea
| | - Chanyong Park
- Department of Mechanical Engineering, Sungkyunkwan University (SKKU), 16419, Suwon, Korea
| | - Jaewon Kim
- Department of Mechanical Engineering, Sungkyunkwan University (SKKU), 16419, Suwon, Korea
| | - Myeongjun Jun
- Department of Mechanical Engineering, Sungkyunkwan University (SKKU), 16419, Suwon, Korea
| | - Sungsu Park
- Department of Mechanical Engineering, Sungkyunkwan University (SKKU), 16419, Suwon, Korea.
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), 16419, Suwon, Korea.
- Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), 16419, Suwon, Korea.
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), 2066 Seobu-ro, 16419, Suwon, Korea.
| |
Collapse
|
47
|
Tijore A, Yao M, Wang YH, Hariharan A, Nematbakhsh Y, Lee Doss B, Lim CT, Sheetz M. Selective killing of transformed cells by mechanical stretch. Biomaterials 2021; 275:120866. [PMID: 34044258 DOI: 10.1016/j.biomaterials.2021.120866] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/13/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022]
Abstract
Cancer cells differ from normal cells in several important features like anchorage independence, Warburg effect and mechanosensing. Further, in recent studies, they respond aberrantly to external mechanical distortion. Consistent with altered mechano-responsiveness, we find that cyclic stretching of tumor cells from many different tissues reduces growth rate and causes apoptosis on soft surfaces. Surprisingly, normal cells behave similarly when transformed by depletion of the rigidity sensor protein (Tropomyosin 2.1). Restoration of rigidity sensing in tumor cells promotes rigidity dependent mechanical behavior, i.e. cyclic stretching enhances growth and reduces apoptosis on soft surfaces. The mechanism of mechanical apoptosis (mechanoptosis) of transformed cells involves calcium influx through the mechanosensitive channel, Piezo1 that activates calpain 2 dependent apoptosis through the BAX molecule and subsequent mitochondrial activation of caspase 3 on both fibronetin and collagen matrices. Thus, it is possible to selectively kill tumor cells by mechanical perturbations, while stimulating the growth of normal cells.
Collapse
Affiliation(s)
- Ajay Tijore
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Mingxi Yao
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Yu-Hsiu Wang
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Anushya Hariharan
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Yasaman Nematbakhsh
- Department of Biomedical Engineering, National University of Singapore, 117575, Singapore
| | - Bryant Lee Doss
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, 117411, Singapore; Department of Biomedical Engineering, National University of Singapore, 117575, Singapore; Institute for Health Innovation and Technology, National University of Singapore, 117599, Singapore
| | - Michael Sheetz
- Mechanobiology Institute, National University of Singapore, 117411, Singapore; Molecular Mechanomedicine Program, Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
48
|
Delineating the heterogeneity of matrix-directed differentiation toward soft and stiff tissue lineages via single-cell profiling. Proc Natl Acad Sci U S A 2021; 118:2016322118. [PMID: 33941688 PMCID: PMC8126831 DOI: 10.1073/pnas.2016322118] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The clinical utility of mesenchymal stromal/stem cells (MSCs) in mediating immunosuppressive effects and supporting regenerative processes is broadly established. However, the inherent heterogeneity of MSCs compromises its biomedical efficacy and reproducibility. To study how cellular variation affects fate decision-making processes, we perform single-cell RNA sequencing at multiple time points during bipotential matrix-directed differentiation toward soft- and stiff tissue lineages. In this manner, we identify distinctive MSC subpopulations that are characterized by their multipotent differentiation capacity and mechanosensitivity. Also, whole-genome screening highlights TPM1 as a potent mechanotransducer of matrix signals and regulator of cell differentiation. Thus, by introducing single-cell methodologies into mechanobiology, we delineate the complexity of adult stem cell responses to extracellular cues in tissue regeneration and immunomodulation. Mesenchymal stromal/stem cells (MSCs) form a heterogeneous population of multipotent progenitors that contribute to tissue regeneration and homeostasis. MSCs assess extracellular elasticity by probing resistance to applied forces via adhesion, cytoskeletal, and nuclear mechanotransducers that direct differentiation toward soft or stiff tissue lineages. Even under controlled culture conditions, MSC differentiation exhibits