1
|
Tidei JJ, Oakes PW, Beach JR. Myosin 2 - A general contractor for the cytoskeleton. Curr Opin Cell Biol 2025; 94:102522. [PMID: 40319507 DOI: 10.1016/j.ceb.2025.102522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/07/2025]
Abstract
Cells derive their shape, and in turn much of their behavior, from the organization of the cytoskeleton. While a myriad of proteins contribute to the regulation and organization of this dynamic structure, two of the principal components are actin filaments, which provide the structure, and myosin motors, which generate the majority of the forces. Here we review recent results on the assembly and kinetics of non-muscle myosin 2, and highlight how the cellular environment modulates local myosin behavior and signaling.
Collapse
Affiliation(s)
- Joseph J Tidei
- Dept. Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA
| | - Patrick W Oakes
- Dept. Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA.
| | - Jordan R Beach
- Dept. Cell & Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL 60153, USA.
| |
Collapse
|
2
|
Suzuki M, Kawauchi K, Machiyama H, Hirata H, Ishiwata S, Fujita H. Dynamic Remodeling of Mechano-Sensing Complexes in Suspended Fibroblast Cell-Sheets Under External Mechanical Stimulus. Biotechnol Bioeng 2025. [PMID: 40270085 DOI: 10.1002/bit.28996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/25/2025]
Abstract
Freestanding cell-sheets are valuable bio-materials for use in regenerative medicine and tissue engineering. Because cell-sheets experience various mechanical stimulations during handling, it is important to understand the responses of cells to these stimulations. Here, we demonstrate changes in the localization of various proteins during the stretching of fibroblast cell-sheets. These proteins are known to be involved in mechano-sensing. Upon stretching, actin filaments appear parallel to the stretching direction. At cell-cell junctions, β-catenin forms clusters that co-localize with accumulated vinculin and zyxin as well as the actin filaments. The p130 Crk-associated substrate, known to be present in focal adhesions, is also recruited to these clusters and phosphorylated. Our results suggest that mechano-sensing machinery is formed at cell-cell junctions when the cell-sheets are stretched.
Collapse
Affiliation(s)
- Madoka Suzuki
- Institute for Protein Research, The University of Osaka, Suita, Osaka, Japan
| | - Keiko Kawauchi
- Faculty of Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Hyogo, Japan
| | - Hiroaki Machiyama
- Department of Immunology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Hiroaki Hirata
- Department of Life Science and Biotechnology, Kanazawa Institute of Technology, Hakusan, Ishikawa, Japan
| | - Shin'ichi Ishiwata
- Department of Physics, Faculty of Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Hideaki Fujita
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Hiroshima, Japan
| |
Collapse
|
3
|
Phua DYZ, Sun X, Alushin GM. Force-activated zyxin assemblies coordinate actin nucleation and crosslinking to orchestrate stress fiber repair. Curr Biol 2025; 35:854-870.e9. [PMID: 39952249 PMCID: PMC11867737 DOI: 10.1016/j.cub.2025.01.042] [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: 06/10/2024] [Revised: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/17/2025]
Abstract
As the cytoskeleton sustains cell and tissue forces, it incurs physical damage that must be repaired to maintain mechanical homeostasis. The LIN-11, Isl-1, and Mec-3 (LIM)-domain protein zyxin detects force-induced ruptures in actin-myosin stress fibers, coordinating downstream repair factors to restore stress fiber integrity through unclear mechanisms. Here, we reconstitute stress fiber repair with purified proteins, uncovering detailed links between zyxin's force-regulated binding interactions and cytoskeletal dynamics. In addition to binding individual tensed actin filaments (F-actin), zyxin's LIM domains form force-dependent assemblies that bridge broken filament fragments. Zyxin assemblies engage repair factors through multivalent interactions, coordinating nucleation of new F-actin by VASP and its crosslinking into aligned bundles by ɑ-actinin. Through these combined activities, stress fiber repair initiates within the cores of micron-scale damage sites in cells, explaining how these F-actin-depleted regions are rapidly restored. Thus, zyxin's force-dependent organization of actin repair machinery inherently operates at the network scale to maintain cytoskeletal integrity.
Collapse
Affiliation(s)
- Donovan Y Z Phua
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA.
| | - Xiaoyu Sun
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA
| | - Gregory M Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
4
|
Oakes PW. Cytoskeletal repair: Zyxin relieves actin stress from the inside out. Curr Biol 2025; 35:R148-R150. [PMID: 39999785 DOI: 10.1016/j.cub.2024.12.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
During the dynamic reorganization of the actin cytoskeleton, filaments occasionally break. To maintain the mechanical integrity of the actin structure, the cell must swiftly repair strained filaments in situ. New work recapitulates this process using purified proteins to reveal the underlying molecular mechanisms that enable this robust and rapid recovery.
Collapse
Affiliation(s)
- Patrick W Oakes
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA.
| |
Collapse
|
5
|
Meijlink B, van der Kooij HR, Wang Y, Li H, Huveneers S, Kooiman K. Ultrasound-activated microbubbles mediate F-actin disruptions and endothelial gap formation during sonoporation. J Control Release 2024; 376:1176-1189. [PMID: 39500409 DOI: 10.1016/j.jconrel.2024.10.066] [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: 10/07/2024] [Accepted: 10/29/2024] [Indexed: 11/14/2024]
Abstract
Locally opening up the endothelial barrier in a safe and controlled way is beneficial for drug delivery into the extravascular tissue. Although ultrasound-induced microbubble oscillations can affect the endothelial barrier integrity, the mechanism remains unknown. Here we uncover a new role for F-actin in microbubble-mediated endothelial gap formation. Unique simultaneous high-resolution confocal microscopy and ultra-high-speed camera imaging (10 million frames per second) reveal that single oscillating microbubbles (radius 1.3-3.8 μm; n = 48) induce sonoporation in all cells in which F-actin remodeling occurred. F-actin disruption only mainly resulted in tunnel formation (75 %), while F-actin stress fiber severing and recoil mainly resulted in cell-cell contact opening within 15 s upon treatment (54 %) and tunnel formation (15 %). F-actin stress fiber severing occurred when the fibers were within reach of the microbubble's maximum radius during oscillation, requiring normal forces of ≥230 nN. In the absence of F-actin stress fibers, oscillating microbubbles induced F-actin remodeling but no cell-cell contact opening. Together, these findings reveal a novel mechanism of microbubble-mediated transendothelial drug delivery, which associates with the underlying cytoskeletal F-actin organization.
Collapse
Affiliation(s)
- Bram Meijlink
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - H Rhodé van der Kooij
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Yuchen Wang
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Hongchen Li
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands
| | - Stephan Huveneers
- Dept. Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Klazina Kooiman
- Biomedical Engineering, Dept. of Cardiology, Cardiovascular Institute, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
6
|
Burgess JK, Gosens R. Mechanotransduction and the extracellular matrix: Key drivers of lung pathologies and drug responsiveness. Biochem Pharmacol 2024; 228:116255. [PMID: 38705536 DOI: 10.1016/j.bcp.2024.116255] [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: 02/02/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
The lung is a biomechanically active organ, with multiscale mechanical forces impacting the organ, tissue and cellular responses within this microenvironment. In chronic lung diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and others, the structure of the lung is drastically altered impeding gas exchange. These changes are, in part, reflected in alterations in the composition, amount and organization of the extracellular matrix within the different lung compartments. The transmission of mechanical forces within lung tissue are broadcast by this complex mix of extracellular matrix components, in particular the collagens, elastin and proteoglycans and the crosslinking of these components. At both a macro and a micro level, the mechanical properties of the microenvironment have a key regulatory role in ascertaining cellular responses and the function of the lung. Cells adhere to, and receive signals from, the extracellular matrix through a number of different surface receptors and complexes which are important for mechanotransduction. This review summarizes the multiscale mechanics in the lung and how the mechanical environment changes in lung disease and aging. We then examine the role of mechanotransduction in driving cell signaling events in lung diseases and finish with a future perspective of the need to consider how such forces may impact pharmacological responsiveness in lung diseases.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| | - Reinoud Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
7
|
Schwarz US. Cracking under stress: How actin might turn failure into action. Biophys J 2024; 123:3281-3282. [PMID: 39244639 PMCID: PMC11480752 DOI: 10.1016/j.bpj.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Affiliation(s)
- Ulrich S Schwarz
- Institute for Theoretical Physics and BioQuant, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
8
|
Saporito S, Panzetta V, Netti PA. Time and space modulation of substrate curvature to regulate cell mechanical identity. Acta Biomater 2024; 186:300-315. [PMID: 39127326 DOI: 10.1016/j.actbio.2024.08.006] [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: 03/27/2024] [Revised: 07/08/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Recently, a variety of microenvironmental biophysical stimuli have been proved to play a crucial role in regulating cell functions. Among them, morpho-physical cues, like curvature, are emerging as key regulators of cellular behavior. Changes in substrate curvature have been shown to impact the arrangement of Focal Adhesions (FAs), influencing the direction and intensity of cytoskeleton generated forces and resulting in an overall alteration of cell mechanical identity. In their native environment, cells encounter varying degrees of substrate curvature, and in specific organs, they are exposed to dynamic changes of curvature due to periodic tissue deformation. However, the mechanism by which cells perceive substrate curvature remains poorly understood. To this aim, a micro-pneumatic device was designed and implemented. This device enables the controlled application of substrate curvature, both statically and dynamically. Employing a combined experimental and simulative approach, human adipose-derived stem cells were exposed to controlled curvature intensity and frequency. During this exposure, measurements were taken on FAs extension and orientation, cytoskeleton organization and cellular/nuclear alignment. The data clearly indicated a significant influence of the substrate curvature on cell adhesion processes. These findings contribute to a better understanding of the mechanisms through which cells perceive and respond to substrate curvature signals. STATEMENT OF SIGNIFICANCE: This work is our contribution to the comprehension of substrate curvature's function as a crucial regulator of cell adhesion at the scale of focal adhesions and cell mechanical identity. In recent years, a large body of knowledge is continuously growing providing comprehension of the role of various microenvironmental biophysical stimuli in regulating cell functions. Nevertheless, little is known about the role of substrate curvature, in particular, when cells are exposed to this stimulus in a dynamic manner. To address the role of substrate curvature on cellular behavior, a micro-pneumatic device was designed and implemented. This device enables the controlled application of substrate curvature, both statically and dynamically. The experiment data made it abundantly evident that the substrate curvature had a major impact on the mechanisms involved in cell adhesion.
