1
|
Guo H, Lan M, Zhang Q, Liu Y, Zhang Y, Zhang Q, Chen W. [Piezo1 Mediates the Regulation of Substrate Stiffness on Primary Cilia in Chondrocytes]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:67-73. [PMID: 38322536 PMCID: PMC10839480 DOI: 10.12182/20240160502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Indexed: 02/08/2024]
Abstract
Objective To investigate how substrate stiffness regulates the morphology of primary cilia in chondrocytes and to illustrate how Piezo1 mediates the morphology regulation of primary cilia by substrate stiffness. Methods Polydimethylsiloxane (PDMS) curing agent and the main agent (Dow Corning, Beijing, China) were mixed at the ratio of 1∶10 (stiff), 1∶50 (medium stiffness), and 1∶70 (soft), respectively, to prepare substrate films with the thickness of 1 mm at different levels of stiffness, including stiff substrate of (2.21±0.12) MPa, medium-stiffness substrate of (54.47±6.06) kPa, and soft substrate of (2.13±0.10) kPa. Chondrocytes were cultured with the substrates of three different levels of stiffness. Then, the cells were treated with Tubastatin A (Tub A) to inhibit histone deacetylase 6 (HDAC6), Piezo1 activator Yoda1, and inhibitor GsMTx4, respectively. The effects of HDAC6, Yoda1, and GsMTx4 on chondrocyte morphology and the length of primary cilia were analyzed through immunofluorescence staining. Results The stiff substrate increased the spread area of the chondrocytes. Immunofluorescence assays showed that the cytoskeleton and the nuclear area of the cells on the stiff substrate were significantly increased (P<0.05) and the primary cilia were significantly extended (P<0.05) compared with those on the medium-stiffness and soft substrates. However, the presence rate of primary cilia was not affected. The HDAC6 activity of chondrocytes increased with the decrease in substrate stiffness. When the activity of HDAC6 was inhibited, the cytoskeletal area, the nuclei area, and the primary cilium length were increased more significantly on the stiff substrate (P<0.05). Further testing showed that Piezo1 activator and inhibitor could regulate the activity of HDAC6 in chondrocytes, and that the length of primary cilia was significantly increased after treatment with the activator Yoda1 (P<0.05). On the other hand, the length of primary cilia was significantly shortened on the stiff substrate after treatment with the inhibitor GsMTx4 (P<0.05). Conclusion Both substrate stiffness and Piezo1 may affect the morphology of chondrocyte primary cilia by regulating HDAC6 activity.
Collapse
Affiliation(s)
- Huaqing Guo
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Minhua Lan
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Qiang Zhang
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yanli Liu
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yanjun Zhang
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- ( 030009) Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030009, China
| | - Quanyou Zhang
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
- ( 030009) Department of Orthopedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030009, China
| | - Weiyi Chen
- ( 030024) College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, China
| |
Collapse
|
2
|
Zhang Y, Tawiah GK, Zhang Y, Wang X, Wei X, Chen W, Qiao X, Zhang Q. HDAC6 inhibition regulates substrate stiffness-mediated inflammation signaling in chondrocytes. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1987-1998. [PMID: 37644773 PMCID: PMC10753363 DOI: 10.3724/abbs.2023144] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/28/2023] [Indexed: 08/31/2023] Open
Abstract
