1
|
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
|
2
|
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
|
3
|
Vakhrusheva A, Murashko A, Trifonova E, Efremov Y, Timashev P, Sokolova O. Role of Actin-binding Proteins in the Regulation of Cellular Mechanics. Eur J Cell Biol 2022; 101:151241. [DOI: 10.1016/j.ejcb.2022.151241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/18/2022] [Accepted: 05/19/2022] [Indexed: 12/25/2022] Open
|
4
|
Altered mechanotransduction in adolescent idiopathic scoliosis osteoblasts: an exploratory in vitro study. Sci Rep 2022; 12:1846. [PMID: 35115632 PMCID: PMC8813918 DOI: 10.1038/s41598-022-05918-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 01/14/2022] [Indexed: 11/16/2022] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is the most prevalent pediatric spinal deformity. We previously demonstrated elongated cilia and an altered molecular mechanosensory response in AIS osteoblasts. The purpose of this exploratory study was to characterize the mechanosensory defect occurring in AIS osteoblasts. We found that cilia length dynamics in response to flow significantly differ in AIS osteoblasts compared to control cells. In addition, strain-induced rearrangement of actin filaments was compromised resulting in a failure of AIS osteoblasts to position or elongate in function of the bidirectional-applied flow. Contrary to control osteoblasts, fluid flow had an inhibitory effect on AIS cell migration. Moreover, flow induced an increase in secreted VEGF-A and PGE2 in control but not AIS cells. Collectively our data demonstrated that in addition to the observed primary cilium defects, there are cytoskeletal abnormalities correlated to impaired mechanotransduction in AIS. Thus, we propose that the AIS etiology could be a result of generalized defects in cellular mechanotransduction given that an adolescent growing spine is under constant stimulation for growth and bone remodeling in response to applied mechanical forces. Recognition of an altered mechanotransduction as part of the AIS pathomechanism must be considered in the conception and development of more effective bracing treatments.
Collapse
|
5
|
Goelzer M, Goelzer J, Ferguson ML, Neu CP, Uzer G. Nuclear envelope mechanobiology: linking the nuclear structure and function. Nucleus 2021; 12:90-114. [PMID: 34455929 PMCID: PMC8432354 DOI: 10.1080/19491034.2021.1962610] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 01/10/2023] Open
Abstract
The nucleus, central to cellular activity, relies on both direct mechanical input as well as its molecular transducers to sense external stimuli and respond by regulating intra-nuclear chromatin organization that determines cell function and fate. In mesenchymal stem cells of musculoskeletal tissues, changes in nuclear structures are emerging as a key modulator of their differentiation and proliferation programs. In this review we will first introduce the structural elements of the nucleoskeleton and discuss the current literature on how nuclear structure and signaling are altered in relation to environmental and tissue level mechanical cues. We will focus on state-of-the-art techniques to apply mechanical force and methods to measure nuclear mechanics in conjunction with DNA, RNA, and protein visualization in living cells. Ultimately, combining real-time nuclear deformations and chromatin dynamics can be a powerful tool to study mechanisms of how forces affect the dynamics of genome function.
Collapse
Affiliation(s)
- Matthew Goelzer
- Materials Science and Engineering, Boise State University, Boise, ID, US
| | | | - Matthew L. Ferguson
- Biomolecular Science, Boise State University, Boise, ID, US
- Physics, Boise State University, Boise, ID, US
| | - Corey P. Neu
- Paul M. Rady Department of Mechanical Engineering, University of Colorado, Boulder, CO, US
| | - Gunes Uzer
- Mechanical and Biomedical Engineering, Boise State University, Boise, ID, US
| |
Collapse
|
6
|
Gould NR, Torre OM, Leser JM, Stains JP. The cytoskeleton and connected elements in bone cell mechano-transduction. Bone 2021; 149:115971. [PMID: 33892173 PMCID: PMC8217329 DOI: 10.1016/j.bone.2021.115971] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/30/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023]
Abstract
Bone is a mechano-responsive tissue that adapts to changes in its mechanical environment. Increases in strain lead to increased bone mass acquisition, whereas decreases in strain lead to a loss of bone mass. Given that mechanical stress is a regulator of bone mass and quality, it is important to understand how bone cells sense and transduce these mechanical cues into biological changes to identify druggable targets that can be exploited to restore bone cell mechano-sensitivity or to mimic mechanical load. Many studies have identified individual cytoskeletal components - microtubules, actin, and intermediate filaments - as mechano-sensors in bone. However, given the high interconnectedness and interaction between individual cytoskeletal components, and that they can assemble into multiple discreet cellular structures, it is likely that the cytoskeleton as a whole, rather than one specific component, is necessary for proper bone cell mechano-transduction. This review will examine the role of each cytoskeletal element in bone cell mechano-transduction and will present a unified view of how these elements interact and work together to create a mechano-sensor that is necessary to control bone formation following mechanical stress.
Collapse
Affiliation(s)
- Nicole R Gould
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Olivia M Torre
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jenna M Leser
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA..
| |
Collapse
|
7
|
Deprez J, Lajoinie G, Engelen Y, De Smedt SC, Lentacker I. Opening doors with ultrasound and microbubbles: Beating biological barriers to promote drug delivery. Adv Drug Deliv Rev 2021; 172:9-36. [PMID: 33705877 DOI: 10.1016/j.addr.2021.02.015] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Apart from its clinical use in imaging, ultrasound has been thoroughly investigated as a tool to enhance drug delivery in a wide variety of applications. Therapeutic ultrasound, as such or combined with cavitating nuclei or microbubbles, has been explored to cross or permeabilize different biological barriers. This ability to access otherwise impermeable tissues in the body makes the combination of ultrasound and therapeutics very appealing to enhance drug delivery in situ. This review gives an overview of the most important biological barriers that can be tackled using ultrasound and aims to provide insight on how ultrasound has shown to improve accessibility as well as the biggest hurdles. In addition, we discuss the clinical applicability of therapeutic ultrasound with respect to the main challenges that must be addressed to enable the further progression of therapeutic ultrasound towards an effective, safe and easy-to-use treatment tailored for drug delivery in patients.