Collapse
Affiliation(s)
- Stefania Saporito
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy
| | - Valeria Panzetta
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy; Interdisciplinary research Center on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy; Interdisciplinary research Center on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy.
| |
Collapse
|
9
|
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
|
10
|
Dutta S, Muraganadan T, Vasudevan M. Evaluation of lamin A/C mechanotransduction under different surface topography in LMNA related muscular dystrophy. Cytoskeleton (Hoboken) 2024. [PMID: 39091017 DOI: 10.1002/cm.21895] [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: 10/16/2023] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024]
Abstract
Most of the single point mutations of the LMNA gene are associated with distinct muscular dystrophies, marked by heterogenous phenotypes but primarily the loss and symmetric weakness of skeletal muscle tissue. The molecular mechanism and phenotype-genotype relationships in these muscular dystrophies are poorly understood. An effort has been here to delineating the adaptation of mechanical inputs into biological response by mutant cells of lamin A associated muscular dystrophy. In this study, we implement engineered smooth and pattern surfaces of particular young modulus to mimic muscle physiological range. Using fluorescence and atomic force microscopy, we present distinct architecture of the actin filament along with abnormally distorted cell and nuclear shape in mutants, which showed a tendency to deviate from wild type cells. Topographic features of pattern surface antagonize the binding of the cell with it. Correspondingly, from the analysis of genome wide expression data in wild type and mutant cells, we report differential expression of the gene products of the structural components of cell adhesion as well as LINC (linkers of nucleoskeleton and cytoskeleton) protein complexes. This study also reveals mis expressed downstream signaling processes in mutant cells, which could potentially lead to onset of the disease upon the application of engineered materials to substitute the role of conventional cues in instilling cellular behaviors in muscular dystrophies. Collectively, these data support the notion that lamin A is essential for proper cellular mechanotransduction from extracellular environment to the genome and impairment of the muscle cell differentiation in the pathogenic mechanism for lamin A associated muscular dystrophy.
Collapse
Affiliation(s)
- Subarna Dutta
- Department of Biochemistry, University of Calcutta, Kolkata, West Bengal, India
- Theomics International Private Limited, Bengaluru, India
| | - T Muraganadan
- CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | | |
Collapse
|
11
|
Kim DS, Cheah JS, Lai TW, Zhao KX, Foust SR, Julie Lee YR, Lo SH, Heinrich V, Yamada S. Tandem LIM domain-containing proteins, LIMK1 and LMO1, directly bind to force-bearing keratin intermediate filaments. Cell Rep 2024; 43:114480. [PMID: 39003737 PMCID: PMC11610427 DOI: 10.1016/j.celrep.2024.114480] [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: 12/18/2023] [Revised: 05/10/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
The cytoskeleton of the cell is constantly exposed to physical forces that regulate cellular functions. Selected members of the LIM (Lin-11, Isl-1, and Mec-3) domain-containing protein family accumulate along force-bearing actin fibers, with evidence supporting that the LIM domain is solely responsible for this force-induced interaction. However, LIM domain's force-induced interactions are not limited to actin. LIMK1 and LMO1, both containing only two tandem LIM domains, are recruited to force-bearing keratin fibers in epithelial cells. This unique recruitment is mediated by their LIM domains and regulated by the sequences outside the LIM domains. Based on in vitro reconstitution of this interaction, LIMK1 and LMO1 directly interact with stretched keratin 8/18 fibers. These results show that LIM domain's mechano-sensing abilities extend to the keratin cytoskeleton, highlighting the diverse role of LIM proteins in force-regulated signaling.
Collapse
Affiliation(s)
- Dah Som Kim
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Joleen S Cheah
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Tzu Wei Lai
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Karen X Zhao
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Skylar R Foust
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Yuh-Ru Julie Lee
- Department of Plant Biology, University of California, Davis, Davis CA 95616, USA
| | - Su Hao Lo
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis CA 95616 USA
| | - Volkmar Heinrich
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA
| | - Soichiro Yamada
- Biomedical Engineering Department, University of California, Davis, Davis CA 95616, USA.
| |
Collapse
|
12
|
Phua DY, Sun X, Alushin GM. Force-activated zyxin assemblies coordinate actin nucleation and crosslinking to orchestrate stress fiber repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.17.594765. [PMID: 38798419 PMCID: PMC11118565 DOI: 10.1101/2024.05.17.594765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
As the cytoskeleton sustains cell and tissue forces, it incurs physical damage that must be repaired to maintain mechanical homeostasis. The LIM-domain protein zyxin detects force-induced ruptures in actin-myosin stress fibers, coordinating downstream repair factors to restore stress fiber integrity through unclear mechanisms. Here, we reconstitute stress fiber repair with purified proteins, uncovering detailed links between zyxin's force-regulated binding interactions and cytoskeletal dynamics. In addition to binding individual tensed actin filaments (F-actin), zyxin's LIM domains form force-dependent assemblies that bridge broken filament fragments. Zyxin assemblies engage repair factors through multi-valent interactions, coordinating nucleation of new F-actin by VASP and its crosslinking into aligned bundles by ɑ-actinin. Through these combined activities, stress fiber repair initiates within the cores of micron-scale damage sites in cells, explaining how these F-actin depleted regions are rapidly restored. Thus, zyxin's force-dependent organization of actin repair machinery inherently operates at the network scale to maintain cytoskeletal integrity.
Collapse
Affiliation(s)
- Donovan Y.Z. Phua
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Xiaoyu Sun
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| | - Gregory M. Alushin
- Laboratory of Structural Biophysics and Mechanobiology, The Rockefeller University, New York, NY, USA
| |
Collapse
|
13
|
Ebrahimighaei R, Tarassova N, Bond SC, McNeill MC, Hathway T, Vohra H, Newby AC, Bond M. Extracellular matrix stiffness controls cardiac fibroblast proliferation via the nuclear factor-Y (NF-Y) transcription factor. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119640. [PMID: 37996060 DOI: 10.1016/j.bbamcr.2023.119640] [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: 08/31/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023]
Abstract
The proliferative expansion of cardiac fibroblasts (CF) contributes towards cardiac fibrosis, which results in myocardial stiffening, cardiac dysfunction, and heart failure. CF sense and respond to increased stiffness of their local extracellular matrix, modulating their phenotype towards increased collagen synthesis and higher proliferation, leading potentially to a vicious circle of positive feedback. Here we describe a novel mechanism that mediates increased CF proliferation in response to a pathologically stiff Exteracellular matrix (ECM). The mechanism we describe is independent of the well-characterised mechano-sensitive transcript factors, YAP-TEAD and MKL1-SRF, which our data indicate are only responsible for part of the genes induced by stiffened ECM. Instead, our data identify Nuclear Factor-Y (NF-Y) as a novel mechanosensitive transcription factor, which mediates enhanced CF proliferation in response to a stiff ECM. We show that levels of NF-YA protein, the major regulatory subunit of NF-Y, and NF-Y transcriptional activity, are increased by a stiff ECM. Indeed, NF-Y activity drives the expression of multiple cell-cycle genes. Furthermore, NF-YA protein levels are dependent on FAK signalling suggesting a mechanistic link to ECM composition. Consistent with its role as a mechano-sensor, inhibition of NF-Y using siRNA or dominant negative mutant blocks CF proliferation on plastic in vitro, which models a stiff ECM, whereas ectopic expression of NF-YA increases the proliferation of cells interacting under conditions that model a physiologically soft ECM. In summary, our data demonstrate that NF-Y is a biomechanically sensitive transcription factor that promotes CF proliferation in a model of pathologically stiffened ECM.
Collapse
Affiliation(s)
- Reza Ebrahimighaei
- Department of Translational Health Sciences, Bristol Medical School, Bristol, BS2 8HW, United Kingdom
| | - Nathalie Tarassova
- Department of Translational Health Sciences, Bristol Medical School, Bristol, BS2 8HW, United Kingdom.
| | - Samuel C Bond
- Clifton High School, Clifton, Bristol, BS8 3JD, United Kingdom.
| | - Madeleine C McNeill
- Department of Translational Health Sciences, Bristol Medical School, Bristol, BS2 8HW, United Kingdom.
| | - Tom Hathway
- Department of Translational Health Sciences, Bristol Medical School, Bristol, BS2 8HW, United Kingdom.
| | - Hunaid Vohra
- Department of Translational Health Sciences, Bristol Medical School, Bristol, BS2 8HW, United Kingdom.
| | - Andrew C Newby
- Department of Translational Health Sciences, Bristol Medical School, Bristol, BS2 8HW, United Kingdom.
| | - Mark Bond
- Department of Translational Health Sciences, Bristol Medical School, Bristol, BS2 8HW, United Kingdom.
| |
Collapse
|
14
|
Alonso A, Ebben A, Dabagh M. Impact of disturbed flow and arterial stiffening on mechanotransduction in endothelial cells. Biomech Model Mechanobiol 2023; 22:1919-1933. [PMID: 37709992 DOI: 10.1007/s10237-023-01743-0] [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: 02/28/2023] [Accepted: 07/05/2023] [Indexed: 09/16/2023]
Abstract
Disturbed flow promotes progression of atherosclerosis at particular regions of arteries where the recent studies show the arterial wall becomes stiffer. Objective of this study is to show how mechanotransduction in subcellular organelles of endothelial cells (ECs) will alter with changes in blood flow profiles applied on ECs surface and mechanical properties of arterial wall where ECs are attached to. We will examine the exposure of ECs to atherogenic flow profiles (disturbed flow) and non-atherogenic flow profiles (purely forward flow), while stiffness and viscoelasticity of arterial wall will change. A multicomponent model of endothelial cell monolayer was applied to quantify the response of subcellular organelles to the changes in their microenvironment. Our results show that arterial stiffening alters mechanotransduction in intra/inter-cellular organelles of ECs by slight increase in the transmitted stresses, particularly over central stress fibers (SFs). We also observed that degradation of glycocalyx and exposure to non-atherogenic flow profiles result in significantly higher stresses in subcellular organelles, while degradation of glycocalyx and exposure to atherogenic flow profiles result in dramatically lower stresses in the organelles. Moreover, we show that increasing the arterial wall viscoelasticity leads to slight increase in the stresses transmitted to subcellular organelles. FAs are particularly influenced with the changes in the arterial wall properties and viscoelasticity. Our study suggests that changes in viscoelasticity of arterial wall and degradation state of glycocalyx have to be considered along with arterial stiffening in designing more efficient treatment strategies for atherosclerosis. Our study provides insight into significant role of mechanotransduction in the localization of atherosclerosis by quantifying the role of ECs mechanosensors and suggests that mechanotransduction may play a key role in design of more efficient and precision therapeutics to slow down or block the progression of atherosclerosis.
Collapse
Affiliation(s)
- Andrea Alonso
- Department of Biomedical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Alessandra Ebben
- Department of Biomedical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Mahsa Dabagh
- Department of Biomedical Engineering, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA.
| |
Collapse
|
15
|
Colin A, Orhant-Prioux M, Guérin C, Savinov M, Cao W, Vianay B, Scarfone I, Roux A, De La Cruz EM, Mogilner A, Théry M, Blanchoin L. Friction patterns guide actin network contraction. Proc Natl Acad Sci U S A 2023; 120:e2300416120. [PMID: 37725653 PMCID: PMC10523593 DOI: 10.1073/pnas.2300416120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 08/09/2023] [Indexed: 09/21/2023] Open
Abstract
The shape of cells is the outcome of the balance of inner forces produced by the actomyosin network and the resistive forces produced by cell adhesion to their environment. The specific contributions of contractile, anchoring and friction forces to network deformation rate and orientation are difficult to disentangle in living cells where they influence each other. Here, we reconstituted contractile actomyosin networks in vitro to study specifically the role of the friction forces between the network and its anchoring substrate. To modulate the magnitude and spatial distribution of friction forces, we used glass or lipids surface micropatterning to control the initial shape of the network. We adapted the concentration of Nucleating Promoting Factor on each surface to induce the assembly of actin networks of similar densities and compare the deformation of the network toward the centroid of the pattern shape upon myosin-induced contraction. We found that actin network deformation was faster and more coordinated on lipid bilayers than on glass, showing the resistance of friction to network contraction. To further study the role of the spatial distribution of these friction forces, we designed heterogeneous micropatterns made of glass and lipids. The deformation upon contraction was no longer symmetric but biased toward the region of higher friction. Furthermore, we showed that the pattern of friction could robustly drive network contraction and dominate the contribution of asymmetric distributions of myosins. Therefore, we demonstrate that during contraction, both the active and resistive forces are essential to direct the actin network deformation.