Osteoarthritis (OA) is a chronic disease and is difficult to cure. Chondrocytes are highly mechanosensitive. Therefore, mechanical therapies have received attention as a therapeutic direction for OA. The stiffness, as a critical cue of the extracellular matrix (ECM), affects cell growth, development, and death. In this study, we use polydimethylsiloxane (PDMS) to create substrates with varying stiffness for chondrocyte growth, interleukin-1β (IL-1β) treatment to mimic the inflammatory environment, and Tubastatin A (Tub A) to inhibit histone deacetylase 6 (HDAC6). Our results show that stiff substrates can be anti-inflammatory and provide a better matrix environment than soft substrates. Inhibition of HDAC6 improves the inflammatory environment caused by IL-1β and coordinates with inflammation to spread the chondrocyte area and primary cilia elongation. Without IL-1β and Tub A treatments, the length of the primary cilia rather than frequency is stiffness-dependent, and their length on stiff substrates are greater than that on soft substrates. In conclusion, we demonstrate that stiff substrates, inflammation, and inhibition of HDAC6 enhance the mechanosensitivity of primary cilia and mediate substrate stiffness to suppress inflammation and protect the matrix.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Histology and EmbryologyShanxi Medical UniversityJinzhong030604China
- College of Biomedical EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Godfred K Tawiah
- College of Biomedical EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Yanjun Zhang
- College of Biomedical EngineeringTaiyuan University of TechnologyTaiyuan030024China
- Department of Orthopaedicsthe Second Hospital of Shanxi Medical UniversityShanxi Key Laboratory of Bone and Soft Tissue Injury RepairShanxi Medical UniversityTaiyuan030001China
| | - Xiaohu Wang
- Department of Orthopaedicsthe Second Hospital of Shanxi Medical UniversityShanxi Key Laboratory of Bone and Soft Tissue Injury RepairShanxi Medical UniversityTaiyuan030001China
| | - Xiaochun Wei
- Department of Orthopaedicsthe Second Hospital of Shanxi Medical UniversityShanxi Key Laboratory of Bone and Soft Tissue Injury RepairShanxi Medical UniversityTaiyuan030001China
| | - Weiyi Chen
- College of Biomedical EngineeringTaiyuan University of TechnologyTaiyuan030024China
| | - Xiaohong Qiao
- Department of Histology and EmbryologyShanxi Medical UniversityJinzhong030604China
- Department of OrthopaedicsLvliang Hospital Affiliated to Shanxi Medical UniversityLvliang033099China
| | - Quanyou Zhang
- College of Biomedical EngineeringTaiyuan University of TechnologyTaiyuan030024China
- Department of Orthopaedicsthe Second Hospital of Shanxi Medical UniversityShanxi Key Laboratory of Bone and Soft Tissue Injury RepairShanxi Medical UniversityTaiyuan030001China
| |
Collapse
|
3
|
Pettenuzzo S, Arduino A, Belluzzi E, Pozzuoli A, Fontanella CG, Ruggieri P, Salomoni V, Majorana C, Berardo A. Biomechanics of Chondrocytes and Chondrons in Healthy Conditions and Osteoarthritis: A Review of the Mechanical Characterisations at the Microscale. Biomedicines 2023; 11:1942. [PMID: 37509581 PMCID: PMC10377681 DOI: 10.3390/biomedicines11071942] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Biomechanical studies are expanding across a variety of fields, from biomedicine to biomedical engineering. From the molecular to the system level, mechanical stimuli are crucial regulators of the development of organs and tissues, their growth and related processes such as remodelling, regeneration or disease. When dealing with cell mechanics, various experimental techniques have been developed to analyse the passive response of cells; however, cell variability and the extraction process, complex experimental procedures and different models and assumptions may affect the resulting mechanical properties. For these purposes, this review was aimed at collecting the available literature focused on experimental chondrocyte and chondron biomechanics with direct connection to their biochemical functions and activities, in order to point out important information regarding the planning of an experimental test or a comparison with the available results. In particular, this review highlighted (i) the most common experimental techniques used, (ii) the results and models adopted by different authors, (iii) a critical perspective on features that could affect the results and finally (iv) the quantification of structural and mechanical changes due to a degenerative pathology such as osteoarthritis.