Collapse
Affiliation(s)
- J Deprez
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, P.O. Box 217, 7500 AE Enschede, Netherlands
| | - Y Engelen
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
8
|
Rubert M, Vetsch JR, Lehtoviita I, Sommer M, Zhao F, Studart AR, Müller R, Hofmann S. Scaffold Pore Geometry Guides Gene Regulation and Bone-like Tissue Formation in Dynamic Cultures. Tissue Eng Part A 2021; 27:1192-1204. [PMID: 33297842 DOI: 10.1089/ten.tea.2020.0121] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cells sense and respond to scaffold pore geometry and mechanical stimuli. Many fabrication methods used in bone tissue engineering render structures with poorly controlled pore geometries. Given that cell-scaffold interactions are complex, drawing a conclusion on how cells sense and respond to uncontrolled scaffold features under mechanical loading is difficult. In this study, monodisperse templated scaffolds (MTSC) were fabricated and used as well-defined porous scaffolds to study the effect of dynamic culture conditions on bone-like tissue formation. Human bone marrow-derived stromal cells were cultured on MTSC or conventional salt-leached scaffolds (SLSC) for up to 7 weeks, either under static or dynamic conditions (wall shear stress [WSS] using spinner flask bioreactors). The influence of controlled spherical pore geometry of MTSC subjected to static or dynamic conditions on osteoblast cell differentiation, bone-like tissue formation, structure, and distribution was investigated. WSS generated within the two idealized geometrical scaffold features was assessed. Distinct response to fluid flow in osteoblast cell differentiation were shown to be dependent on scaffold pore geometry. As revealed by collagen staining and microcomputed tomography images, dynamic conditions promoted a more regular extracellular matrix (ECM) formation and mineral distribution in both scaffold types compared with static conditions. The results showed that regulation of bone-related genes and the amount and the structure of mineralized ECM were dependent on scaffold pore geometry and the mechanical cues provided by the two different culture conditions. Under dynamic conditions, SLSC favored osteoblast cell differentiation and ECM formation, whereas MTSC enhanced ECM mineralization. The spherical pore shape in MTSC supported a more trabecular bone-like structure under dynamic conditions compared with MTSC statically cultured or to SLSC under either static or dynamic conditions. These results suggest that cell activity and bone-like tissue formation is driven not only by the pore geometry but also by the mechanical environment. This should be taken into account in the future design of complex scaffolds, which should favor cell differentiation while guiding the formation, structure, and distribution of the engineered bone tissue. This could help to mimic the anatomical complexity of the bone tissue structure and to adapt to each bone defect needs. Impact statement Aging of the human population leads to an increasing need for medical implants with high success rate. We provide evidence that cell activity and the amount and structure of bone-like tissue formation is dependent on the scaffold pore geometry and on the mechanical environment. Fabrication of complex scaffolds comprising concave and planar pore geometries might represent a promising direction toward the tunability and mimicry the structural complexity of the bone tissue. Moreover, the use of fabrication methods that allow a systematic fabrication of reproducible and geometrically controlled structures would simplify scaffold design optimization.
Collapse
Affiliation(s)
- Marina Rubert
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Jolanda Rita Vetsch
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Iina Lehtoviita
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Marianne Sommer
- Complex Materials, Department of Materials, ETH Zurich, Zurich, Switzerland
| | - Feihu Zhao
- Zienkiewicz Centre for Computational Engineering, College of Engineering, Swansea University, Swansea, United Kingdom
| | - André R Studart
- Complex Materials, Department of Materials, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Sandra Hofmann
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology Eindhoven, The Netherlands
| |
Collapse
|
9
|
Hernández-Cáceres MP, Munoz L, Pradenas JM, Pena F, Lagos P, Aceiton P, Owen GI, Morselli E, Criollo A, Ravasio A, Bertocchi C. Mechanobiology of Autophagy: The Unexplored Side of Cancer. Front Oncol 2021; 11:632956. [PMID: 33718218 PMCID: PMC7952994 DOI: 10.3389/fonc.2021.632956] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Proper execution of cellular function, maintenance of cellular homeostasis and cell survival depend on functional integration of cellular processes and correct orchestration of cellular responses to stresses. Cancer transformation is a common negative consequence of mismanagement of coordinated response by the cell. In this scenario, by maintaining the balance among synthesis, degradation, and recycling of cytosolic components including proteins, lipids, and organelles the process of autophagy plays a central role. Several environmental stresses activate autophagy, among those hypoxia, DNA damage, inflammation, and metabolic challenges such as starvation. In addition to these chemical challenges, there is a requirement for cells to cope with mechanical stresses stemming from their microenvironment. Cells accomplish this task by activating an intrinsic mechanical response mediated by cytoskeleton active processes and through mechanosensitive protein complexes which interface the cells with their mechano-environment. Despite autophagy and cell mechanics being known to play crucial transforming roles during oncogenesis and malignant progression their interplay is largely overlooked. In this review, we highlight the role of physical forces in autophagy regulation and their potential implications in both physiological as well as pathological conditions. By taking a mechanical perspective, we wish to stimulate novel questions to further the investigation of the mechanical requirements of autophagy and appreciate the extent to which mechanical signals affect this process.
Collapse
Affiliation(s)
- Maria Paz Hernández-Cáceres
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Leslie Munoz
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Javiera M. Pradenas
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Pena
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Lagos
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Aceiton
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Gareth I. Owen
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
- Facultad De Odontología, Instituto De Investigación En Ciencias Odontológicas (ICOD), Universidad De Chile, Santiago, Chile
| | - Andrea Ravasio
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristina Bertocchi
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| |
Collapse
|
10
|
Wade EM, Halliday BJ, Jenkins ZA, O'Neill AC, Robertson SP. The X‐linked filaminopathies: Synergistic insights from clinical and molecular analysis. Hum Mutat 2020; 41:865-883. [DOI: 10.1002/humu.24002] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/30/2020] [Accepted: 02/24/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Emma M. Wade
- Department of Women's and Children's Health, Dunedin School of MedicineUniversity of Otago Dunedin New Zealand
| | - Benjamin J. Halliday
- Department of Women's and Children's Health, Dunedin School of MedicineUniversity of Otago Dunedin New Zealand
| | - Zandra A. Jenkins
- Department of Women's and Children's Health, Dunedin School of MedicineUniversity of Otago Dunedin New Zealand
| | - Adam C. O'Neill
- Department of Women's and Children's Health, Dunedin School of MedicineUniversity of Otago Dunedin New Zealand
| | - Stephen P. Robertson
- Department of Women's and Children's Health, Dunedin School of MedicineUniversity of Otago Dunedin New Zealand
| |
Collapse
|
11
|
Chen L, Childs RD, Landis WJ. Correlations between gene expression and mineralization in the avian leg tendon. Bone 2019; 121:42-59. [PMID: 30419319 DOI: 10.1016/j.bone.2018.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/02/2018] [Accepted: 11/07/2018] [Indexed: 10/27/2022]
Abstract
Certain avian tendons have been studied previously as a model system for normal mineralization of vertebrates in general. In this regard, the gastrocnemius tendon in the legs of turkeys mineralizes in a well defined temporal and spatial manner such that changes in the initial and subsequent events of mineral formation can be associated with time and specific locations in the tissue. In the present investigation, these parameters and mineral deposition have been correlated with the expression of several genes and the synthesis and secretion of their related extracellular matrix proteins by the composite tenocytes of the tendon. Quantitative polymerase chain reaction analysis demonstrates that mRNA expression of the non-collagenous genes of bone sialoprotein, osteopontin, and osteocalcin corresponds well with the temporal and spatial onset and progression of mineralization. Immunolocalization separately confirms the synthesis and secretion of these matrix molecules. The expression of other non-collagenous genes such as decorin does not show strong correlation with turkey leg tendon mineralization, and expression of vimentin, a cytoskeletal component which may be regulated by biomechanical factors in the tendon, may lead to inhibition of osteocalcin expression during the development and mineralization of the tissue. The overall results of this work provide insight into direct temporal and spatial relations between the genes and proteins of interest as well as the formation and deposition of mineral in the avian tendon model.