Collapse
Affiliation(s)
- Alexandra Colin
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Magali Orhant-Prioux
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Christophe Guérin
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Mariya Savinov
- Courant Institute of Mathematical Sciences, New York University, New York, NY10012
| | - Wenxiang Cao
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT06520-8114
| | - Benoit Vianay
- University of Paris, INSERM, Commissariat à l'énergie atomique et aux énergies alternatives, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, Paris75010, France
| | - Ilaria Scarfone
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
| | - Aurélien Roux
- Department of Biochemistry, University of Geneva, CH-1211Geneva, Switzerland
| | - Enrique M. De La Cruz
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT06520-8114
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, New York, NY10012
| | - Manuel Théry
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
- University of Paris, INSERM, Commissariat à l'énergie atomique et aux énergies alternatives, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, Paris75010, France
| | - Laurent Blanchoin
- Université Grenoble-Alpes, CEA, CNRS, UMR5168, Interdisciplinary Research Institute of Grenoble, CytoMorpho Lab, Grenoble38054, France
- University of Paris, INSERM, Commissariat à l'énergie atomique et aux énergies alternatives, UMRS1160, Institut de Recherche Saint Louis, CytoMorpho Lab, Hôpital Saint Louis, Paris75010, France
| |
Collapse
|
16
|
Segretain D, Di Marco M, Dufeu C, Carette D, Trubuil A, Pointis G. Cooperative cell-cell actin network remodeling to perform Gap junction endocytosis. Basic Clin Androl 2023; 33:20. [PMID: 37533006 PMCID: PMC10399049 DOI: 10.1186/s12610-023-00194-y] [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: 12/19/2022] [Accepted: 04/16/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND The endocytosis of Gap junction plaques (GJP) requires cytoskeletal forces to internalize such large membranous structures. Actin, which partners the connexin proteins constituting Gap junctions and is located close to Annular Gap Junctions (AGJ), could be actively involved in this physiological process. RESULTS Electron Microscopy and Light Microscopy images, associated with time-lapse analysis and 3D reconstruction, used at high resolution and enhanced using ImageJ based software analysis, revealed that: i) actin cables, originating from Donor cells, insert on the edge of GJP and contribute to their invagination, giving rise to AGJ, whereas actin cables on the Acceptor cell side of the plaque are not modified; ii) actin cables from the Donor cell are continuous with the actin network present over the entire GJP surface. These actin cables fuse at a single point distant from the plaque, which then detaches itself from the membrane, condensing to form an actin mass during the final internalization process; iii) the Acceptor cell participates in the last step of the endocytic invagination process by forming an annular actin structure known as an actin ring. CONCLUSIONS Together, these data suggest that the endocytosis of GJP is an example of a unique cooperative mechanism between the Donor (the traction of its actin cables) and the Acceptor cells (forming the actin ring).
Collapse
Affiliation(s)
- Dominique Segretain
- UMR S1147, Université Paris Descartes, 45 Rue Des Saints-Pères, 75006, Paris, France.
| | - Mathilde Di Marco
- UMR S1147, Université Paris Descartes, 45 Rue Des Saints-Pères, 75006, Paris, France
- Present Address: Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, 75005, Paris, France
| | - Chloé Dufeu
- Faculté de Pharmacie, Université Paris-Saclay, Saclay, France
| | | | - Alain Trubuil
- MaIAGE, INRAE, Université Paris-Saclay, 78352, Jouy-en-Josas, France
| | - Georges Pointis
- INSERM U 1065, Team 5 Physiopathological Control of Germ Cell Proliferation: Genomic and Non-Genomic Mechanisms, University of Nice Sophia-Antipolis, 151 Route Saint-Antoine de Ginestière BP 2 3194, 06204, Nice Cedex 3, France
| |
Collapse
|
17
|
Amatu JB, Baudouin C, Trinh L, Labbé A, Buffault J. [Corneal epithelial biomechanics: Resistance to stress and role in healing and remodeling]. J Fr Ophtalmol 2023; 46:287-299. [PMID: 36759249 DOI: 10.1016/j.jfo.2022.09.026] [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: 09/04/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 02/10/2023]
Abstract
The corneal epithelium is one of the first tissue barriers of the eye against the environment. In recent years, many studies provided better knowledge of its healing, its behavior and its essential role in the optical system of the eye. At the crossroads of basic science and clinical medicine, the study of the mechanical stresses applied to the cornea makes it possible to learn the behavior of epithelial cells and better understand ocular surface disease. We describe herein the current knowledge about the adhesion systems of the corneal epithelium and their resistance to mechanical stress. We will also describe the involvement of these mechanisms in corneal healing and their role in epithelial dynamics. Adhesion molecules of the epithelial cells, especially hemidesmosomes, allow the tissue cohesion required to maintain the integrity of the corneal epithelium against the shearing forces of the eyelids as well as external forces. Their regeneration after a corneal injury is mandatory for the restoration of a healthy epithelium. Mechanotransduction plays a significant role in regulating epithelial cell behavior, and the study of the epithelium's response to mechanical forces helps to better understand the evolution of epithelial profiles after refractive surgery. A better understanding of corneal epithelial biomechanics could also help improve future therapies, particularly in the field of tissue engineering.
Collapse
Affiliation(s)
- J-B Amatu
- Department of Ophthalmology III, CHNO des Quinze-Vingts, IHU FOReSIGHT, 28, rue de Charenton, 75012 Paris, France.
| | - C Baudouin
- Department of Ophthalmology III, CHNO des Quinze-Vingts, IHU FOReSIGHT, 28, rue de Charenton, 75012 Paris, France; Institut de La Vision, Sorbonne Université, Inserm, CNRS, IHU FOReSIGHT, 17, rue Moreau, 75012 Paris, France; Department of Ophthalmology, Ambroise Paré Hospital, AP-HP, University of Versailles Saint-Quentin-en-Yvelines, Boulogne-Billancourt, France
| | - L Trinh
- Department of Ophthalmology III, CHNO des Quinze-Vingts, IHU FOReSIGHT, 28, rue de Charenton, 75012 Paris, France
| | - A Labbé
- Department of Ophthalmology III, CHNO des Quinze-Vingts, IHU FOReSIGHT, 28, rue de Charenton, 75012 Paris, France; Institut de La Vision, Sorbonne Université, Inserm, CNRS, IHU FOReSIGHT, 17, rue Moreau, 75012 Paris, France; Department of Ophthalmology, Ambroise Paré Hospital, AP-HP, University of Versailles Saint-Quentin-en-Yvelines, Boulogne-Billancourt, France
| | - J Buffault
- Department of Ophthalmology III, CHNO des Quinze-Vingts, IHU FOReSIGHT, 28, rue de Charenton, 75012 Paris, France; Institut de La Vision, Sorbonne Université, Inserm, CNRS, IHU FOReSIGHT, 17, rue Moreau, 75012 Paris, France; Department of Ophthalmology, Ambroise Paré Hospital, AP-HP, University of Versailles Saint-Quentin-en-Yvelines, Boulogne-Billancourt, France
| |
Collapse
|
18
|
Hakeem RM, Subramanian BC, Hockenberry MA, King ZT, Butler MT, Legant WR, Bear JE. A Photopolymerized Hydrogel System with Dual Stiffness Gradients Reveals Distinct Actomyosin-Based Mechano-Responses in Fibroblast Durotaxis. ACS NANO 2023; 17:197-211. [PMID: 36475639 PMCID: PMC9839609 DOI: 10.1021/acsnano.2c05941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Durotaxis, migration of cells directed by a stiffness gradient, is critical in development and disease. To distinguish durotaxis-specific migration mechanisms from those on uniform substrate stiffnesses, we engineered an all-in-one photopolymerized hydrogel system containing areas of stiffness gradients with dual slopes (steep and shallow), adjacent to uniform stiffness (soft and stiff) regions. While fibroblasts rely on nonmuscle myosin II (NMII) activity and the LIM-domain protein Zyxin, ROCK and the Arp2/3 complex are surprisingly dispensable for durotaxis on either stiffness gradient. Additionally, loss of either actin-elongator Formin-like 3 (FMNL3) or actin-bundler fascin has little impact on durotactic response on stiffness gradients. However, lack of Arp2/3 activity results in a filopodia-based durotactic migration that is equally as efficient as that of lamellipodia-based durotactic migration. Importantly, we uncover essential and specific roles for FMNL3 and fascin in the formation and asymmetric distribution of filopodia during filopodia-based durotaxis response to the stiffness gradients. Together, our tunable all-in-one hydrogel system serves to identify both conserved as well as distinct molecular mechanisms that underlie mechano-responses of cells experiencing altered slopes of stiffness gradients.
Collapse
Affiliation(s)
- Reem M Hakeem
- Department of Biochemistry and Biophysics, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Bhagawat C Subramanian
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Max A Hockenberry
- Department of Cell Biology and Physiology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Zayna T King
- Department of Cell Biology and Physiology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Mitchell T Butler
- Department of Cell Biology and Physiology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - Wesley R Legant
- Department of Pharmacology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| | - James E Bear
- Department of Cell Biology and Physiology, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
- UNC Lineberger Comprehensive Cancer Center, UNC-Chapel Hill School of Medicine, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
19
|
Basu A, Paul MK, Weiss S. The actin cytoskeleton: Morphological changes in pre- and fully developed lung cancer. BIOPHYSICS REVIEWS 2022; 3:041304. [PMID: 38505516 PMCID: PMC10903407 DOI: 10.1063/5.0096188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 12/09/2022] [Indexed: 03/21/2024]
Abstract
Actin, a primary component of the cell cytoskeleton can have multiple isoforms, each of which can have specific properties uniquely suited for their purpose. These monomers are then bound together to form polymeric filaments utilizing adenosine triphosphate hydrolysis as a source of energy. Proteins, such as Arp2/3, VASP, formin, profilin, and cofilin, serve important roles in the polymerization process. These filaments can further be linked to form stress fibers by proteins called actin-binding proteins, such as α-actinin, myosin, fascin, filamin, zyxin, and epsin. These stress fibers are responsible for mechanotransduction, maintaining cell shape, cell motility, and intracellular cargo transport. Cancer metastasis, specifically epithelial mesenchymal transition (EMT), which is one of the key steps of the process, is accompanied by the formation of thick stress fibers through the Rho-associated protein kinase, MAPK/ERK, and Wnt pathways. Recently, with the advent of "field cancerization," pre-malignant cells have also been demonstrated to possess stress fibers and related cytoskeletal features. Analytical methods ranging from western blot and RNA-sequencing to cryo-EM and fluorescent imaging have been employed to understand the structure and dynamics of actin and related proteins including polymerization/depolymerization. More recent methods involve quantifying properties of the actin cytoskeleton from fluorescent images and utilizing them to study biological processes, such as EMT. These image analysis approaches exploit the fact that filaments have a unique structure (curvilinear) compared to the noise or other artifacts to separate them. Line segments are extracted from these filament images that have assigned lengths and orientations. Coupling such methods with statistical analysis has resulted in development of a new reporter for EMT in lung cancer cells as well as their drug responses.