Collapse
Affiliation(s)
- Sofia Pettenuzzo
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
| | - Alessandro Arduino
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
| | - Elisa Belluzzi
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), Via Giustiniani 3, 35128 Padova, Italy
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), 35128 Padova, Italy
| | - Assunta Pozzuoli
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), Via Giustiniani 3, 35128 Padova, Italy
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), 35128 Padova, Italy
| | | | - Pietro Ruggieri
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology, University of Padova (DiSCOG), 35128 Padova, Italy
| | - Valentina Salomoni
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
- Department of Management and Engineering (DTG), Stradella S. Nicola 3, 36100 Vicenza, Italy
| | - Carmelo Majorana
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
| | - Alice Berardo
- Department of Civil, Environmental and Architectural Engineering, University of Padova, 35131 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
4
|
Kroupa KR, Gangi LR, Zimmerman BK, Hung CT, Ateshian GA. Superficial zone chondrocytes can get compacted under physiological loading: A multiscale finite element analysis. Acta Biomater 2023; 163:248-258. [PMID: 36243365 PMCID: PMC10324087 DOI: 10.1016/j.actbio.2022.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/27/2022] [Accepted: 10/05/2022] [Indexed: 11/01/2022]
Abstract
Recent in vivo and in vitro studies have demonstrated that superficial zone (SZ) chondrocytes within articular layers of diarthrodial joints die under normal physiologic loading conditions. In order to further explore the implications of this observation in future investigations, we first needed to understand the mechanical environment of SZ chondrocytes that might cause them to die under physiological sliding contact conditions. In this study we performed a multiscale finite element analysis of articular contact to track the temporal evolution of a SZ chondrocyte's interstitial fluid pressure, hydraulic permeability, and volume under physiologic loading conditions. The effect of the pericellular matrix modulus and permeability was parametrically investigated. Results showed that SZ chondrocytes can lose ninety percent of their intracellular fluid after several hours of intermittent or continuous contact loading, resulting in a reduction of intracellular hydraulic permeability by more than three orders of magnitude. These findings are consistent with loss of cell viability due to the impediment of cellular metabolic pathways induced by the loss of fluid. They suggest that there is a simple mechanical explanation for the vulnerability of SZ chondrocytes to sustained physiological loading conditions. Future studies will focus on validating these specific findings experimentally. STATEMENT OF SIGNIFICANCE: As with any mechanical system, normal 'wear and tear' of cartilage tissue lining joints is expected. Yet incidences of osteoarthritis are uncommon in individuals younger than 45. This counter-intuitive observation suggests there must be an intrinsic repair mechanism compensating for this wear and tear over many decades of life. Recent experimental studies have shown superficial zone chondrocytes die under physiologic loading conditions, suggesting that this repair mechanism may involve cell replenishment. To better understand the mechanical environment of these cells, we performed a multiscale computational analysis of articular contact under loading. Results indicated that normal activities like walking or standing can induce significant loss of intracellular fluid volume, potentially hindering metabolic activity and fluid transport properties, and causing cell death.
Collapse
Affiliation(s)
- Kimberly R Kroupa
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, 220 S.W. Mudd, New York, NY 10027, USA
| | - Lianna R Gangi
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA
| | - Brandon K Zimmerman
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, 220 S.W. Mudd, New York, NY 10027, USA
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA; Department of Orthopedic Surgery, Columbia University, 622 West 168th Street PH 11 - Center, New York, NY 10032, USA
| | - Gerard A Ateshian
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, 220 S.W. Mudd, New York, NY 10027, USA; Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027, USA.
| |
Collapse
|
5
|
Petropoulou K, Platania V, Chatzinikolaidou M, Mitraki A. A Doubly Fmoc-Protected Aspartic Acid Self-Assembles into Hydrogels Suitable for Bone Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2022; 15:8928. [PMID: 36556733 PMCID: PMC9784766 DOI: 10.3390/ma15248928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
Hydrogels have been used as scaffolds for biomineralization in tissue engineering and regenerative medicine for the repair and treatment of many tissue types. In the present work, we studied an amino acid-based material that is attached to protecting groups and self-assembles into biocompatible and stable nanostructures that are suitable for tissue engineering applications. Specifically, the doubly protected aspartic residue (Asp) with fluorenyl methoxycarbonyl (Fmoc) protecting groups have been shown to lead to the formation of well-ordered fibrous structures. Many amino acids and small peptides which are modified with protecting groups display relatively fast self-assembly and exhibit remarkable physicochemical properties leading to three-dimensional (3D) networks, the trapping of solvent molecules, and forming hydrogels. In this study, the self-assembling fibrous structures are targeted toward calcium binding and act as nucleation points for the binding of the available phosphate groups. The cell viability, proliferation, and osteogenic differentiation of pre-osteoblastic cells cultured on the formed hydrogel under various conditions demonstrate that hydrogel formation in CaCl2 and CaCl2-Na2HPO4 solutions lead to calcium ion binding onto the hydrogels and enrichment with phosphate groups, respectively, rendering these mechanically stable hydrogels osteoinductive scaffolds for bone tissue engineering.