Collapse
Affiliation(s)
- Ling Chen
- Department of Polymer Science, University of Akron, Akron, OH, USA
| | | | - William J Landis
- Department of Polymer Science, University of Akron, Akron, OH, USA.
| |
Collapse
|
12
|
Irmak G, Demirtaş TT, Gümüşderelioǧlu M. Highly Methacrylated Gelatin Bioink for Bone Tissue Engineering. ACS Biomater Sci Eng 2018; 5:831-845. [DOI: 10.1021/acsbiomaterials.8b00778] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
13
|
Xu H, Duan J, Ren L, Yang P, Yang R, Li W, Zhao D, Shang P, Jiang JX. Impact of flow shear stress on morphology of osteoblast-like IDG-SW3 cells. J Bone Miner Metab 2018; 36:529-536. [PMID: 29027016 PMCID: PMC6282752 DOI: 10.1007/s00774-017-0870-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/11/2017] [Indexed: 11/26/2022]
Abstract
This study constructed an in situ cell culture, real-time observation system based originally on a microfluidic channel, and reported the morphological changes of late osteoblast-like IDG-SW3 cells in response to flow shear stress (FSS). The effects of high (1.2 Pa) and low (0.3 Pa) magnitudes of unidirectional FSS and three concentrations of extracellular Type I collagen (0.1, 0.5, and 1 mg/mL) coating on cell morphology were investigated. IDG-SW3 cells were cultured in polydimethylsiloxane microfluidic channels. Cell images were recorded real-time under microscope at intervals of 1 min. Cell morphology was characterized by five parameters: cellular area, cell elongation index, cellular alignment, cellular process length, and number of cellular process per cell. Immunofluorescence assay was used to detect stress fiber distribution and vinculin expression. The results showed that 1.2 Pa, but not 0.3 Pa of FSS induced a significant morphological change in late osteoblast-like IDG-SW3 cells, which may be caused by the alteration of cellular adhesion with matrix in response to FSS. Moreover, the amount of collagen matrix, alignment of fiber stress and expression of vinculin were closely correlated with the morphological changes of IDG-SW3 cells. This study suggests that osteoblasts are very responsive to the magnitudes of FSS, and extracellular collagen matrix and focal adhesion are directly involved in the morphological changes adaptive to FSS.
Collapse
Affiliation(s)
- Huiyun Xu
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China.
| | - Jing Duan
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Li Ren
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Pengfei Yang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Ruixin Yang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Wenbin Li
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Dongdong Zhao
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Peng Shang
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Jean X Jiang
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
14
|
Uchida T, Sakashita Y, Kitahata K, Yamashita Y, Tomida C, Kimori Y, Komatsu A, Hirasaka K, Ohno A, Nakao R, Higashitani A, Higashibata A, Ishioka N, Shimazu T, Kobayashi T, Okumura Y, Choi I, Oarada M, Mills EM, Teshima-Kondo S, Takeda S, Tanaka E, Tanaka K, Sokabe M, Nikawa T. Reactive oxygen species upregulate expression of muscle atrophy-associated ubiquitin ligase Cbl-b in rat L6 skeletal muscle cells. Am J Physiol Cell Physiol 2018. [DOI: 10.1152/ajpcell.00184.2017] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Unloading-mediated muscle atrophy is associated with increased reactive oxygen species (ROS) production. We previously demonstrated that elevated ubiquitin ligase casitas B-lineage lymphoma-b (Cbl-b) resulted in the loss of muscle volume (Nakao R, Hirasaka K, Goto J, Ishidoh K, Yamada C, Ohno A, Okumura Y, Nonaka I, Yasutomo K, Baldwin KM, Kominami E, Higashibata A, Nagano K, Tanaka K, Yasui N, Mills EM, Takeda S, Nikawa T. Mol Cell Biol 29: 4798–4811, 2009). However, the pathological role of ROS production associated with unloading-mediated muscle atrophy still remains unknown. Here, we showed that the ROS-mediated signal transduction caused by microgravity or its simulation contributes to Cbl-b expression. In L6 myotubes, the assessment of redox status revealed that oxidized glutathione was increased under microgravity conditions, and simulated microgravity caused a burst of ROS, implicating ROS as a critical upstream mediator linking to downstream atrophic signaling. ROS generation activated the ERK1/2 early-growth response protein (Egr)1/2-Cbl-b signaling pathway, an established contributing pathway to muscle volume loss. Interestingly, antioxidant treatments such as N-acetylcysteine and TEMPOL, but not catalase, blocked the clinorotation-mediated activation of ERK1/2. The increased ROS induced transcriptional activity of Egr1 and/or Egr2 to stimulate Cbl-b expression through the ERK1/2 pathway in L6 myoblasts, since treatment with Egr1/2 siRNA and an ERK1/2 inhibitor significantly suppressed clinorotation-induced Cbl-b and Egr expression, respectively. Promoter and gel mobility shift assays revealed that Cbl-b was upregulated via an Egr consensus oxidative responsive element at −110 to −60 bp of the Cbl-b promoter. Together, this indicates that under microgravity conditions, elevated ROS may be a crucial mechanotransducer in skeletal muscle cells, regulating muscle mass through Cbl-b expression activated by the ERK-Egr signaling pathway.
Collapse
Affiliation(s)
- Takayuki Uchida
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Yoshihiro Sakashita
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Kanako Kitahata
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Yui Yamashita
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Chisato Tomida
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Yuki Kimori
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Akio Komatsu
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Katsuya Hirasaka
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
- Graduate School of Fisheries Science and Environmental Studies, Nagasaki University, Nagasaki, Japan
| | - Ayako Ohno
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| | - Reiko Nakao
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
- Biological Clock Research Group, Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | | | - Akira Higashibata
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Tsukuba, Ibaraki, Japan
| | - Noriaki Ishioka
- Institute of Space and Astronautical Science, Japan Aerospace Exploration Agency, Tsukuba, Ibaraki, Japan
| | | | - Takeshi Kobayashi
- Department of Physiology, Nagoya University, School of Medicine, Nagoya, Japan
| | - Yuushi Okumura
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
- Faculty of Nutritional Science, Sagami Women’s University, Kanagawa, Japan
| | - Inho Choi
- Institute of Space Biology, Yonsei University, Wonju, South Korea
| | - Motoko Oarada
- Faculty of Nutritional Science, Sagami Women’s University, Kanagawa, Japan
| | - Edward M. Mills
- Division of Pharmacology/Toxicology, College of Pharmacy, University of Texas, Austin, Texas
| | - Shigetada Teshima-Kondo
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
- Department of Nutrition, Graduate School of Comprehensive Rehabilitation, Osaka Prefecture University, Osaka, Japan
| | - Shin’ichi Takeda
- Translational Research Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Eiji Tanaka
- Department of Orthodontic Dentistry, Institute of Medical Biosciences, Tokushima University Graduate School, Tokushima, Japan
| | - Keiji Tanaka
- Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masahiro Sokabe
- Department of Physiology, Nagoya University, School of Medicine, Nagoya, Japan
| | - Takeshi Nikawa
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
15
|
Riehl BD, Lee JS, Ha L, Kwon IK, Lim JY. Flowtaxis of osteoblast migration under fluid shear and the effect of RhoA kinase silencing. PLoS One 2017; 12:e0171857. [PMID: 28199362 PMCID: PMC5310897 DOI: 10.1371/journal.pone.0171857] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/26/2017] [Indexed: 02/07/2023] Open
Abstract
Despite the important role of mechanical signals in bone remodeling, relatively little is known about how fluid shear affects osteoblastic cell migration behavior. Here we demonstrated that MC3T3-E1 osteoblast migration could be activated by physiologically-relevant levels of fluid shear in a shear stress-dependent manner. Interestingly, shear-sensitive osteoblast migration behavior was prominent only during the initial period after the onset of the steady flow (for about 30 min), exhibiting shear stress-dependent migration speed, displacement, arrest coefficient, and motility coefficient. For example, cell speed at 1 min was 0.28, 0.47, 0.51, and 0.84 μm min-1 for static, 2, 15, and 25 dyne cm-2 shear stress, respectively. Arrest coefficient (measuring how often cells are paused during migration) assessed for the first 30 min was 0.40, 0.26, 0.24, and 0.12 respectively for static, 2, 15, and 25 dyne cm-2. After this initial period, osteoblasts under steady flow showed decreased migration capacity and diminished shear stress dependency. Molecular interference of RhoA kinase (ROCK), a regulator of cytoskeletal tension signaling, was found to increase the shear-sensitive window beyond the initial period. Cells with ROCK-shRNA had increased migration in the flow direction and continued shear sensitivity, resulting in greater root mean square displacement at the end of 120 min of measurement. It is notable that the transient osteoblast migration behavior was in sharp contrast to mesenchymal stem cells that exhibited sustained shear sensitivity (as we recently reported, J. R. Soc. Interface. 2015; 12:20141351). The study of fluid shear as a driving force for cell migration, i.e., "flowtaxis", and investigation of molecular mechanosensors governing such behavior (e.g., ROCK as tested in this study) may provide new and improved insights into the fundamental understanding of cell migration-based homeostasis.