Collapse
Affiliation(s)
| | | | - Shimon Weiss
- Author to whom correspondence should be addressed:
| |
Collapse
|
20
|
Bellingham-Johnstun K, Commer B, Levesque B, Tyree ZL, Laplante C. Imp2p forms actin-dependent clusters and imparts stiffness to the contractile ring. Mol Biol Cell 2022; 33:ar145. [PMID: 36287824 PMCID: PMC9727792 DOI: 10.1091/mbc.e22-06-0221] [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] [Indexed: 02/04/2023] Open
Abstract
The contractile ring must anchor to the plasma membrane and cell wall to transmit its tension. F-BAR domain containing proteins including Imp2p and Cdc15p in fission yeast are likely candidate anchoring proteins based on their mutant phenotypes. Cdc15p is a node component, links the actin bundle to the plasma membrane, recruits Bgs1p to the division plane, prevents contractile ring sliding, and contributes to the stiffness of the contractile ring. Less is known about Imp2p. We found that similarly to Cdc15p, Imp2p contributes to the stiffness of the contractile ring and assembles into protein clusters. Imp2p clusters contain approximately eight Imp2p dimers and depend on the actin network for their stability at the division plane. Importantly, Imp2p and Cdc15p reciprocally affect the amount of each other in the contractile ring, indicating that the two proteins influence each other during cytokinesis, which may partially explain their similar phenotypes.
Collapse
Affiliation(s)
| | - Blake Commer
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607
| | - Brié Levesque
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607
| | - Zoe L Tyree
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607
| | - Caroline Laplante
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607
| |
Collapse
|
21
|
Moshtohry M, Bellingham-Johnstun K, Elting MW, Laplante C. Laser ablation reveals the impact of Cdc15p on the stiffness of the contractile. Mol Biol Cell 2022; 33:br9. [PMID: 35274981 PMCID: PMC9265155 DOI: 10.1091/mbc.e21-10-0515] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The mechanics that govern the constriction of the contractile ring remain poorly understood yet are critical to understanding the forces that drive cytokinesis. We used laser ablation in fission yeast cells to unravel these mechanics focusing on the role of Cdc15p as a putative anchoring protein. Our work shows that the severed constricting contractile ring recoils to a finite point leaving a gap that can heal if less than ∼1 µm. Severed contractile rings in Cdc15p-depleted cells exhibit an exaggerated recoil, which suggests that the recoil is limited by the anchoring of the ring to the plasma membrane. Based on a physical model of the severed contractile ring, we propose that Cdc15p impacts the stiffness of the contractile ring more than the viscous drag.
Collapse
Affiliation(s)
- Mohamed Moshtohry
- Department of Physics, North Carolina State University, Raleigh, NC 27607
| | | | - Mary Williard Elting
- Department of Physics, North Carolina State University, Raleigh, NC 27607.,Cluster for Quantitative and Computational Developmental Biology, North Carolina State University, Raleigh, NC 27607
| | - Caroline Laplante
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607.,Cluster for Quantitative and Computational Developmental Biology, North Carolina State University, Raleigh, NC 27607
| |
Collapse
|
22
|
Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6:426. [PMID: 34916490 PMCID: PMC8674418 DOI: 10.1038/s41392-021-00830-x] [Citation(s) in RCA: 431] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Hydrogel is a type of versatile platform with various biomedical applications after rational structure and functional design that leverages on material engineering to modulate its physicochemical properties (e.g., stiffness, pore size, viscoelasticity, microarchitecture, degradability, ligand presentation, stimulus-responsive properties, etc.) and influence cell signaling cascades and fate. In the past few decades, a plethora of pioneering studies have been implemented to explore the cell-hydrogel matrix interactions and figure out the underlying mechanisms, paving the way to the lab-to-clinic translation of hydrogel-based therapies. In this review, we first introduced the physicochemical properties of hydrogels and their fabrication approaches concisely. Subsequently, the comprehensive description and deep discussion were elucidated, wherein the influences of different hydrogels properties on cell behaviors and cellular signaling events were highlighted. These behaviors or events included integrin clustering, focal adhesion (FA) complex accumulation and activation, cytoskeleton rearrangement, protein cyto-nuclei shuttling and activation (e.g., Yes-associated protein (YAP), catenin, etc.), cellular compartment reorganization, gene expression, and further cell biology modulation (e.g., spreading, migration, proliferation, lineage commitment, etc.). Based on them, current in vitro and in vivo hydrogel applications that mainly covered diseases models, various cell delivery protocols for tissue regeneration and disease therapy, smart drug carrier, bioimaging, biosensor, and conductive wearable/implantable biodevices, etc. were further summarized and discussed. More significantly, the clinical translation potential and trials of hydrogels were presented, accompanied with which the remaining challenges and future perspectives in this field were emphasized. Collectively, the comprehensive and deep insights in this review will shed light on the design principles of new biomedical hydrogels to understand and modulate cellular processes, which are available for providing significant indications for future hydrogel design and serving for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China.
| | - Kun Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China.
| |
Collapse
|
23
|
Castaneda N, Feuillie C, Molinari M, Kang EH. Actin Bundle Nanomechanics and Organization Are Modulated by Macromolecular Crowding and Electrostatic Interactions. Front Mol Biosci 2021; 8:760950. [PMID: 34901154 PMCID: PMC8662701 DOI: 10.3389/fmolb.2021.760950] [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: 08/19/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022] Open
Abstract
The structural and mechanical properties of actin bundles are essential to eukaryotic cells, aiding in cell motility and mechanical support of the plasma membrane. Bundle formation occurs in crowded intracellular environments composed of various ions and macromolecules. Although the roles of cations and macromolecular crowding in the mechanics and organization of actin bundles have been independently established, how changing both intracellular environmental conditions influence bundle mechanics at the nanoscale has yet to be established. Here we investigate how electrostatics and depletion interactions modulate the relative Young’s modulus and height of actin bundles using atomic force microscopy. Our results demonstrate that cation- and depletion-induced bundles display an overall reduction of relative Young’s modulus depending on either cation or crowding concentrations. Furthermore, we directly measure changes to cation- and depletion-induced bundle height, indicating that bundles experience alterations to filament packing supporting the reduction to relative Young’s modulus. Taken together, our work suggests that electrostatic and depletion interactions may act counteractively, impacting actin bundle nanomechanics and organization.
Collapse
Affiliation(s)
- Nicholas Castaneda
- NanoScience Technology Center, University of Central Florida, Orlando, FL, United States.,Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Cecile Feuillie
- Institute of Chemistry and Biology of Membranes and Nano-objects, CBMN CNRS UMR 5248, IPB, Université de Bordeaux, Pessac, France
| | - Michael Molinari
- Institute of Chemistry and Biology of Membranes and Nano-objects, CBMN CNRS UMR 5248, IPB, Université de Bordeaux, Pessac, France
| | - Ellen Hyeran Kang
- NanoScience Technology Center, University of Central Florida, Orlando, FL, United States.,Department of Physics, University of Central Florida, Orlando, FL, United States.,Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, United States
| |
Collapse
|
24
|
Marsal M, Hernández-Vega A, Pouille PA, Martin-Blanco E. Rab5ab-Mediated Yolk Cell Membrane Endocytosis Is Essential for Zebrafish Epiboly and Mechanical Equilibrium During Gastrulation. Front Cell Dev Biol 2021; 9:697097. [PMID: 34778246 PMCID: PMC8585776 DOI: 10.3389/fcell.2021.697097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 09/01/2021] [Indexed: 11/13/2022] Open
Abstract
Morphogenesis in early embryos demands the coordinated distribution of cells and tissues to their final destination in a spatio-temporal controlled way. Spatial and scalar differences in adhesion and contractility are essential for these morphogenetic movements, while the role that membrane remodeling may play remains less clear. To evaluate how membrane turnover modulates tissue arrangements we studied the role of endocytosis in zebrafish epiboly. Experimental analyses and modeling have shown that the expansion of the blastoderm relies on an asymmetry of mechanical tension in the yolk cell generated as a result of actomyosin-dependent contraction and membrane removal. Here we show that the GTPase Rab5ab is essential for the endocytosis and the removal of the external yolk cell syncytial layer (E-YSL) membrane. Interfering in its expression exclusively in the yolk resulted in the reduction of yolk cell actomyosin contractility, the disruption of cortical and internal flows, a disequilibrium in force balance and epiboly impairment. We conclude that regulated membrane remodeling is crucial for directing cell and tissue mechanics, preserving embryo geometry and coordinating morphogenetic movements during epiboly.
Collapse
Affiliation(s)
- Maria Marsal
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Parc Cientific de Barcelona, Barcelona, Spain
| | - Amayra Hernández-Vega
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Parc Cientific de Barcelona, Barcelona, Spain
| | - Philippe-Alexandre Pouille
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Parc Cientific de Barcelona, Barcelona, Spain
| | - Enrique Martin-Blanco
- Instituto de Biología Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Parc Cientific de Barcelona, Barcelona, Spain
| |
Collapse
|
25
|
Correlation of cellular traction forces and dissociation kinetics of adhesive protein zyxin revealed by multi-parametric live cell microscopy. PLoS One 2021; 16:e0251411. [PMID: 33974655 PMCID: PMC8112686 DOI: 10.1371/journal.pone.0251411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/26/2021] [Indexed: 01/02/2023] Open
Abstract
Cells exert traction forces on the extracellular matrix to which they are adhered through the formation of focal adhesions. Spatial-temporal regulation of traction forces is crucial in cell adhesion, migration, cellular division, and remodeling of the extracellular matrix. By cultivating cells on polyacrylamide hydrogels of different stiffness we were able to investigate the effects of substrate stiffness on the generation of cellular traction forces by Traction Force Microscopy (TFM), and characterize the molecular dynamics of the focal adhesion protein zyxin by Fluorescence Correlation Spectroscopy (FCS) and Fluorescence Recovery After Photobleaching (FRAP). As the rigidity of the substrate increases, we observed an increment of both, cellular traction generation and zyxin residence time at the focal adhesions, while its diffusion would not be altered. Moreover, we found a positive correlation between the traction forces exerted by cells and the residence time of zyxin at the substrate elasticities studied. We found that this correlation persists at the subcellular level, even if there is no variation in substrate stiffness, revealing that focal adhesions that exert greater traction present longer residence time for zyxin, i.e., zyxin protein has less probability to dissociate from the focal adhesion.