substantial cell-to-cell variation that remains poorly characterized. By single-cell transcriptional profiling of nonconditioned, matrix-conditioned, and early differentiating cells, we identified distinct MSC subpopulations with distinct mechanosensitivities, differentiation capacities, and cell cycling. We show that soft matrices support adipogenesis of multipotent cells and early endochondral ossification of nonadipogenic cells, whereas intramembranous ossification and preosteoblast proliferation are directed by stiff matrices. Using diffusion pseudotime mapping, we outline hierarchical matrix-directed differentiation and perform whole-genome screening of mechanoresponsive genes. Specifically, top-ranked tropomyosin-1 is highly sensitive to stiffness cues both at RNA and protein levels, and changes in TPM1 expression determine the differentiation toward soft versus stiff tissue lineage. Consistent with actin stress fiber stabilization, tropomyosin-1 overexpression maintains YAP1 nuclear localization, activates YAP1 target genes, and directs osteogenic differentiation. Knockdown of tropomyosin-1 reversed YAP1 nuclear localization consistent with relaxation of cellular contractility, suppressed osteogenesis, activated early endochondral ossification genes after 3 d of culture in induction medium, and facilitated adipogenic differentiation after 1 wk. Our results delineate cell-to-cell variation of matrix-directed MSC differentiation and highlight tropomyosin-mediated matrix sensing.
Collapse
|
49
|
Lavrenyuk K, Conway D, Dahl KN. Imaging methods in mechanosensing: a historical perspective and visions for the future. Mol Biol Cell 2021; 32:842-854. [PMID: 33788578 PMCID: PMC8108522 DOI: 10.1091/mbc.e20-10-0671] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Over the past three decades, as mechanobiology has become a distinct area of study, researchers have developed novel imaging tools to discover the pathways of biomechanical signaling. Early work with substrate engineering and particle tracking demonstrated the importance of cell–extracellular matrix interactions on the cell cycle as well as the mechanical flux of the intracellular environment. Most recently, tension sensor approaches allowed directly measuring tension in cell–cell and cell–substrate interactions. We retrospectively analyze how these various optical techniques progressed the field and suggest our vision forward for a unified theory of cell mechanics, mapping cellular mechanosensing, and novel biomedical applications for mechanobiology.
Collapse
Affiliation(s)
- Kirill Lavrenyuk
- Carnegie Mellon University, College of Engineering, Pittsburgh, PA 15213
| | - Daniel Conway
- Virginia Commonwealth University, College of Engineering, Richmond, VA 23284
| | - Kris Noel Dahl
- Carnegie Mellon University, College of Engineering, Pittsburgh, PA 15213
| |
Collapse
|
50
|
Riehl BD, Kim E, Bouzid T, Lim JY. The Role of Microenvironmental Cues and Mechanical Loading Milieus in Breast Cancer Cell Progression and Metastasis. Front Bioeng Biotechnol 2021; 8:608526. [PMID: 33585411 PMCID: PMC7874074 DOI: 10.3389/fbioe.2020.608526] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/22/2020] [Indexed: 01/08/2023] Open
Abstract
Cancer can disrupt the microenvironments and mechanical homeostatic actions in multiple scales from large tissue modification to altered cellular signaling pathway in mechanotransduction. In this review, we highlight recent progresses in breast cancer cell mechanobiology focusing on cell-microenvironment interaction and mechanical loading regulation of cells. First, the effects of microenvironmental cues on breast cancer cell progression and metastasis will be reviewed with respect to substrate stiffness, chemical/topographic substrate patterning, and 2D vs. 3D cultures. Then, the role of mechanical loading situations such as tensile stretch, compression, and flow-induced shear will be discussed in relation to breast cancer cell mechanobiology and metastasis prevention. Ultimately, the substrate microenvironment and mechanical signal will work together to control cancer cell progression and metastasis. The discussions on breast cancer cell responsiveness to mechanical signals, from static substrate and dynamic loading, and the mechanotransduction pathways involved will facilitate interdisciplinary knowledge transfer, enabling further insights into prognostic markers, mechanically mediated metastasis pathways for therapeutic targets, and model systems required to advance cancer mechanobiology.
Collapse
Affiliation(s)
- Brandon D Riehl
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Eunju Kim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Tasneem Bouzid
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|