Collapse
Affiliation(s)
- Brandon D. Riehl
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Jeong Soon Lee
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Ligyeom Ha
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Il Keun Kwon
- The Graduate School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Jung Yul Lim
- Department of Mechanical and Materials Engineering, College of Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- The Graduate School of Dentistry, Kyung Hee University, Seoul, Korea
- * E-mail:
| |
Collapse
|
16
|
Alioscha-Perez M, Benadiba C, Goossens K, Kasas S, Dietler G, Willaert R, Sahli H. A Robust Actin Filaments Image Analysis Framework. PLoS Comput Biol 2016; 12:e1005063. [PMID: 27551746 PMCID: PMC4995035 DOI: 10.1371/journal.pcbi.1005063] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 07/15/2016] [Indexed: 11/18/2022] Open
Abstract
The cytoskeleton is a highly dynamical protein network that plays a central role in numerous cellular physiological processes, and is traditionally divided into three components according to its chemical composition, i.e. actin, tubulin and intermediate filament cytoskeletons. Understanding the cytoskeleton dynamics is of prime importance to unveil mechanisms involved in cell adaptation to any stress type. Fluorescence imaging of cytoskeleton structures allows analyzing the impact of mechanical stimulation in the cytoskeleton, but it also imposes additional challenges in the image processing stage, such as the presence of imaging-related artifacts and heavy blurring introduced by (high-throughput) automated scans. However, although there exists a considerable number of image-based analytical tools to address the image processing and analysis, most of them are unfit to cope with the aforementioned challenges. Filamentous structures in images can be considered as a piecewise composition of quasi-straight segments (at least in some finer or coarser scale). Based on this observation, we propose a three-steps actin filaments extraction methodology: (i) first the input image is decomposed into a ‘cartoon’ part corresponding to the filament structures in the image, and a noise/texture part, (ii) on the ‘cartoon’ image, we apply a multi-scale line detector coupled with a (iii) quasi-straight filaments merging algorithm for fiber extraction. The proposed robust actin filaments image analysis framework allows extracting individual filaments in the presence of noise, artifacts and heavy blurring. Moreover, it provides numerous parameters such as filaments orientation, position and length, useful for further analysis. Cell image decomposition is relatively under-exploited in biological images processing, and our study shows the benefits it provides when addressing such tasks. Experimental validation was conducted using publicly available datasets, and in osteoblasts grown in two different conditions: static (control) and fluid shear stress. The proposed methodology exhibited higher sensitivity values and similar accuracy compared to state-of-the-art methods. We propose a novel actin filaments cytoskeleton analysis framework that allows extracting quasi-straight individual fibers in a robust manner, and provides their respective position, orientation, and length as output. The proposed framework is defined as a three-steps processing sequence, that can explicitly cope with high-throughput imaging related issues, such as noise/artifacts presence and heavy blurring, and can similarly process artifacts-free and well-focused images.
Collapse
Affiliation(s)
- Mitchel Alioscha-Perez
- Electronics and Informatics Dept (ETRO), AVSP Lab, Vrije Universiteit Brussel, Brussels, Belgium
- VUB-EPFL International Joint Research Group (IJRG) NanoBiotechnology and NanoMedicine (NANO), Brussels, Belgium
- * E-mail: (MAP); (HS)
| | - Carine Benadiba
- VUB-EPFL International Joint Research Group (IJRG) NanoBiotechnology and NanoMedicine (NANO), Brussels, Belgium
- Laboratoire de Physique de la Matière Vivante (LPMV), EPFL, Cubotron, Lausanne, Switzerland
| | - Katty Goossens
- VUB-EPFL International Joint Research Group (IJRG) NanoBiotechnology and NanoMedicine (NANO), Brussels, Belgium
- Department of Bioengineering Sciences (DBIT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Sandor Kasas
- VUB-EPFL International Joint Research Group (IJRG) NanoBiotechnology and NanoMedicine (NANO), Brussels, Belgium
- Laboratoire de Physique de la Matière Vivante (LPMV), EPFL, Cubotron, Lausanne, Switzerland
| | - Giovanni Dietler
- VUB-EPFL International Joint Research Group (IJRG) NanoBiotechnology and NanoMedicine (NANO), Brussels, Belgium
- Laboratoire de Physique de la Matière Vivante (LPMV), EPFL, Cubotron, Lausanne, Switzerland
| | - Ronnie Willaert
- VUB-EPFL International Joint Research Group (IJRG) NanoBiotechnology and NanoMedicine (NANO), Brussels, Belgium
- Department of Bioengineering Sciences (DBIT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Hichem Sahli
- Electronics and Informatics Dept (ETRO), AVSP Lab, Vrije Universiteit Brussel, Brussels, Belgium
- VUB-EPFL International Joint Research Group (IJRG) NanoBiotechnology and NanoMedicine (NANO), Brussels, Belgium
- Interuniversity Microelectronics Centre (IMEC), Heverlee, Belgium
- * E-mail: (MAP); (HS)
| |
Collapse
|
17
|
Grady ME, Composto RJ, Eckmann DM. Cell elasticity with altered cytoskeletal architectures across multiple cell types. J Mech Behav Biomed Mater 2016; 61:197-207. [PMID: 26874250 DOI: 10.1016/j.jmbbm.2016.01.022] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/18/2016] [Accepted: 01/22/2016] [Indexed: 12/18/2022]
Abstract
The cytoskeleton is primarily responsible for providing structural support, localization and transport of organelles, and intracellular trafficking. The structural support is supplied by actin filaments, microtubules, and intermediate filaments, which contribute to overall cell elasticity to varying degrees. We evaluate cell elasticity in five different cell types with drug-induced cytoskeletal derangements to probe how actin filaments and microtubules contribute to cell elasticity and whether it is conserved across cell type. Specifically, we measure elastic stiffness in primary chondrocytes, fibroblasts, endothelial cells (HUVEC), hepatocellular carcinoma cells (HUH-7), and fibrosarcoma cells (HT 1080) subjected to two cytoskeletal destabilizers: cytochalasin D and nocodazole, which disrupt actin and microtubule polymerization, respectively. Elastic stiffness is measured by atomic force microscopy (AFM) and the disruption of the cytoskeleton is confirmed using fluorescence microscopy. The two cancer cell lines showed significantly reduced elastic moduli values (~0.5kPa) when compared to the three healthy cell lines (~2kPa). Non-cancer cells whose actin filaments were disrupted using cytochalasin D showed a decrease of 60-80% in moduli values compared to untreated cells of the same origin, whereas the nocodazole-treated cells showed no change in elasticity. Overall, we demonstrate actin filaments contribute more to elastic stiffness than microtubules but this result is cell type dependent. Cancer cells behaved differently, exhibiting increased stiffness as well as stiffness variability when subjected to nocodazole. We show that disruption of microtubule dynamics affects cancer cell elasticity, suggesting therapeutic drugs targeting microtubules be monitored for significant elastic changes.