Collapse
|
26
|
Cheah JS, Jacobs KA, Lai TW, Caballelo R, Yee JL, Ueda S, Heinrich V, Yamada S. Spatial proximity of proteins surrounding zyxin under force-bearing conditions. Mol Biol Cell 2021; 32:1221-1228. [PMID: 33909446 PMCID: PMC8351546 DOI: 10.1091/mbc.e19-10-0568] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Sensing physical forces is a critical first step in mechano-transduction of cells. Zyxin, a LIM domain-containing protein, is recruited to force-bearing actin filaments and is thought to repair and strengthen them. Yet, the precise force-induced protein interactions surrounding zyxin remain unclear. Using BioID analysis, we identified proximal proteins surrounding zyxin under normal and force-bearing conditions by label-free mass spectrometry analysis. Under force-bearing conditions, increased biotinylation of α-actinin 1, α-actinin 4, and AFAP1 were detected, and these proteins accumulated along force-bearing actin fibers independently from zyxin, albeit at a lower intensity than zyxin. VASP also accumulated along force-bearing actin fibers in a zyxin-dependent manner, but the biotinylation of VASP remained constant regardless of force, supporting the model of a free zyxin-VASP complex in the cytoplasm being corecruited to tensed actin fibers. In addition, ARHGAP42, a RhoA GAP, was also identified as a proximal protein of zyxin and colocalized with zyxin along contractile actin bundles. The overexpression of ARHGAP42 reduced the rate of small wound closure, a zyxin-dependent process. These results demonstrate that the application of proximal biotinylation can resolve the proximity and composition of protein complexes as a function of force, which had not been possible with traditional biochemical analysis.
Collapse
Affiliation(s)
- Joleen S Cheah
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616.,Biosciences Program, Stanford University, Stanford, CA 94305
| | - Kyle A Jacobs
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Tzu Wei Lai
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Reca Caballelo
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Jacqueline L Yee
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Shuji Ueda
- Graduate School of Agricultural Science, Kobe University, Kobe, Japan 657
| | - Volkmar Heinrich
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| | - Soichiro Yamada
- Biomedical Engineering Department, University of California Davis, Davis, CA 95616
| |
Collapse
|
27
|
Wang Y, Zhang C, Yang W, Shao S, Xu X, Sun Y, Li P, Liang L, Wu C. LIMD1 phase separation contributes to cellular mechanics and durotaxis by regulating focal adhesion dynamics in response to force. Dev Cell 2021; 56:1313-1325.e7. [PMID: 33891898 DOI: 10.1016/j.devcel.2021.04.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/25/2020] [Accepted: 03/30/2021] [Indexed: 01/09/2023]
Abstract
Cells sense and respond to extracellular mechanical cues through cell-matrix adhesions. Interestingly, the maturation of focal adhesions (FAs) is reciprocally force dependent. How biomechanical cues dictate the status of cell motility and how FAs spatial temporally coordinate force sensing and self-organization remain enigmatic. Here, we identify that LIMD1, a member of the LIM domain scaffolding proteins, undergoes force-sensitive condensation at the FAs. We also unveil that the multivalent interactions of LIMD1 intrinsically disordered region (IDR) and the LIM domains concertedly drive this phase transition under the regulation of phosphorylation. Intriguingly, formation of condensed LIMD1 protein compartments is sufficient to specifically enrich and localize late FA proteins. We further discover that LIMD1 regulates cell spreading, maintains FA dynamics and cellular contractility, and is critical for durotaxis-the ability of cells to crawl along gradients of substrate stiffness. Our results suggest a model that recruitment of LIMD1 to the FAs, via mechanical force triggered inter-molecular interaction, serves as a phase separation hub to assemble and organize matured FAs, thus allowing for efficient mechano-transduction and cell migration.
Collapse
Affiliation(s)
- Yuan Wang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China
| | - Chunlei Zhang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China
| | - Wenzhong Yang
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China
| | - ShiPeng Shao
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China; Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Xinmin Xu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China
| | - Yujie Sun
- State Key Laboratory of Membrane Biology, Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, Peking University, Beijing 100871, China
| | - Pilong Li
- Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ling Liang
- Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing 100191, China.
| |
Collapse
|
28
|
Mahmoudi M, Farghadan A, McConnell DR, Barker AJ, Wentzel JJ, Budoff MJ, Arzani A. The Story of Wall Shear Stress in Coronary Artery Atherosclerosis: Biochemical Transport and Mechanotransduction. J Biomech Eng 2021; 143:041002. [PMID: 33156343 DOI: 10.1115/1.4049026] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Indexed: 12/20/2022]
Abstract
Coronary artery atherosclerosis is a local, multifactorial, complex disease, and the leading cause of death in the US. Complex interactions between biochemical transport and biomechanical forces influence disease growth. Wall shear stress (WSS) affects coronary artery atherosclerosis by inducing endothelial cell mechanotransduction and by controlling the near-wall transport processes involved in atherosclerosis. Each of these processes is controlled by WSS differently and therefore has complicated the interpretation of WSS in atherosclerosis. In this paper, we present a comprehensive theory for WSS in atherosclerosis. First, a short review of shear stress-mediated mechanotransduction in atherosclerosis was presented. Next, subject-specific computational fluid dynamics (CFD) simulations were performed in ten coronary artery models of diseased and healthy subjects. Biochemical-specific mass transport models were developed to study low-density lipoprotein, nitric oxide, adenosine triphosphate, oxygen, monocyte chemoattractant protein-1, and monocyte transport. The transport results were compared with WSS vectors and WSS Lagrangian coherent structures (WSS LCS). High WSS magnitude protected against atherosclerosis by increasing the production or flux of atheroprotective biochemicals and decreasing the near-wall localization of atherogenic biochemicals. Low WSS magnitude promoted atherosclerosis by increasing atherogenic biochemical localization. Finally, the attracting WSS LCS's role was more complex where it promoted or prevented atherosclerosis based on different biochemicals. We present a summary of the different pathways by which WSS influences coronary artery atherosclerosis and compare different mechanotransduction and biotransport mechanisms.
Collapse
Affiliation(s)
- Mostafa Mahmoudi
- Department of Mechanical Engineering, Northern Arizona University, Flagstaff, AZ 86011
| | - Ali Farghadan
- Department of Mechanical Engineering, Northern Arizona University, Flagstaff, AZ 86011
| | - Daniel R McConnell
- Department of Mechanical Engineering, Northern Arizona University, Flagstaff, AZ 86011
| | - Alex J Barker
- Department of Pediatric Radiology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO 80045
| | - Jolanda J Wentzel
- Department of Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, The Netherlands
| | | | - Amirhossein Arzani
- Department of Mechanical Engineering, Northern Arizona University, Flagstaff, AZ 86011
| |
Collapse
|
29
|
Vignaud T, Copos C, Leterrier C, Toro-Nahuelpan M, Tseng Q, Mahamid J, Blanchoin L, Mogilner A, Théry M, Kurzawa L. Stress fibres are embedded in a contractile cortical network. NATURE MATERIALS 2021; 20:410-420. [PMID: 33077951 PMCID: PMC7610471 DOI: 10.1038/s41563-020-00825-z] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 09/14/2020] [Indexed: 05/06/2023]
Abstract
Contractile actomyosin networks are responsible for the production of intracellular forces. There is increasing evidence that bundles of actin filaments form interconnected and interconvertible structures with the rest of the network. In this study, we explored the mechanical impact of these interconnections on the production and distribution of traction forces throughout the cell. By using a combination of hydrogel micropatterning, traction force microscopy and laser photoablation, we measured the relaxation of traction forces in response to local photoablations. Our experimental results and modelling of the mechanical response of the network revealed that bundles were fully embedded along their entire length in a continuous and contractile network of cortical filaments. Moreover, the propagation of the contraction of these bundles throughout the entire cell was dependent on this embedding. In addition, these bundles appeared to originate from the alignment and coalescence of thin and unattached cortical actin filaments from the surrounding mesh.
Collapse
Affiliation(s)
- Timothée Vignaud
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France
- Clinique de Chirurgie Digestive et Endocrinienne, Hôtel Dieu, Nantes, France
| | - Calina Copos
- Courant Institute and Department of Biology, New York University, New York, NY, USA
| | - Christophe Leterrier
- NeuroCyto, Institute of NeuroPhysiopathology (INP), CNRS, Aix Marseille Université, Marseille, France
| | - Mauricio Toro-Nahuelpan
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Qingzong Tseng
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France
| | - Julia Mahamid
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Laurent Blanchoin
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France
| | - Alex Mogilner
- Courant Institute and Department of Biology, New York University, New York, NY, USA.
| | - Manuel Théry
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France.
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France.
| | - Laetitia Kurzawa
- CytoMorpho Lab, Interdisciplinary Research Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, Grenoble-Alpes University/CEA/CNRS/INRA, Grenoble, France.
- CytoMorpho Lab, Hôpital Saint Louis, Institut Universitaire d'Hématologie, Université Paris Diderot/CEA/INSERM, Paris, France.
| |
Collapse
|
30
|
Park J, Lee M, Lee B, Castaneda N, Tetard L, Kang EH. Crowding tunes the organization and mechanics of actin bundles formed by crosslinking proteins. FEBS Lett 2020; 595:26-40. [PMID: 33020904 DOI: 10.1002/1873-3468.13949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 01/05/2023]
Abstract
Fascin and α-actinin form higher-ordered actin bundles that mediate numerous cellular processes including cell morphogenesis and movement. While it is understood crosslinked bundle formation occurs in crowded cytoplasm, how crowding affects the bundling activities of the two crosslinking proteins is not known. Here, we demonstrate how solution crowding modulates the organization and mechanical properties of fascin- and α-actinin-induced bundles, utilizing total internal reflection fluorescence and atomic force microscopy imaging. Molecular dynamics simulations support the inference that crowding reduces binding interaction between actin filaments and fascin or the calponin homology 1 domain of α-actinin evidenced by interaction energy and hydrogen bonding analysis. Based on our findings, we suggest a mechanism of crosslinked actin bundle assembly and mechanics in crowded intracellular environments.
Collapse
Affiliation(s)
- Jinho Park
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA.,Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, USA
| | - Myeongsang Lee
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA
| | - Briana Lee
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA
| | - Nicholas Castaneda
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA.,Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Laurene Tetard
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA.,Department of Physics, University of Central Florida, Orlando, FL, USA
| | - Ellen Hyeran Kang
- NanoScience Technology Center, University of Central Florida, Orlando, FL, USA.,Department of Materials Science and Engineering, University of Central Florida, Orlando, FL, USA.,Department of Physics, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
31
|
Actin polymerization downstream of integrins: signaling pathways and mechanotransduction. Biochem J 2020; 477:1-21. [PMID: 31913455 DOI: 10.1042/bcj20170719] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 11/17/2019] [Accepted: 12/16/2019] [Indexed: 01/04/2023]
Abstract
A cell constantly adapts to its environment. Cell decisions to survive, to proliferate or to migrate are dictated not only by soluble growth factors, but also through the direct interaction of the cell with the surrounding extracellular matrix (ECM). Integrins and their connections to the actin cytoskeleton are crucial for monitoring cell attachment and the physical properties of the substratum. Cell adhesion dynamics are modulated in complex ways by the polymerization of branched and linear actin arrays, which in turn reinforce ECM-cytoskeleton connection. This review describes the major actin regulators, Ena/VASP proteins, formins and Arp2/3 complexes, in the context of signaling pathways downstream of integrins. We focus on the specific signaling pathways that transduce the rigidity of the substrate and which control durotaxis, i.e. directed migration of cells towards increased ECM rigidity. By doing so, we highlight several recent findings on mechanotransduction and put them into a broad integrative perspective that is the result of decades of intense research on the actin cytoskeleton and its regulation.