Collapse
Affiliation(s)
| | - Varvara Platania
- Department of Materials Science and Technology, University of Crete, 70013 Heraklion, Greece
| | - Maria Chatzinikolaidou
- Department of Materials Science and Technology, University of Crete, 70013 Heraklion, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FO.R.T.H), 70013 Heraklion, Greece
| | - Anna Mitraki
- Department of Materials Science and Technology, University of Crete, 70013 Heraklion, Greece
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FO.R.T.H), 70013 Heraklion, Greece
| |
Collapse
|
6
|
Bashir KMI, Lee S, Jung DH, Basu SK, Cho MG, Wierschem A. Narrow-Gap Rheometry: A Novel Method for Measuring Cell Mechanics. Cells 2022; 11:2010. [PMID: 35805094 PMCID: PMC9265971 DOI: 10.3390/cells11132010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/18/2022] Open
Abstract
The viscoelastic properties of a cell cytoskeleton contain abundant information about the state of a cell. Cells show a response to a specific environment or an administered drug through changes in their viscoelastic properties. Studies of single cells have shown that chemical agents that interact with the cytoskeleton can alter mechanical cell properties and suppress mitosis. This envisions using rheological measurements as a non-specific tool for drug development, the pharmacological screening of new drug agents, and to optimize dosage. Although there exists a number of sophisticated methods for studying mechanical properties of single cells, studying concentration dependencies is difficult and cumbersome with these methods: large cell-to-cell variations demand high repetition rates to obtain statistically significant data. Furthermore, method-induced changes in the cell mechanics cannot be excluded when working in a nonlinear viscoelastic range. To address these issues, we not only compared narrow-gap rheometry with commonly used single cell techniques, such as atomic force microscopy and microfluidic-based approaches, but we also compared existing cell monolayer studies used to estimate cell mechanical properties. This review provides insight for whether and how narrow-gap rheometer could be used as an efficient drug screening tool, which could further improve our current understanding of the mechanical issues present in the treatment of human diseases.
Collapse
Affiliation(s)
- Khawaja Muhammad Imran Bashir
- German Engineering Research and Development Center, LSTME-Busan Branch, Busan 46742, Korea; (K.M.I.B.); (S.L.); (D.H.J.); (M.-G.C.)
| | - Suhyang Lee
- German Engineering Research and Development Center, LSTME-Busan Branch, Busan 46742, Korea; (K.M.I.B.); (S.L.); (D.H.J.); (M.-G.C.)
- Institute of Fluid Mechanics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany;
| | - Dong Hee Jung
- German Engineering Research and Development Center, LSTME-Busan Branch, Busan 46742, Korea; (K.M.I.B.); (S.L.); (D.H.J.); (M.-G.C.)
- Division of Energy and Bioengineering, Dongseo University, Busan 47011, Korea
| | - Santanu Kumar Basu
- Institute of Fluid Mechanics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany;
| | - Man-Gi Cho
- German Engineering Research and Development Center, LSTME-Busan Branch, Busan 46742, Korea; (K.M.I.B.); (S.L.); (D.H.J.); (M.-G.C.)
- Division of Energy and Bioengineering, Dongseo University, Busan 47011, Korea
| | - Andreas Wierschem
- German Engineering Research and Development Center, LSTME-Busan Branch, Busan 46742, Korea; (K.M.I.B.); (S.L.); (D.H.J.); (M.-G.C.)
- Institute of Fluid Mechanics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany;
| |
Collapse
|
7
|
Rothbauer M, Reihs EI, Fischer A, Windhager R, Jenner F, Toegel S. A Progress Report and Roadmap for Microphysiological Systems and Organ-On-A-Chip Technologies to Be More Predictive Models in Human (Knee) Osteoarthritis. Front Bioeng Biotechnol 2022; 10:886360. [PMID: 35782494 PMCID: PMC9240813 DOI: 10.3389/fbioe.2022.886360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022] Open
Abstract
Osteoarthritis (OA), a chronic debilitating joint disease affecting hundreds of million people globally, is associated with significant pain and socioeconomic costs. Current treatment modalities are palliative and unable to stop the progressive degeneration of articular cartilage in OA. Scientific attention has shifted from the historical view of OA as a wear-and-tear cartilage disorder to its recognition as a whole-joint disease, highlighting the contribution of other knee joint tissues in OA pathogenesis. Despite much progress in the field of microfluidic systems/organs-on-a-chip in other research fields, current in vitro models in use do not yet accurately reflect the complexity of the OA pathophenotype. In this review, we provide: 1) a detailed overview of the most significant recent developments in the field of microsystems approaches for OA modeling, and 2) an OA-pathophysiology-based bioengineering roadmap for the requirements of the next generation of more predictive and authentic microscale systems fit for the purpose of not only disease modeling but also of drug screening to potentially allow OA animal model reduction and replacement in the near future.