Collapse
Affiliation(s)
- Martha E Grady
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, 3231 Walnut Street, Philadelphia, PA 19104, United States; Department of Anesthesiology and Critical Care, School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, PA 19104, United States
| | - Russell J Composto
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, 3231 Walnut Street, Philadelphia, PA 19104, United States
| | - David M Eckmann
- Department of Anesthesiology and Critical Care, School of Medicine, University of Pennsylvania, 3620 Hamilton Walk, Philadelphia, PA 19104, United States.
| |
Collapse
|
18
|
Zhao Y, Shapiro SS, Eto M. F-actin clustering and cell dysmotility induced by the pathological W148R missense mutation of filamin B at the actin-binding domain. Am J Physiol Cell Physiol 2015; 310:C89-98. [PMID: 26491051 DOI: 10.1152/ajpcell.00274.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/19/2015] [Indexed: 11/22/2022]
Abstract
Filamin B (FLNB) is a dimeric actin-binding protein that orchestrates the reorganization of the actin cytoskeleton. Congenital mutations of FLNB at the actin-binding domain (ABD) are known to cause abnormalities of skeletal development, such as atelosteogenesis types I and III and Larsen's syndrome, although the underlying mechanisms are poorly understood. Here, using fluorescence microscopy, we characterized the reorganization of the actin cytoskeleton in cells expressing each of six pathological FLNB mutants that have been linked to skeletal abnormalities. The subfractionation assay showed a greater accumulation of the FLNB ABD mutants W148R and E227K than the wild-type protein to the cytoskeleton. Ectopic expression of FLNB-W148R and, to a lesser extent, FLNB-E227K induced prominent F-actin accumulations and the consequent rearrangement of focal adhesions, myosin II, and septin filaments and results in a delayed directional migration of the cells. The W148R protein-induced cytoskeletal rearrangement was partially attenuated by the inhibition of myosin II, p21-activated protein kinase, or Rho-associated protein kinase. The expression of a single-head ABD fragment with the mutations partially mimicked the rearrangement induced by the dimer. The F-actin clustering through the interaction with the mutant FLNB ABD may limit the cytoskeletal reorganization, preventing normal skeletal development.
Collapse
Affiliation(s)
- Yongtong Zhao
- Department of Molecular Physiology and Biophysics, Sidney Kimmel Medical College at Thomas Jefferson University, and Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Sandor S Shapiro
- Department of Molecular Physiology and Biophysics, Sidney Kimmel Medical College at Thomas Jefferson University, and Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Masumi Eto
- Department of Molecular Physiology and Biophysics, Sidney Kimmel Medical College at Thomas Jefferson University, and Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
19
|
Bouet G, Marchat D, Cruel M, Malaval L, Vico L. In VitroThree-Dimensional Bone Tissue Models: From Cells to Controlled and Dynamic Environment. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:133-56. [DOI: 10.1089/ten.teb.2013.0682] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Guenaelle Bouet
- Laboratoire de Biologie du Tissu Osseux, Institut National de la Santé et de la Recherche Médicale—U1059, Université de Lyon—Université Jean Monnet, Saint-Etienne, France
| | - David Marchat
- Center for Biomedical and Healthcare Engineering, Ecole Nationale Supérieure des Mines, CIS-EMSE, CNRS:UMR 5307, Saint-Etienne, France
| | - Magali Cruel
- University of Lyon, LTDS, UMR CNRS 5513, Ecole Centrale de Lyon, Ecully, France
| | - Luc Malaval
- Laboratoire de Biologie du Tissu Osseux, Institut National de la Santé et de la Recherche Médicale—U1059, Université de Lyon—Université Jean Monnet, Saint-Etienne, France
| | - Laurence Vico
- Laboratoire de Biologie du Tissu Osseux, Institut National de la Santé et de la Recherche Médicale—U1059, Université de Lyon—Université Jean Monnet, Saint-Etienne, France
| |
Collapse
|
20
|
Altmann B, Löchner A, Swain M, Kohal RJ, Giselbrecht S, Gottwald E, Steinberg T, Tomakidi P. Differences in morphogenesis of 3D cultured primary human osteoblasts under static and microfluidic growth conditions. Biomaterials 2014; 35:3208-19. [DOI: 10.1016/j.biomaterials.2013.12.088] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/22/2013] [Indexed: 11/30/2022]
|
21
|
Jahed Z, Shams H, Mehrbod M, Mofrad MRK. Mechanotransduction pathways linking the extracellular matrix to the nucleus. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 310:171-220. [PMID: 24725427 DOI: 10.1016/b978-0-12-800180-6.00005-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cells contain several mechanosensing components that transduce mechanical signals into biochemical cascades. During cell-ECM adhesion, a complex network of molecules mechanically couples the extracellular matrix (ECM), cytoskeleton, and nucleoskeleton. The network comprises transmembrane receptor proteins and focal adhesions, which link the ECM and cytoskeleton. Additionally, recently identified protein complexes extend this linkage to the nucleus by linking the cytoskeleton and the nucleoskeleton. Despite numerous studies in this field, due to the complexity of this network, our knowledge of the mechanisms of cell-ECM adhesion at the molecular level remains remarkably incomplete. Herein, we present a review of the structures of key molecules involved in cell-ECM adhesion, along with an evaluation of their predicted roles in mechanical sensing. Additionally, specific binding events prompted by force-induced conformational changes of each molecule are discussed. Finally, we propose a model for the biomechanical events prominent in cell-ECM adhesion.