Collapse
|
32
|
Su Z, Chang Q, Drelich A, Shelite T, Judy B, Liu Y, Xiao J, Zhou C, He X, Jin Y, Saito T, Tang S, Soong L, Wakamiya M, Fang X, Bukreyev A, Ksiazek T, Russell WK, Gong B. Annexin A2 depletion exacerbates the intracerebral microhemorrhage induced by acute rickettsia and Ebola virus infections. PLoS Negl Trop Dis 2020; 14:e0007960. [PMID: 32687500 PMCID: PMC7392349 DOI: 10.1371/journal.pntd.0007960] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 07/30/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022] Open
Abstract
Intracerebral microhemorrhages (CMHs) are small foci of hemorrhages in the cerebrum. Acute infections induced by some intracellular pathogens, including rickettsia, can result in CMHs. Annexin a2 (ANXA2) has been documented to play a functional role during intracellular bacterial adhesion. Here we report that ANXA2-knockout (KO) mice are more susceptible to CMHs in response to rickettsia and Ebola virus infections, suggesting an essential role of ANXA2 in protecting vascular integrity during these intracellular pathogen infections. Proteomic analysis via mass spectrometry of whole brain lysates and brain-derived endosomes from ANXA2-KO and wild-type (WT) mice post-infection with R. australis revealed that a variety of significant proteins were differentially expressed, and the follow-up function enrichment analysis had identified several relevant cell-cell junction functions. Immunohistology study confirmed that both infected WT and infected ANXA2-KO mice were subjected to adherens junctional protein (VE-cadherin) damages. However, key blood-brain barrier (BBB) components, tight junctional proteins ZO-1 and occludin, were disorganized in the brains from R. australis-infected ANXA2-KO mice, but not those of infected WT mice. Similar ANXA2-KO dependent CMHs and fragments of ZO-1 and occludin were also observed in Ebola virus-infected ANXA2-KO mice, but not found in infected WT mice. Overall, our study revealed a novel role of ANXA2 in the formation of CMHs during R. australis and Ebola virus infections; and the underlying mechanism is relevant to the role of ANXA2-regulated tight junctions and its role in stabilizing the BBB in these deadly infections.
Collapse
Affiliation(s)
- Zhengchen Su
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Aleksandra Drelich
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas Shelite
- Department of Internal Medicine, Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Barbara Judy
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yakun Liu
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jie Xiao
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Changchen Zhou
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Xi He
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University Medical Campus, Boston, Massachusetts, United States of America
| | - Tais Saito
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
| | - Shaojun Tang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lynn Soong
- Galveston National Laboratory, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Maki Wakamiya
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Xiang Fang
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
| | - William K. Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Galveston National Laboratory, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
33
|
Chan FY, Silva AM, Carvalho AX. Using the Four-Cell C. elegans Embryo to Study Contractile Ring Dynamics During Cytokinesis. Methods Mol Biol 2020; 2101:297-325. [PMID: 31879911 DOI: 10.1007/978-1-0716-0219-5_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cytokinesis is the process that completes cell division by partitioning the contents of the mother cell between the two daughter cells. It involves the highly regulated assembly and constriction of an actomyosin contractile ring, whose function is to pinch the mother cell in two. Research on the contractile ring has particularly focused on the signaling mechanisms that dictate when and where the ring is formed. In vivo studies of ring constriction are however scarce and its mechanistic understanding is therefore limited. Here we present several experimental approaches for monitoring ring constriction in vivo, using the four-cell C. elegans embryo as model. These approaches allow for the ring to be perturbed only after it forms and include the combination of live imaging with acute drug treatments, temperature-sensitive mutants and rapid temperature shifts, as well as laser microsurgery. In addition, we explain how to combine these with RNAi-mediated depletion of specific components of the cytokinetic machinery.
Collapse
Affiliation(s)
- Fung Yi Chan
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Ana Marta Silva
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal
| | - Ana Xavier Carvalho
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Porto, Portugal.
| |
Collapse
|
34
|
Inhibition of protein FAK enhances 5-FU chemosensitivity to gastric carcinoma via p53 signaling pathways. Comput Struct Biotechnol J 2019; 18:125-136. [PMID: 31969973 PMCID: PMC6961071 DOI: 10.1016/j.csbj.2019.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 12/17/2019] [Accepted: 12/18/2019] [Indexed: 02/05/2023] Open
Abstract
The small molecule drug 5-fluorouracil (5-FU) is widely used in the treatment for gastric cancer (GC), however, it exerts poor efficacy and is associated with acquired and intrinsic resistance. Focal adhesion kinase (FAK), a non-receptor tyrosine kinase, plays a key role in adhesion, migration, and proliferation of gastric carcinoma cells, suggesting that this kinase may be a promising therapeutic target. Differentially expressed FAK in GC tissue was detected by RT-qPCR and TCGA database analysis. To investigate the biological functions of FAK, loss-of-function experiments were performed. CCK-8 assay, colony formation assay, flow cytometry, dual-luciferase reporter assays, and western blot assays were conducted to determine the underlying mechanisms of FAK in 5-FU chemosensitivity in GC. FAK is overexpressed in GC patients, and positively correlated with poor prognosis. The use of shRNA interference to target FAK decreased proliferation and increased apoptosis of GC cells in vitro. Importantly, FAK silencing enhanced the therapeutic efficacy of 5-FU, leading to reduced tumor growth in vivo. We further demonstrated that FAK silencing increased 5-FU-induced caspase-3 activity, and promoted p53 transcriptional activities. Clinical data also has shown that patients with higher levels of FAK had significantly shorter overall survival (OS) and time to first progression (FP) than those with lower levels of FAK. These findings indicate that FAK plays a critical role in 5-FU chemosensitivity in GC, and the use of FAK inhibitors as an adjunct to 5-FU might be an effective strategy for patients who undergo chemotherapy.
Collapse
|
35
|
Catalano S, Panza S, Augimeri G, Giordano C, Malivindi R, Gelsomino L, Marsico S, Giordano F, Győrffy B, Bonofiglio D, Andò S, Barone I. Phosphodiesterase 5 (PDE5) Is Highly Expressed in Cancer-Associated Fibroblasts and Enhances Breast Tumor Progression. Cancers (Basel) 2019; 11:cancers11111740. [PMID: 31698786 PMCID: PMC6895904 DOI: 10.3390/cancers11111740] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/25/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
The overexpression of phosphodiesterase (PDE) 5 is frequently found in various human cancers, such as those of the breast. However, PDE5’s role in the tumor microenvironment is still unknown. As PDE5 represents a high-value therapeutic target, we investigated whether the expression and function of PDE5 in breast cancer-associated fibroblasts (CAFs) may be clinically relevant to malignant progression. PDE5 expression was increased in human breast cancer stroma compared with normal stroma and was correlated to a shorter overall survival. Treatment of CAFs, isolated from breast tumor biopsies, with selective PDE5 inhibitors inhibited their proliferation, motility, and invasiveness, and negatively controlled tumor–stroma interactions in both ‘in vitro’ and ‘in vivo’ models. PDE5 stable overexpression transformed immortalized mouse embryonic fibroblasts (MEFs) towards an activated fibroblast phenotype, impacting their intrinsic characteristics and paracrine effects on breast cancer cell growth and migration through an enhanced production of the C-X-C motif chemokine 16 (CXCL16). On the other hand, CAF exposure to PDE5 inhibitors was associated with reduced CXCL16 expression and secretion. Importantly, CXCL16 levels in breast cancer stroma showed a strong correlation with PDE5 levels and poor patient outcomes. In conclusion, PDE5 is overexpressed in breast cancer stroma, enhances the tumor-stimulatory activities of fibroblasts, and impacts clinical outcomes; thus, we propose this enzyme as an attractive candidate for prognosis and a potential target for treatments in breast cancer patients.
Collapse
Affiliation(s)
- Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
| | - Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
| | - Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
- Centro Sanitario, University of Calabria, 87036 Rende (CS), Italy
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Semmelweis University 2nd Dept. of Pediatrics, 1094 Budapest, Hungary;
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
- Centro Sanitario, University of Calabria, 87036 Rende (CS), Italy
- Correspondence: (S.A.); (I.B.); Tel.: +39-0984-496201 (S.A.); +39-0984-496216 (I.B.); Fax: +39-0984-496203 (S.A. & I.B.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende (CS), Italy; (S.C.); (S.P.); (G.A.); (C.G.); (R.M.); (L.G.); (S.M.); (F.G.); (D.B.)
- Correspondence: (S.A.); (I.B.); Tel.: +39-0984-496201 (S.A.); +39-0984-496216 (I.B.); Fax: +39-0984-496203 (S.A. & I.B.)
| |
Collapse
|
36
|
Abstract
The cytoskeleton provides structural integrity to cells and serves as a key component in mechanotransduction. Tensins are thought to provide a force-bearing linkage between integrins and the actin cytoskeleton; yet, direct evidence of tensin’s role in mechanotransduction is lacking. We here report that local force application to epithelial cells using a micrometer-sized needle leads to rapid accumulation of cten (tensin 4), but not tensin 1, along a fibrous intracellular network. Surprisingly, cten-positive fibers are not actin fibers; instead, these fibers are keratin intermediate filaments. The dissociation of cten from tension-free keratin fibers depends on the duration of cell stretch, demonstrating that the external force favors maturation of cten−keratin network interactions over time and that keratin fibers retain remarkable structural memory of a cell’s force-bearing state. These results establish the keratin network as an integral part of force-sensing elements recruiting distinct proteins like cten and suggest the existence of a mechanotransduction pathway via keratin network.
Collapse
|
37
|
Rauskolb C, Cervantes E, Madere F, Irvine KD. Organization and function of tension-dependent complexes at adherens junctions. J Cell Sci 2019; 132:jcs.224063. [PMID: 30837288 DOI: 10.1242/jcs.224063] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 02/22/2019] [Indexed: 12/17/2022] Open
Abstract
Adherens junctions provide attachments between neighboring epithelial cells and a physical link to the cytoskeleton, which enables them to sense and transmit forces and to initiate biomechanical signaling. Examination of the Ajuba LIM protein Jub in Drosophila embryos revealed that it is recruited to adherens junctions in tissues experiencing high levels of myosin activity, and that the pattern of Jub recruitment varies depending upon how tension is organized. In cells with high junctional myosin, Jub is recruited to puncta near intercellular vertices, which are distinct from Ena-containing puncta, but can overlap Vinc-containing puncta. We identify roles for Jub in modulating tension and cellular organization, which are shared with the cytohesin Step, and the cytohesin adapter Sstn, and show that Jub and Sstn together recruit Step to adherens junctions under tension. Our observations establish Jub as a reporter of tension experienced at adherens junctions, and identify distinct types of tension-dependent and tension-independent junctional complexes. They also identify a role for Jub in mediating a feedback loop that modulates the distribution of tension and cellular organization in epithelia.