Collapse
Affiliation(s)
- Mario Rothbauer
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Faculty of Technical Chemistry, Vienna University of Technology, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Eva I. Reihs
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Faculty of Technical Chemistry, Vienna University of Technology, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Anita Fischer
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Reinhard Windhager
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Florien Jenner
- Veterinary Tissue Engineering and Regenerative Medicine Vienna (VETERM), Equine Surgery Unit, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Stefan Toegel
- Karl Chiari Lab for Orthopeadic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| |
Collapse
|
8
|
Willantarra I, Leung S, Choi YS, Chhana A, McGlashan SR. Chondrocyte-specific response to stiffness-mediated primary cilia formation and centriole positioning. Am J Physiol Cell Physiol 2022; 323:C236-C247. [PMID: 35649254 DOI: 10.1152/ajpcell.00135.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mechanical stress and the stiffness of the extracellular matrix are key drivers of tissue development and homeostasis. Aberrant mechanosensation is associated with a wide range of pathologies, including osteoarthritis. Matrix (or substrate) stiffness plays a major role in cell spreading, adhesion, proliferation and differentiation. However, how specific cells sense substrate stiffness still remains unclude. The primary cilium is an essential cellular organelle that senses and integrates mechanical and chemical signals from the extracellular environment. We hypothesised that the primary cilium dynamically alters its length and position to fine-tune cell mechanosignalling based on substrate stiffness alone. We used a hydrogel system of varying substrate stiffness to examine the role of stiffness on cilia frequency, length and centriole position as well as cell and nuclei area over time. Contrary to other cell types, we show that chondrocyte primary cilia shorten on softer substrates demonstrating tissue-specific mechanosensing which is aligned with the tissue stiffness the cells originate from. We further show that stiffness determines centriole positioning to either the basal or apical membrane during attachment and spreading, with centriole positioned towards the basal membrane on stiffer substrates. These phenomena are mediated by force generation actin-myosin stress fibres in a time-dependent manner. Finally we show on stiff substrates, that primary cilia are involved in tension-mediated cell spreading. We propose that substrate stiffness plays a role in cilia positioning, regulating cellular responses to external forces, and may be a key driver of mechanosignalling-associated diseases.
Collapse
Affiliation(s)
- Ivanna Willantarra
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sophia Leung
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Yu Suk Choi
- School of Human Sciences, University of Western Australia, Perth, Australia
| | - Ashika Chhana
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sue R McGlashan
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
9
|
Kim E, Lee H. Mechanical characterization of soft microparticles prepared by droplet microfluidics. JOURNAL OF POLYMER SCIENCE 2022. [DOI: 10.1002/pol.20220110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Eunseo Kim
- Department of Chemical Engineering Pohang University of Science and Technology (POSTECH) Pohang South Korea
| | - Hyomin Lee
- Department of Chemical Engineering Pohang University of Science and Technology (POSTECH) Pohang South Korea
| |
Collapse
|
10
|
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
|
11
|
Khoeini R, Nosrati H, Akbarzadeh A, Eftekhari A, Kavetskyy T, Khalilov R, Ahmadian E, Nasibova A, Datta P, Roshangar L, Deluca DC, Davaran S, Cucchiarini M, Ozbolat IT. Natural and Synthetic Bioinks for 3D Bioprinting. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000097] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Roghayeh Khoeini
- Department of Medicinal Chemistry Faculty of Pharmacy Tabriz University of Medical Sciences P.O. Box: 51664-14766 Tabriz Iran
- Drug Applied Research Center Tabriz University of Medical Sciences P.O. Box: 51656-65811 Tabriz Iran
| | - Hamed Nosrati
- Drug Applied Research Center Tabriz University of Medical Sciences P.O. Box: 51656-65811 Tabriz Iran
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
| | - Abolfazl Akbarzadeh
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Department of Medical Nanotechnology Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences P.O. Box: 516615731 Tabriz Iran
| | - Aziz Eftekhari
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Russian Institute for Advanced Study Moscow State Pedagogical University 1/1, Malaya Pirogovskaya Street Moscow 119991 Russian Federation
- Pharmacology and Toxicology Department Maragheh University of Medical Sciences 78151-55158 Maragheh Iran
- Department of Synthesis and Characterization of Polymers Polymer Institute Slovak Academy of Sciences (SAS) Dúbravská cesta 9 845 41 Bratislava Slovakia
| | - Taras Kavetskyy
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Department of Biology and Chemistry Drohobych Ivan Franko State Pedagogical University 24, I. Franko Str. 82100 Drohobych Ukraine