Collapse
Affiliation(s)
- Zeinab Jahed
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California at Berkeley, Berkeley, California, USA
| | - Hengameh Shams
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California at Berkeley, Berkeley, California, USA
| | - Mehrdad Mehrbod
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California at Berkeley, Berkeley, California, USA
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California at Berkeley, Berkeley, California, USA.
| |
Collapse
|
22
|
Dumaual CM, Steere BA, Walls CD, Wang M, Zhang ZY, Randall SK. Integrated analysis of global mRNA and protein expression data in HEK293 cells overexpressing PRL-1. PLoS One 2013; 8:e72977. [PMID: 24019887 PMCID: PMC3760866 DOI: 10.1371/journal.pone.0072977] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 07/17/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The protein tyrosine phosphatase PRL-1 represents a putative oncogene with wide-ranging cellular effects. Overexpression of PRL-1 can promote cell proliferation, survival, migration, invasion, and metastasis, but the underlying mechanisms by which it influences these processes remain poorly understood. METHODOLOGY To increase our comprehension of PRL-1 mediated signaling events, we employed transcriptional profiling (DNA microarray) and proteomics (mass spectrometry) to perform a thorough characterization of the global molecular changes in gene expression that occur in response to stable PRL-1 overexpression in a relevant model system (HEK293). PRINCIPAL FINDINGS Overexpression of PRL-1 led to several significant changes in the mRNA and protein expression profiles of HEK293 cells. The differentially expressed gene set was highly enriched in genes involved in cytoskeletal remodeling, integrin-mediated cell-matrix adhesion, and RNA recognition and splicing. In particular, members of the Rho signaling pathway and molecules that converge on this pathway were heavily influenced by PRL-1 overexpression, supporting observations from previous studies that link PRL-1 to the Rho GTPase signaling network. In addition, several genes not previously associated with PRL-1 were found to be significantly altered by its expression. Most notable among these were Filamin A, RhoGDIα, SPARC, hnRNPH2, and PRDX2. CONCLUSIONS AND SIGNIFICANCE This systems-level approach sheds new light on the molecular networks underlying PRL-1 action and presents several novel directions for future, hypothesis-based studies.
Collapse
Affiliation(s)
- Carmen M. Dumaual
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| | - Boyd A. Steere
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Chad D. Walls
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Mu Wang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Zhong-Yin Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Stephen K. Randall
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, United States of America
| |
Collapse
|
23
|
Mitra J, Tripathi G, Sharma A, Basu B. Scaffolds for bone tissue engineering: role of surface patterning on osteoblast response. RSC Adv 2013. [DOI: 10.1039/c3ra23315d] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
24
|
Macro and microfluidic flows for skeletal regenerative medicine. Cells 2012; 1:1225-45. [PMID: 24710552 PMCID: PMC3901127 DOI: 10.3390/cells1041225] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 11/07/2012] [Accepted: 12/04/2012] [Indexed: 11/16/2022] Open
Abstract
Fluid flow has a great potential as a cell stimulatory tool for skeletal regenerative medicine, because fluid flow-induced bone cell mechanotransduction in vivo plays a critical role in maintaining healthy bone homeostasis. Applications of fluid flow for skeletal regenerative medicine are reviewed at macro and microscale. Macroflow in two dimensions (2D), in which flow velocity varies along the normal direction to the flow, has explored molecular mechanisms of bone forming cell mechanotransduction responsible for flow-regulated differentiation, mineralized matrix deposition, and stem cell osteogenesis. Though 2D flow set-ups are useful for mechanistic studies due to easiness in in situ and post-flow assays, engineering skeletal tissue constructs should involve three dimensional (3D) flows, e.g., flow through porous scaffolds. Skeletal tissue engineering using 3D flows has produced promising outcomes, but 3D flow conditions (e.g., shear stress vs. chemotransport) and scaffold characteristics should further be tailored. Ideally, data gained from 2D flows may be utilized to engineer improved 3D bone tissue constructs. Recent microfluidics approaches suggest a strong potential to mimic in vivo microscale interstitial flows in bone. Though there have been few microfluidics studies on bone cells, it was demonstrated that microfluidic platform can be used to conduct high throughput screening of bone cell mechanotransduction behavior under biomimicking flow conditions.
Collapse
|
25
|
Honjo T, Kubota S, Kamioka H, Sugawara Y, Ishihara Y, Yamashiro T, Takigawa M, Takano-Yamamoto T. Promotion of Ccn2 expression and osteoblastic differentiation by actin polymerization, which is induced by laminar fluid flow stress. J Cell Commun Signal 2012; 6:225-32. [PMID: 22956334 DOI: 10.1007/s12079-012-0177-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 08/17/2012] [Indexed: 01/02/2023] Open
Abstract
Fluid flow stress (FSS) is a major mechanical stress that induces bone remodeling upon orthodontic tooth movement, whereas CCN family protein 2 (CCN2) is a potent regenerator of bone defects. In this study, we initially evaluated the effect of laminar FSS on Ccn2 expression and investigated its mechanism in osteoblastic MC3T3-E1 cells. The Ccn2 expression was drastically induced by uniform FSS in an intensity dependent manner. Of note, the observed effect was inhibited by a Rho kinase inhibitor Y27632. Moreover, the inhibition of actin polymerization blocked the FSS-induced activation of Ccn2, whereas inducing F-actin formation using cytochalasin D and jasplakinolide enhanced Ccn2 expression in the same cells. Finally, F-actin formation was found to induce osteoblastic differentiation. In addition, activation of cyclic AMP-dependent kinase, which inhibits Rho signaling, abolished the effect of FSS. Collectively, these findings indicate the critical role of actin polymerization and Rho signaling in CCN2 induction and bone remodeling provoked by FSS.
Collapse
Affiliation(s)
- Tadashi Honjo
- Department of Orthodontics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Muthukumaran P, Lim CT, Lee T. Estradiol influences the mechanical properties of human fetal osteoblasts through cytoskeletal changes. Biochem Biophys Res Commun 2012; 423:503-8. [PMID: 22683634 DOI: 10.1016/j.bbrc.2012.05.149] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 05/26/2012] [Indexed: 10/28/2022]
Abstract
Estrogen is known to have a direct effect on bone forming osteoblasts and bone resorbing osteoclasts. The cellular and molecular effects of estrogen on osteoblasts and osteoblasts-like cells have been extensively studied. However, the effect of estrogen on the mechanical property of osteoblasts has not been studied yet. It is important since mechanical property of the mechanosensory osteoblasts could be pivotal to its functionality in bone remodeling. This is the first study aimed to assess the direct effect of estradiol on the apparent elastic modulus (E∗) and corresponding cytoskeletal changes of human fetal osteoblasts (hFOB 1.19). The cells were cultured in either medium alone or medium supplemented with β-estradiol and then subjected to Atomic Force Microscopy indentation (AFM) to determine E∗. The underlying changes in cytoskeleton were studied by staining the cells with TRITC-Phalloidin. Following estradiol treatment, the cells were also tested for proliferation, alkaline phosphatase activity and mineralization. With estradiol treatment, E∗ of osteoblasts significantly decreased by 43-46%. The confocal images showed that the changes in f-actin network observed in estradiol treated cells can give rise to the changes in the stiffness of the cells. Estradiol also increases the inherent alkaline phosphatase activity of the cells. Estradiol induced stiffness changes of osteoblasts were not associated with changes in the synthesized mineralized matrix of the cells. Thus, a decrease in osteoblast stiffness with estrogen treatment was demonstrated in this study, with positive links to cytoskeletal changes. The estradiol associated changes in osteoblast mechanical properties could bear implications for bone remodeling and its mechanical integrity.