Collapse
Affiliation(s)
- Cordelia Rauskolb
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Estelle Cervantes
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Ferralita Madere
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Kenneth D Irvine
- Waksman Institute and Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
38
|
Dasbiswas K, Hu S, Schnorrer F, Safran SA, Bershadsky AD. Ordering of myosin II filaments driven by mechanical forces: experiments and theory. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0114. [PMID: 29632266 DOI: 10.1098/rstb.2017.0114] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2017] [Indexed: 12/27/2022] Open
Abstract
Myosin II filaments form ordered superstructures in both cross-striated muscle and non-muscle cells. In cross-striated muscle, myosin II (thick) filaments, actin (thin) filaments and elastic titin filaments comprise the stereotypical contractile units of muscles called sarcomeres. Linear chains of sarcomeres, called myofibrils, are aligned laterally in registry to form cross-striated muscle cells. The experimentally observed dependence of the registered organization of myofibrils on extracellular matrix elasticity has been proposed to arise from the interactions of sarcomeric contractile elements (considered as force dipoles) through the matrix. Non-muscle cells form small bipolar filaments built of less than 30 myosin II molecules. These filaments are associated in registry forming superstructures ('stacks') orthogonal to actin filament bundles. Formation of myosin II filament stacks requires the myosin II ATPase activity and function of the actin filament crosslinking, polymerizing and depolymerizing proteins. We propose that the myosin II filaments embedded into elastic, intervening actin network (IVN) function as force dipoles that interact attractively through the IVN. This is in analogy with the theoretical picture developed for myofibrils where the elastic medium is now the actin cytoskeleton itself. Myosin stack formation in non-muscle cells provides a novel mechanism for the self-organization of the actin cytoskeleton at the level of the entire cell.This article is part of the theme issue 'Self-organization in cell biology'.
Collapse
Affiliation(s)
- Kinjal Dasbiswas
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA
| | - Shiqiong Hu
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore.,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, 13288 Marseille, France
| | - Samuel A Safran
- Department of Chemical and Biological Physics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Republic of Singapore .,Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
39
|
Ben-Shmuel A, Joseph N, Sabag B, Barda-Saad M. Lymphocyte mechanotransduction: The regulatory role of cytoskeletal dynamics in signaling cascades and effector functions. J Leukoc Biol 2019; 105:1261-1273. [DOI: 10.1002/jlb.mr0718-267r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/19/2018] [Accepted: 01/21/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Aviad Ben-Shmuel
- Laboratory of Molecular and Applied Immunology; Bar-Ilan University; The Mina and Everard Goodman Faculty of Life Sciences; Ramat-Gan Israel
| | - Noah Joseph
- Laboratory of Molecular and Applied Immunology; Bar-Ilan University; The Mina and Everard Goodman Faculty of Life Sciences; Ramat-Gan Israel
| | - Batel Sabag
- Laboratory of Molecular and Applied Immunology; Bar-Ilan University; The Mina and Everard Goodman Faculty of Life Sciences; Ramat-Gan Israel
| | - Mira Barda-Saad
- Laboratory of Molecular and Applied Immunology; Bar-Ilan University; The Mina and Everard Goodman Faculty of Life Sciences; Ramat-Gan Israel
| |
Collapse
|
40
|
Landau S, Moriel A, Livne A, Zheng MH, Bouchbinder E, Levenberg S. Tissue-Level Mechanosensitivity: Predicting and Controlling the Orientation of 3D Vascular Networks. NANO LETTERS 2018; 18:7698-7708. [PMID: 30427693 DOI: 10.1021/acs.nanolett.8b03373] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Understanding the mechanosensitivity of tissues is a fundamentally important problem having far-reaching implications for tissue engineering. Here we study vascular networks formed by a coculture of fibroblasts and endothelial cells embedded in three-dimensional biomaterials experiencing external, physiologically relevant forces. We show that cyclic stretching of the biomaterial orients the newly formed network perpendicular to the stretching direction, independent of the geometric aspect ratio of the biomaterial's sample. A two-dimensional theory explains this observation in terms of the network's stored elastic energy if the cell-embedded biomaterial features a vanishing effective Poisson's ratio, which we directly verify. We further show that under a static stretch, vascular networks orient parallel to the stretching direction due to force-induced anisotropy of the biomaterial polymer network. Finally, static stretching followed by cyclic stretching reveals a competition between the two mechanosensitive mechanisms. These results demonstrate tissue-level mechanosensitivity and constitute an important step toward developing enhanced tissue repair capabilities using well-oriented vascular networks.
Collapse
Affiliation(s)
- Shira Landau
- Department of Biomedical Engineering , Technion-Israel Institute of Technology , Haifa , 3200002 , Israel
| | - Avraham Moriel
- Department of Chemical and Biological Physics , Weizmann Institute of Science , Rehovot 7610001 , Israel
| | - Ariel Livne
- Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot 7610001 , Israel
| | - Ming H Zheng
- Centre for Orthopaedic Research, School of Surgery , The University of Western Australia , Perth , Western Australia , Australia
| | - Eran Bouchbinder
- Department of Chemical and Biological Physics , Weizmann Institute of Science , Rehovot 7610001 , Israel
| | - Shulamit Levenberg
- Department of Biomedical Engineering , Technion-Israel Institute of Technology , Haifa , 3200002 , Israel
| |
Collapse
|
41
|
Activation of Farnesoid X Receptor impairs the tumor-promoting function of breast cancer-associated fibroblasts. Cancer Lett 2018; 437:89-99. [DOI: 10.1016/j.canlet.2018.08.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/30/2018] [Accepted: 08/16/2018] [Indexed: 12/29/2022]
|
42
|
Dharmarajan A, Floren M, Cox L, Ding Y, Johnson R, Tan W. Mechanochemical Effects on Extracellular Signal-Regulated Kinase Dynamics in Stem Cell Differentiation. Tissue Eng Part A 2018; 24:1179-1189. [PMID: 29969368 PMCID: PMC6080114 DOI: 10.1089/ten.tea.2017.0365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/22/2018] [Indexed: 12/20/2022] Open
Abstract
Understanding how key signaling molecules are coregulated by biochemical agents and physical stimuli during stem cell differentiation is critical but often lacking. Due to the important role of extracellular signal-regulated kinase (ERK), this study has examined its temporal dynamics to determine the coregulation of mechanochemical cues on ERK phosphorylation for smooth muscle cell (SMC) differentiation. To assess ERK1/2 activity, a fluorescence resonance energy transfer-based biosensor was transfected into mesenchymal stem cells. The influences of nanopatterned substrates, growth factors, and drugs on ERK activities were related to their effects on SMC differentiation. Results revealed that nanopatterned substrates significantly increased ERK activity in cells, overriding ERK response from administered biochemical factors. The nanopatterned substrates reduced expression of SMC markers after a 48-h biochemical treatment, except for the combination with ERK inhibitor PD98059 treatment, which enhanced expression of mature SMC marker MYH11. Immunofluorescent staining for focal adhesion proteins, vinculin and zyxin, indicated no significant differences in vinculin cluster distribution or dimension, while the location of zyxin changed from adhesion sites of cell periphery on nonpatterned substrate to actin filaments on nanopatterned substrate. The zyxin-reinforced stress fibers likely enhanced the cytoskeletal tension to increase ERK dynamics. Collectively, results suggest that physical stimuli play a dominating role in initial ERK signaling and early-stage differentiation through focal adhesion changes, and the capability of monitoring signaling events in real time could be exploited to guide the engineering of cell microenvironment.
Collapse
Affiliation(s)
- Anirudh Dharmarajan
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Michael Floren
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Lewis Cox
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Yifu Ding
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Richard Johnson
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| | - Wei Tan
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado
| |
Collapse
|
43
|
Martino F, Perestrelo AR, Vinarský V, Pagliari S, Forte G. Cellular Mechanotransduction: From Tension to Function. Front Physiol 2018; 9:824. [PMID: 30026699 PMCID: PMC6041413 DOI: 10.3389/fphys.2018.00824] [Citation(s) in RCA: 595] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
Living cells are constantly exposed to mechanical stimuli arising from the surrounding extracellular matrix (ECM) or from neighboring cells. The intracellular molecular processes through which such physical cues are transformed into a biological response are collectively dubbed as mechanotransduction and are of fundamental importance to help the cell timely adapt to the continuous dynamic modifications of the microenvironment. Local changes in ECM composition and mechanics are driven by a feed forward interplay between the cell and the matrix itself, with the first depositing ECM proteins that in turn will impact on the surrounding cells. As such, these changes occur regularly during tissue development and are a hallmark of the pathologies of aging. Only lately, though, the importance of mechanical cues in controlling cell function (e.g., proliferation, differentiation, migration) has been acknowledged. Here we provide a critical review of the recent insights into the molecular basis of cellular mechanotransduction, by analyzing how mechanical stimuli get transformed into a given biological response through the activation of a peculiar genetic program. Specifically, by recapitulating the processes involved in the interpretation of ECM remodeling by Focal Adhesions at cell-matrix interphase, we revise the role of cytoskeleton tension as the second messenger of the mechanotransduction process and the action of mechano-responsive shuttling proteins converging on stage and cell-specific transcription factors. Finally, we give few paradigmatic examples highlighting the emerging role of malfunctions in cell mechanosensing apparatus in the onset and progression of pathologies.
Collapse
Affiliation(s)
- Fabiana Martino
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, Brno, Czechia
| | - Ana R. Perestrelo
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Vladimír Vinarský
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, Brno, Czechia
| | - Stefania Pagliari
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
| | - Giancarlo Forte
- Center for Translational Medicine, International Clinical Research Center, St. Anne’s University Hospital, Brno, Czechia
- Competence Center for Mechanobiology in Regenerative Medicine, INTERREG ATCZ133, Brno, Czechia
- Department of Biomaterials Science, Institute of Dentistry, University of Turku, Turku, Finland
| |
Collapse
|
44
|
Live cell imaging reveals focal adhesions mechanoresponses in mammary epithelial cells under sustained equibiaxial stress. Sci Rep 2018; 8:9788. [PMID: 29955093 PMCID: PMC6023913 DOI: 10.1038/s41598-018-27948-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/08/2018] [Indexed: 12/22/2022] Open
Abstract
Mechanical stimuli play a key role in many cell functions such as proliferation, differentiation and migration. In the mammary gland, mechanical signals such as the distension of mammary epithelial cells due to udder filling are proposed to be directly involved during lactation and involution. However, the evolution of focal adhesions -specialized multiprotein complexes that mechanically connect cells with the extracellular matrix- during the mammary gland development, as well as the influence of the mechanical stimuli involved, remains unclear. Here we present the use of an equibiaxial stretching device for exerting a sustained normal strain to mammary epithelial cells while quantitatively assessing cell responses by fluorescence imaging techniques. Using this approach, we explored changes in focal adhesion dynamics in HC11 mammary cells in response to a mechanical sustained stress, which resembles the physiological stimuli. We studied the relationship between a global stress and focal adhesion assembly/disassembly, observing an enhanced persistency of focal adhesions under strain as well as an increase in their size. At a molecular level, we evaluated the mechanoresponses of vinculin and zyxin, two focal adhesion proteins postulated as mechanosensors, observing an increment in vinculin molecular tension and a slower zyxin dynamics while increasing the applied normal strain.