- Department of Surface Engineering The John Paul II Catholic University of Lublin 20-950 Lublin Poland
| | - Rovshan Khalilov
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Russian Institute for Advanced Study Moscow State Pedagogical University 1/1, Malaya Pirogovskaya Street Moscow 119991 Russian Federation
- Department of Biophysics and Biochemistry Faculty of Biology Baku State University Baku AZ 1143 Azerbaijan
- Institute of Radiation Problems National Academy of Sciences of Azerbaijan Baku AZ 1143 Azerbaijan
| | - Elham Ahmadian
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Kidney Research Center Tabriz University of Medical Sciences P.O. Box: 5166/15731 Tabriz Iran
| | - Aygun Nasibova
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Institute of Radiation Problems National Academy of Sciences of Azerbaijan Baku AZ 1143 Azerbaijan
| | - Pallab Datta
- Department of Pharmaceutics National Institute of Pharmaceutical Education and Research Kolkata West Bengal 700054 India
| | - Leila Roshangar
- Stem Cell Research Center Tabriz University of Medical Sciences P.O. Box: 5166/15731 Tabriz Iran
| | - Dante C. Deluca
- Agricultural and Biological Engineering Department Penn State University University Park 16802 PA USA
| | - Soodabeh Davaran
- Department of Medicinal Chemistry Faculty of Pharmacy Tabriz University of Medical Sciences P.O. Box: 51664-14766 Tabriz Iran
- Drug Applied Research Center Tabriz University of Medical Sciences P.O. Box: 51656-65811 Tabriz Iran
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Department of Medical Nanotechnology Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences P.O. Box: 516615731 Tabriz Iran
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics Saarland University Medical Center Kirrbergerstr. Bldg 37 D-66421 Homburg/Saar Germany
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics Department Penn State University University Park 16802 PA USA
- The Huck Institutes of the Life Sciences Penn State University University Park 16802 PA USA
- Biomedical Engineering Department Penn State University University Park 16802 PA USA
- Materials Research Institute Penn State University University Park 16802 PA USA
- Department of Neurosurgery Penn State University Hershey 17033 PA USA
| |
Collapse
|
12
|
Delineating the heterogeneity of matrix-directed differentiation toward soft and stiff tissue lineages via single-cell profiling. Proc Natl Acad Sci U S A 2021; 118:2016322118. [PMID: 33941688 PMCID: PMC8126831 DOI: 10.1073/pnas.2016322118] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The clinical utility of mesenchymal stromal/stem cells (MSCs) in mediating immunosuppressive effects and supporting regenerative processes is broadly established. However, the inherent heterogeneity of MSCs compromises its biomedical efficacy and reproducibility. To study how cellular variation affects fate decision-making processes, we perform single-cell RNA sequencing at multiple time points during bipotential matrix-directed differentiation toward soft- and stiff tissue lineages. In this manner, we identify distinctive MSC subpopulations that are characterized by their multipotent differentiation capacity and mechanosensitivity. Also, whole-genome screening highlights TPM1 as a potent mechanotransducer of matrix signals and regulator of cell differentiation. Thus, by introducing single-cell methodologies into mechanobiology, we delineate the complexity of adult stem cell responses to extracellular cues in tissue regeneration and immunomodulation. Mesenchymal stromal/stem cells (MSCs) form a heterogeneous population of multipotent progenitors that contribute to tissue regeneration and homeostasis. MSCs assess extracellular elasticity by probing resistance to applied forces via adhesion, cytoskeletal, and nuclear mechanotransducers that direct differentiation toward soft or stiff tissue lineages. Even under controlled culture conditions, MSC differentiation exhibits substantial cell-to-cell variation that remains poorly characterized. By single-cell transcriptional profiling of nonconditioned, matrix-conditioned, and early differentiating cells, we identified distinct MSC subpopulations with distinct mechanosensitivities, differentiation capacities, and cell cycling. We show that soft matrices support adipogenesis of multipotent cells and early endochondral ossification of nonadipogenic cells, whereas intramembranous ossification and preosteoblast proliferation are directed by stiff matrices. Using diffusion pseudotime mapping, we outline hierarchical matrix-directed differentiation and perform whole-genome screening of mechanoresponsive genes. Specifically, top-ranked tropomyosin-1 is highly sensitive to stiffness cues both at RNA and protein levels, and changes in TPM1 expression determine the differentiation toward soft versus stiff tissue lineage. Consistent with actin stress fiber stabilization, tropomyosin-1 overexpression maintains YAP1 nuclear localization, activates YAP1 target genes, and directs osteogenic differentiation. Knockdown of tropomyosin-1 reversed YAP1 nuclear localization consistent with relaxation of cellular contractility, suppressed osteogenesis, activated early endochondral ossification genes after 3 d of culture in induction medium, and facilitated adipogenic differentiation after 1 wk. Our results delineate cell-to-cell variation of matrix-directed MSC differentiation and highlight tropomyosin-mediated matrix sensing.