Collapse
|
27
|
Abstract
Filamins are essential, evolutionarily conserved, modular, multidomain, actin-binding proteins that organize the actin cytoskeleton and maintain extracellular matrix connections by anchoring actin filaments to transmembrane receptors. By cross-linking and anchoring actin filaments, filamins stabilize the plasma membrane, provide cellular cortical rigidity, and contribute to the mechanical stability of the plasma membrane and the cell cortex. In addition to binding actin, filamins interact with more than 90 other binding partners including intracellular signaling molecules, receptors, ion channels, transcription factors, and cytoskeletal and adhesion proteins. Thus, filamins scaffold a wide range of signaling pathways and are implicated in the regulation of a diverse array of cellular functions including motility, maintenance of cell shape, and differentiation. Here, we review emerging structural and functional evidence that filamins are mechanosensors and/or mechanotransducers playing essential roles in helping cells detect and respond to physical forces in their local environment.
Collapse
Affiliation(s)
- Ziba Razinia
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | |
Collapse
|
28
|
Yang N, Schindeler A, McDonald MM, Seto JT, Houweling PJ, Lek M, Hogarth M, Morse AR, Raftery JM, Balasuriya D, MacArthur DG, Berman Y, Quinlan KGR, Eisman JA, Nguyen TV, Center JR, Prince RL, Wilson SG, Zhu K, Little DG, North KN. α-Actinin-3 deficiency is associated with reduced bone mass in human and mouse. Bone 2011; 49:790-8. [PMID: 21784188 DOI: 10.1016/j.bone.2011.07.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 06/28/2011] [Accepted: 07/07/2011] [Indexed: 10/18/2022]
Abstract
Bone mineral density (BMD) is a complex trait that is the single best predictor of the risk of osteoporotic fractures. Candidate gene and genome-wide association studies have identified genetic variations in approximately 30 genetic loci associated with BMD variation in humans. α-Actinin-3 (ACTN3) is highly expressed in fast skeletal muscle fibres. There is a common null-polymorphism R577X in human ACTN3 that results in complete deficiency of the α-actinin-3 protein in approximately 20% of Eurasians. Absence of α-actinin-3 does not cause any disease phenotypes in muscle because of compensation by α-actinin-2. However, α-actinin-3 deficiency has been shown to be detrimental to athletic sprint/power performance. In this report we reveal additional functions for α-actinin-3 in bone. α-Actinin-3 but not α-actinin-2 is expressed in osteoblasts. The Actn3(-/-) mouse displays significantly reduced bone mass, with reduced cortical bone volume (-14%) and trabecular number (-61%) seen by microCT. Dynamic histomorphometry indicated this was due to a reduction in bone formation. In a cohort of postmenopausal Australian women, ACTN3 577XX genotype was associated with lower BMD in an additive genetic model, with the R577X genotype contributing 1.1% of the variance in BMD. Microarray analysis of cultured osteoprogenitors from Actn3(-/-) mice showed alterations in expression of several genes regulating bone mass and osteoblast/osteoclast activity, including Enpp1, Opg and Wnt7b. Our studies suggest that ACTN3 likely contributes to the regulation of bone mass through alterations in bone turnover. Given the high frequency of R577X in the general population, the potential role of ACTN3 R577X as a factor influencing variations in BMD in elderly humans warrants further study.
Collapse
Affiliation(s)
- Nan Yang
- Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Sydney 2145, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Lück S, Fichtl A, Sailer M, Joos H, Brenner RE, Walther P, Schmidt V. Statistical analysis of the intermediate filament network in cells of mesenchymal lineage by greyvalue-oriented image segmentation. Comput Stat 2011. [DOI: 10.1007/s00180-011-0265-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
30
|
Filamin A mediates interactions between cytoskeletal proteins that control cell adhesion. FEBS Lett 2010; 585:18-22. [PMID: 21095189 DOI: 10.1016/j.febslet.2010.11.033] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 11/09/2010] [Accepted: 11/17/2010] [Indexed: 11/22/2022]
Abstract
Cell adhesion, spreading and migration on extracellular matrices are regulated by complex processes that involve the cytoskeleton and a large array of adhesion receptors, including the β1 integrin. Filamin A is a large, multi-domain, homodimeric actin binding protein that contributes to the mechanical stability of cells and interacts with several proteins that regulate cell adhesion including β1 integrin and several protein kinases. Here we review current data on the structure, mechanical properties and intracellular signaling functions of filamin that regulate cell adhesion. We also consider new data showing that interactions of filamin A with intermediate filaments and protein kinase C enable tight regulation of β1 integrin function and consequently early events in cell adhesion and migration on extracellular matrix proteins.
Collapse
|
31
|
McCoy RJ, O'Brien FJ. Influence of shear stress in perfusion bioreactor cultures for the development of three-dimensional bone tissue constructs: a review. TISSUE ENGINEERING PART B-REVIEWS 2010; 16:587-601. [PMID: 20799909 DOI: 10.1089/ten.teb.2010.0370] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Bone tissue engineering aims to generate clinically applicable bone graft substitutes in an effort to ease the demands and reduce the potential risks associated with traditional autograft and allograft bone replacement procedures. Biomechanical stimuli play an important role under physiologically relevant conditions in the normal formation, development, and homeostasis of bone tissue--predominantly, strain (predicted levels in vivo for humans <2000 με) caused by physical deformation, and fluid shear stress (0.8-3 Pa), generated by interstitial fluid movement through lacunae caused by compression and tension under loading. Therefore, in vitro bone tissue cultivation strategies seek to incorporate biochemical stimuli in an effort to create more physiologically relevant constructs for grafting. This review is focused on collating information pertaining to the relationship between fluid shear stress, cellular deformation, and osteogenic differentiation, providing further insight into the optimal culture conditions for the creation of bone tissue substitutes.