Collapse
|
45
|
Lee S, Kassianidou E, Kumar S. Actomyosin stress fiber subtypes have unique viscoelastic properties and roles in tension generation. Mol Biol Cell 2018; 29:1992-2004. [PMID: 29927349 PMCID: PMC6232976 DOI: 10.1091/mbc.e18-02-0106] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Actomyosin stress fibers (SFs) support cell shape and migration by directing intracellular tension to the extracellular matrix (ECM) via focal adhesions. Migrating cells exhibit three SF subtypes (dorsal SFs, transverse arcs, and ventral SFs), which differ in their origin, location, and ECM connectivity. While each subtype is hypothesized to play unique structural roles, this idea has not been directly tested at the single-SF level. Here, we interrogate the mechanical properties of single SFs of each subtype based on their retraction kinetics following laser incision. While each SF subtype bears distinct mechanical properties, these properties are highly interdependent, with incision of dorsal fibers producing centripetal recoil of adjacent transverse arcs and the retraction of incised transverse arcs being limited by attachment points to dorsal SFs. These observations hold whether cells are allowed to spread freely or are confined to crossbow ECM patterns. Consistent with this interdependence, subtype-specific knockdown of dorsal SFs (palladin) or transverse arcs (mDia2) influences ventral SF retraction. These altered mechanics are partially phenocopied in cells cultured on ECM microlines that preclude assembly of dorsal SFs and transverse arcs. Our findings directly demonstrate that different SF subtypes play distinct roles in generating tension and form a mechanically interdependent network.
Collapse
Affiliation(s)
- Stacey Lee
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762.,Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762
| | - Elena Kassianidou
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762.,Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762
| | - Sanjay Kumar
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762.,Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720-1762.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720-1762
| |
Collapse
|
46
|
Dabagh M, Jalali P, Butler PJ, Randles A, Tarbell JM. Mechanotransmission in endothelial cells subjected to oscillatory and multi-directional shear flow. J R Soc Interface 2018; 14:rsif.2017.0185. [PMID: 28515328 DOI: 10.1098/rsif.2017.0185] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/21/2017] [Indexed: 12/27/2022] Open
Abstract
Local haemodynamics are linked to the non-uniform distribution of atherosclerosic lesions in arteries. Low and oscillatory (reversing in the axial flow direction) wall shear stress (WSS) induce inflammatory responses in endothelial cells (ECs) mediating disease localization. The objective of this study is to investigate computationally how the flow direction (reflected in WSS variation on the EC surface over time) influences the forces experienced by structural components of ECs that are believed to play important roles in mechanotransduction. A three-dimensional, multi-scale, multi-component, viscoelastic model of focally adhered ECs is developed, in which oscillatory WSS (reversing or non-reversing) parallel to the principal flow direction, or multi-directional oscillatory WSS with reversing axial and transverse components are applied over the EC surface. The computational model includes the glycocalyx layer, actin cortical layer, nucleus, cytoskeleton, focal adhesions (FAs), stress fibres and adherens junctions (ADJs). We show the distinct effects of atherogenic flow profiles (reversing unidirectional flow and reversing multi-directional flow) on subcellular structures relative to non-atherogenic flow (non-reversing flow). Reversing flow lowers stresses and strains due to viscoelastic effects, and multi-directional flow alters stress on the ADJs perpendicular to the axial flow direction. The simulations predict forces on integrins, ADJ filaments and other substructures in the range that activate mechanotransduction.
Collapse
Affiliation(s)
- Mahsa Dabagh
- Department of Biomedical Engineering, Duke University, Durham, NC, USA .,School of Energy Systems, Lappeenranta University of Technology, Lappeenranta, Finland
| | - Payman Jalali
- School of Energy Systems, Lappeenranta University of Technology, Lappeenranta, Finland
| | - Peter J Butler
- Department of Biomedical Engineering, The Pennsylvania State University, Pennsylvania, PA, USA
| | - Amanda Randles
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - John M Tarbell
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| |
Collapse
|
47
|
Mollaeian K, Liu Y, Bi S, Ren J. Atomic force microscopy study revealed velocity-dependence and nonlinearity of nanoscale poroelasticity of eukaryotic cells. J Mech Behav Biomed Mater 2018; 78:65-73. [DOI: 10.1016/j.jmbbm.2017.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 10/30/2017] [Accepted: 11/01/2017] [Indexed: 11/25/2022]
|
48
|
Kassianidou E, Hughes JH, Kumar S. Activation of ROCK and MLCK tunes regional stress fiber formation and mechanics via preferential myosin light chain phosphorylation. Mol Biol Cell 2017; 28:3832-3843. [PMID: 29046396 PMCID: PMC5739298 DOI: 10.1091/mbc.e17-06-0401] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 10/10/2017] [Accepted: 10/12/2017] [Indexed: 01/21/2023] Open
Abstract
Graded induction of regulatory light chain (RLC) activators MLCK and ROCK were used to explore the relationship between RLC phosphorylation and actin-myosin stress fiber viscoelasticity. MLCK controls peripheral stress fiber mechanics by monophosphorylation of RLC, whereas ROCK acts on central stress fibers via diphosphorylation. The assembly and mechanics of actomyosin stress fibers (SFs) depend on myosin regulatory light chain (RLC) phosphorylation, which is driven by myosin light chain kinase (MLCK) and Rho-associated kinase (ROCK). Although previous work suggests that MLCK and ROCK control distinct pools of cellular SFs, it remains unclear how these kinases differ in their regulation of RLC phosphorylation or how phosphorylation influences individual SF mechanics. Here, we combine genetic approaches with biophysical tools to explore relationships between kinase activity, RLC phosphorylation, SF localization, and SF mechanics. We show that graded MLCK overexpression increases RLC monophosphorylation (p-RLC) in a graded manner and that this p-RLC localizes to peripheral SFs. Conversely, graded ROCK overexpression preferentially increases RLC diphosphorylation (pp-RLC), with pp-RLC localizing to central SFs. Interrogation of single SFs with subcellular laser ablation reveals that MLCK and ROCK quantitatively regulate the viscoelastic properties of peripheral and central SFs, respectively. The effects of MLCK and ROCK on single-SF mechanics may be correspondingly phenocopied by overexpression of mono- and diphosphomimetic RLC mutants. Our results point to a model in which MLCK and ROCK regulate peripheral and central SF viscoelastic properties through mono- and diphosphorylation of RLC, offering new quantitative connections between kinase activity, RLC phosphorylation, and SF viscoelasticity.
Collapse
Affiliation(s)
- Elena Kassianidou
- Department of Bioengineering.,UC Berkeley-UCSF Graduate Program in Bioengineering, and
| | - Jasmine H Hughes
- Department of Bioengineering.,UC Berkeley-UCSF Graduate Program in Bioengineering, and
| | - Sanjay Kumar
- Department of Bioengineering .,UC Berkeley-UCSF Graduate Program in Bioengineering, and.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
49
|
Hoffman L, Jensen CC, Yoshigi M, Beckerle M. Mechanical signals activate p38 MAPK pathway-dependent reinforcement of actin via mechanosensitive HspB1. Mol Biol Cell 2017; 28:2661-2675. [PMID: 28768826 PMCID: PMC5620374 DOI: 10.1091/mbc.e17-02-0087] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/24/2017] [Accepted: 07/28/2017] [Indexed: 01/12/2023] Open
Abstract
Mechanical force induces protein phosphorylations, subcellular redistributions, and actin remodeling. We show that mechanical activation of the p38 MAPK pathway leads to phosphorylation of HspB1 (hsp25/27), which redistributes to cytoskeletal structures, and contributes to the actin cytoskeletal remodeling induced by mechanical stimulation. Despite the importance of a cell’s ability to sense and respond to mechanical force, the molecular mechanisms by which physical cues are converted to cell-instructive chemical information to influence cell behaviors remain to be elucidated. Exposure of cultured fibroblasts to uniaxial cyclic stretch results in an actin stress fiber reinforcement response that stabilizes the actin cytoskeleton. p38 MAPK signaling is activated in response to stretch, and inhibition of p38 MAPK abrogates stretch-induced cytoskeletal reorganization. Here we show that the small heat shock protein HspB1 (hsp25/27) is phosphorylated in stretch-stimulated mouse fibroblasts via a p38 MAPK-dependent mechanism. Phosphorylated HspB1 is recruited to the actin cytoskeleton, displaying prominent accumulation on actin “comet tails” that emanate from focal adhesions in stretch-stimulated cells. Site-directed mutagenesis to block HspB1 phosphorylation inhibits the protein’s cytoskeletal recruitment in response to mechanical stimulation. HspB1-null cells, generated by CRISPR/Cas9 nuclease genome editing, display an abrogated stretch-stimulated actin reinforcement response and increased cell migration. HspB1 is recruited to sites of increased traction force in cells geometrically constrained on micropatterned substrates. Our findings elucidate a molecular pathway by which a mechanical signal is transduced via activation of p38 MAPK to influence actin remodeling and cell migration via a zyxin-independent process.
Collapse
Affiliation(s)
- Laura Hoffman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112.,Department of Biology, University of Utah, Salt Lake City, UT 84112
| | | | - Masaaki Yoshigi
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112
| | - Mary Beckerle
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112 .,Department of Biology, University of Utah, Salt Lake City, UT 84112.,Department of Pediatrics, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
50
|
Endlich K, Kliewe F, Endlich N. Stressed podocytes-mechanical forces, sensors, signaling and response. Pflugers Arch 2017; 469:937-949. [PMID: 28687864 DOI: 10.1007/s00424-017-2025-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 06/22/2017] [Indexed: 02/07/2023]
Abstract
Increased glomerular capillary pressure (glomerular hypertension) and increased glomerular filtration rate (glomerular hyperfiltration) have been proven to cause glomerulosclerosis in animal models and are likely to be operative in patients. Since podocytes cover the glomerular basement membrane, they are exposed to tensile stress due to circumferential wall tension and to fluid shear stress arising from filtrate flow through the narrow filtration slits and through Bowman's space. In vitro evidence documents that podocytes respond to tensile stress as well as to fluid shear stress. Several proteins are discussed in this review that are expressed in podocytes and could act as mechanosensors converting mechanical force via a conformational change into a biochemical signal. The cation channels P2X4 and TRPC6 were shown to be involved in mechanosignaling in podocytes. P2X4 is activated by stretch-induced ATP release, while TRPC6 might be inherently mechanosensitive. Membrane, slit diaphragm and cell-matrix contact proteins are connected to the sublemmal actin network in podocytes via various linker proteins. Therefore, actin-associated proteins, like the proven mechanosensor filamin, are ideal candidates to sense forces in the podocyte cytoskeleton. Furthermore, podocytes express talin, p130Cas, and fibronectin that are known to undergo a conformational change in response to mechanical force exposing cryptic binding sites. Downstream of mechanosensors, experimental evidence suggests the involvement of MAP kinases, Ca2+ and COX2 in mechanosignaling and an emerging role of YAP/TAZ. In summary, our understanding of mechanotransduction in podocytes is still sketchy, but future progress holds promise to identify targets to alleviate conditions of increased mechanical load.
Collapse
Affiliation(s)
- Karlhans Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17489, Greifswald, Germany.
- Institut für Anatomie and Zellbiologie, Universitätsmedizin Greifswald, Friedrich-Loeffler-Str. 23c, 17489, Greifswald, Germany.
| | - Felix Kliewe
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17489, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, 17489, Greifswald, Germany
| |
Collapse
|