Collapse
|
13
|
Tabatabaei M, Tafazzoli-Shadpour M, Khani MM. Altered mechanical properties of actin fibers due to breast cancer invasion: parameter identification based on micropipette aspiration and multiscale tensegrity modeling. Med Biol Eng Comput 2021; 59:547-560. [PMID: 33559086 DOI: 10.1007/s11517-021-02318-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 01/15/2021] [Indexed: 12/01/2022]
Abstract
The biophysical properties of cells change with cancer invasion to fulfill their metastatic behavior. Cell softening induced by cancer is highly associated with alterations in cytoskeleton fibers. Changes in the mechanical properties of cytoskeletal fibers have not been quantified due to technical limitations. In this study, we used the micropipette aspiration technique to calculate and compare the viscoelastic properties of non-invasive and invasive breast cancer cells. We evaluated the mechanical properties of actin fibers and microtubules of two cancerous cell lines by using multiscale tensegrity modeling and an optimization method. Cancer invasion caused altered viscoelastic behavior of cells and the results of modeling showed changes in mechanical properties of major cytoskeleton fibers. The stiffness and viscosity constant of actin fibers in non-invasive cells were 1.28 and 2.27 times higher than those of the invasive cells, respectively. However, changes in mechanical properties of microtubules were minor. Immunofluorescent staining of fibers and their quantified distributions confirmed altered actin distribution among two cell lines, in contrast to microtubule distribution. This study highlights the function of cytoskeletal fibers in cancer progression, which could be of interest in designing therapeutic strategies to target cancer progress. Firstly, the viscoelastic behavior of non-invasive and invasive cells is examined with micropipette aspiration tests. A tensegrity model of cells is developed to mimic the viscoelastic behavior of cells, and tensegrity element stiffness is evaluated in an optimization procedure based on micropipette aspiration tests. Finally, by using immunofluorescent staining and confocal imaging, mechanical properties of actin filaments and microtubules of cancer cells are investigated during the course of metastasis.
Collapse
Affiliation(s)
- Mohammad Tabatabaei
- Cardiovascular Engineering Lab, Faculty of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Mohammad Tafazzoli-Shadpour
- Cardiovascular Engineering Lab, Faculty of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Mohammad Mehdi Khani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Schwab A, Levato R, D’Este M, Piluso S, Eglin D, Malda J. Printability and Shape Fidelity of Bioinks in 3D Bioprinting. Chem Rev 2020; 120:11028-11055. [PMID: 32856892 PMCID: PMC7564085 DOI: 10.1021/acs.chemrev.0c00084] [Citation(s) in RCA: 446] [Impact Index Per Article: 111.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Indexed: 12/23/2022]
Abstract
Three-dimensional bioprinting uses additive manufacturing techniques for the automated fabrication of hierarchically organized living constructs. The building blocks are often hydrogel-based bioinks, which need to be printed into structures with high shape fidelity to the intended computer-aided design. For optimal cell performance, relatively soft and printable inks are preferred, although these undergo significant deformation during the printing process, which may impair shape fidelity. While the concept of good or poor printability seems rather intuitive, its quantitative definition lacks consensus and depends on multiple rheological and chemical parameters of the ink. This review discusses qualitative and quantitative methodologies to evaluate printability of bioinks for extrusion- and lithography-based bioprinting. The physicochemical parameters influencing shape fidelity are discussed, together with their importance in establishing new models, predictive tools and printing methods that are deemed instrumental for the design of next-generation bioinks, and for reproducible comparison of their structural performance.