Collapse
Affiliation(s)
- Ryan J McCoy
- Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | |
Collapse
|
32
|
Kim H, Nakamura F, Lee W, Hong C, Pérez-Sala D, McCulloch CA. Regulation of cell adhesion to collagen via β1 integrins is dependent on interactions of filamin A with vimentin and protein kinase C epsilon. Exp Cell Res 2010; 316:1829-44. [DOI: 10.1016/j.yexcr.2010.02.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 02/06/2010] [Accepted: 02/08/2010] [Indexed: 12/16/2022]
|
33
|
Pan J, Zhang T, Mi L, Zhang B, Wang B, Yang L, Deng L, Wang L. Stepwise increasing and decreasing fluid shear stresses differentially regulate the functions of osteoblasts. Cell Mol Bioeng 2010; 3:376-386. [PMID: 21603107 DOI: 10.1007/s12195-010-0132-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
It is well accepted that osteoblasts respond to fluid shear stress (FSS) depending on the loading magnitude, rate, and temporal profiles. Although in vivo observations demonstrated that bone mineral density changes as the training intensity gradually increases/decreases, whether osteoblasts perceive such slow temporal changes in the strength of stimulation remains unclear. In this study, we hypothesized that osteoblasts can detect and respond differentially to the temporal gradients of FSS. In specific, we hypothesized that when the temporal FSS gradient is high enough, i) the increasing FSS inhibits the osteoblastic potential in supporting osteoclastogenesis and enhances the osteoblastic anabolic responses; ii) on the other hand, the deceasing FSS would have opposite effects on osteoclastogenesis and anabolic responses. To test the hypotheses, stepwise varying FSS was applied on primary osteoblasts and osteogenic and resorption markers were analyzed. The cells were subjected to FSS increasing from 5, 10, to 15 or decreasing from 15, 10, to 5 dyn/cm(2) at a step of 5 dyn/cm(2) for either 6 or 12 hours. In a subset experiment, the cells were stimulated with stepwise increasing or decreasing FSS at a higher step (10 dyn/cm(2)) for 12 hours. Our results showed that, with the step of 5 dyn/cm(2), the stepwise increasing FSS inhibited the osteoclastogenesis with a 3- to 4-fold decrease in RANKL/OPG gene expression versus static controls, while the stepwise decreasing FSS increased RANKL/OPG ratio by 2- to 2.5-fold versus static controls. Both increasing and decreasing FSS enhanced alkaline phosphatase expression and calcium deposition by 1.0- to 1.8 fold versus static controls. For a higher FSS temporal gradient (three steps of 10 dyn/cm(2) over 12 hour stimulation), the increasing FSS enhanced the expression of alkaline phosphatase expression and calcium deposition by 1.3 fold, while the decreasing FSS slightly inhibited them by -10% compared with static controls. Taken together, our results suggested that osteoblasts can detect the slow temporal gradients of FSS and respond differentially in a dose-dependent manner, which may account for the observed bone mineral density changes in response to the gradual increasing/decreasing exercise in vivo. The stepwise FSS can be a useful model to study bone cell responses to long-term mechanical usage or disuse. These studies will complement the short-term studies and provide additional clinically relevant insights on bone adaptation.
Collapse
Affiliation(s)
- Jun Pan
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, and "National 985 Project" Institute of Biorheology and Gene Regulation, Bioengineering College, Chongqing University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Liu X, Zhang X, Lee I. A quantitative study on morphological responses of osteoblastic cells to fluid shear stress. Acta Biochim Biophys Sin (Shanghai) 2010; 42:195-201. [PMID: 20213044 DOI: 10.1093/abbs/gmq004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Fluid shear stress (FSS) is widely explored regarding its influence on osteoblasts. In vitro studies have shown that the cytoskeleton is very important in cellular responses to FSS. However, morphological changes, which would reflect the cytoskeleton changes as well as other cellular responses, were rarely quantitatively studied in the past years. Therefore, FSS-induced morphological changes in osteoblasts were quantified in this study. Real-time rapid morphological responses were observed by exposing osteoblasts to FSS with magnitude of 1.2, 1.6, and 1.9 Pa for 1 h. Afterward, osteoblast actin cytoskeleton was labeled with rhodamine phalloidin and observed using fluorescence microscopy. The results showed that 1.6 and 1.9 Pa FFS resulted in significant cellular elongation and reorientation along the direction of fluid flow. Besides, along with the enhancement of FSS magnitude, cytoskeleton aggregated more remarkably. Furthermore, extracellular Ca(2+)-depleted fluid flow was also used to stimulate osteoblasts for 1 h with magnitude of 1.6 and 1.9 Pa. No morphological change was observed after removing extracellular calcium. Our study suggested that the level of FSS from 1.2 to 1.9 Pa is capable of influencing cellular morphology, and extracellular calcium might play a role in osteoblasts' response to FSS stimulation.
Collapse
Affiliation(s)
- Xiaoli Liu
- State Key Laboratory of Bioactive Materials, School of Physics, Nankai University, Tianjin 300071, China
| | | | | |
Collapse
|
35
|
Liu J, Zou L, Wang J, Zhao Z. Validation of beta-actin used as endogenous control for gene expression analysis in mechanobiology studies. Stem Cells 2009; 27:2371-2. [PMID: 19551905 DOI: 10.1002/stem.160] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
36
|
Kim H, Nakamura F, Lee W, Shifrin Y, Arora P, McCulloch CA. Filamin A is required for vimentin-mediated cell adhesion and spreading. Am J Physiol Cell Physiol 2009; 298:C221-36. [PMID: 19776392 DOI: 10.1152/ajpcell.00323.2009] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Cell adhesion and spreading are regulated by complex interactions involving the cytoskeleton and extracellular matrix proteins. We examined the interaction of the intermediate filament protein vimentin with the actin cross-linking protein filamin A in regulation of spreading in HEK-293 and 3T3 cells. Filamin A and vimentin-expressing cells were well spread on collagen and exhibited numerous cell extensions enriched with filamin A and vimentin. By contrast, cells treated with small interfering RNA (siRNA) to knock down filamin A or vimentin were poorly spread; both of these cell populations exhibited >50% reductions of cell adhesion, cell surface beta1 integrin expression, and beta1 integrin activation. Knockdown of filamin A reduced vimentin phosphorylation and blocked recruitment of vimentin to cell extensions, whereas knockdown of filamin and/or vimentin inhibited the formation of cell extensions. Reduced vimentin phosphorylation, cell spreading, and beta1 integrin surface expression, and activation were phenocopied in cells treated with the protein kinase C inhibitor bisindolylmaleimide; cell spreading was also reduced by siRNA knockdown of protein kinase C-epsilon. By immunoprecipitation of cell lysates and by pull-down assays using purified proteins, we found an association between filamin A and vimentin. Filamin A also associated with protein kinase C-epsilon, which was enriched in cell extensions. These data indicate that filamin A associates with vimentin and to protein kinase C-epsilon, thereby enabling vimentin phosphorylation, which is important for beta1 integrin activation and cell spreading on collagen.
Collapse
Affiliation(s)
- Hugh Kim
- Canadian Institutes of Health Group in Matrix Dynamics, University of Toronto, Toronto, Ontario, M5S 3E2, Canada.
| | | | | | | | | | | |
Collapse
|
37
|
Krishnan V, Davidovitch Z. On a Path to Unfolding the Biological Mechanisms of Orthodontic Tooth Movement. J Dent Res 2009; 88:597-608. [DOI: 10.1177/0022034509338914] [Citation(s) in RCA: 202] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Orthodontic forces deform the extracellular matrix and activate cells of the paradental tissues, facilitating tooth movement. Discoveries in mechanobiology have illuminated sequential cellular and molecular events, such as signal generation and transduction, cytoskeletal re-organization, gene expression, differentiation, proliferation, synthesis and secretion of specific products, and apoptosis. Orthodontists work in a unique biological environment, wherein applied forces engender remodeling of both mineralized and non-mineralized paradental tissues, including the associated blood vessels and neural elements. This review aims at identifying events that affect the sequence, timing, and significance of factors that determine the nature of the biological response of each paradental tissue to orthodontic force. The results of this literature review emphasize the fact that mechanoresponses and inflammation are both essential for achieving tooth movement clinically. If both are working in concert, orthodontists might be able to accelerate or decelerate tooth movement by adding adjuvant methods, whether physical, chemical, or surgical.
Collapse
Affiliation(s)
- V. Krishnan
- Department of Orthodontics, Rajas Dental College, Tirunelveli District, Tamilnadu, India; and
- Department of Orthodontics, Case Western Reserve University, Cleveland, OH, USA
| | - Z. Davidovitch
- Department of Orthodontics, Rajas Dental College, Tirunelveli District, Tamilnadu, India; and
- Department of Orthodontics, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|