Collapse
Affiliation(s)
- Andrea Schwab
- AO
Research Institute Davos, Clavadelerstrasse 8, 7270 Davos Platz, Switzerland
| | - Riccardo Levato
- Department
of Orthopaedics, University Medical Center
Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Department
of Clinical Sciences, Faculty of Veterinary
Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| | - Matteo D’Este
- AO
Research Institute Davos, Clavadelerstrasse 8, 7270 Davos Platz, Switzerland
| | - Susanna Piluso
- Department
of Orthopaedics, University Medical Center
Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Department
of Developmental BioEngineering, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - David Eglin
- AO
Research Institute Davos, Clavadelerstrasse 8, 7270 Davos Platz, Switzerland
| | - Jos Malda
- Department
of Orthopaedics, University Medical Center
Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
- Department
of Clinical Sciences, Faculty of Veterinary
Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| |
Collapse
|
15
|
Bachmann B, Spitz S, Schädl B, Teuschl AH, Redl H, Nürnberger S, Ertl P. Stiffness Matters: Fine-Tuned Hydrogel Elasticity Alters Chondrogenic Redifferentiation. Front Bioeng Biotechnol 2020; 8:373. [PMID: 32426347 PMCID: PMC7204401 DOI: 10.3389/fbioe.2020.00373] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/03/2020] [Indexed: 12/20/2022] Open
Abstract
Biomechanical cues such as shear stress, stretching, compression, and matrix elasticity are vital in the establishment of next generation physiological in vitro tissue models. Matrix elasticity, for instance, is known to guide stem cell differentiation, influence healing processes and modulate extracellular matrix (ECM) deposition needed for tissue development and maintenance. To better understand the biomechanical effect of matrix elasticity on the formation of articular cartilage analogs in vitro, this study aims at assessing the redifferentiation capacity of primary human chondrocytes in three different hydrogel matrices of predefined matrix elasticities. The hydrogel elasticities were chosen to represent a broad spectrum of tissue stiffness ranging from very soft tissues with a Young’s modulus of 1 kPa up to elasticities of 30 kPa, representative of the perichondral-space. In addition, the interplay of matrix elasticity and transforming growth factor beta-3 (TGF-β3) on the redifferentiation of primary human articular chondrocytes was studied by analyzing both qualitative (viability, morphology, histology) and quantitative (RT-qPCR, sGAG, DNA) parameters, crucial to the chondrotypic phenotype. Results show that fibrin hydrogels of 30 kPa Young’s modulus best guide chondrocyte redifferentiation resulting in a native-like morphology as well as induces the synthesis of physiologic ECM constituents such as glycosaminoglycans (sGAG) and collagen type II. This comprehensive study sheds light onto the mechanobiological impact of matrix elasticity on formation and maintenance of articular cartilage and thus represents a major step toward meeting the need for advanced in vitro tissue models to study both re- and degeneration of articular cartilage.
Collapse
Affiliation(s)
- Barbara Bachmann
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Competence Center MechanoBiology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sarah Spitz
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Barbara Schädl
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Andreas H Teuschl
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
| | - Heinz Redl
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sylvia Nürnberger
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Division of Trauma-Surgery, Department of Orthopedics and Trauma-Surgery, Medical University of Vienna, Vienna, Austria
| | - Peter Ertl
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
16
|
Chondrocyte and Pericellular Matrix Deformation and Strain in the Growth Plate Cartilage Reserve Zone Under Compressive Loading. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/978-3-030-43195-2_43] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
17
|
Pérez-Madrigal MM, Shaw JE, Arno MC, Hoyland JA, Richardson SM, Dove AP. Robust alginate/hyaluronic acid thiol-yne click-hydrogel scaffolds with superior mechanical performance and stability for load-bearing soft tissue engineering. Biomater Sci 2019; 8:405-412. [PMID: 31729512 DOI: 10.1039/c9bm01494b] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hydrogels based on hyaluronic acid (HA) exhibit great potential as tissue engineering (TE) scaffolds as a consequence of their unique biological features. Herein, we examine how the advantages of two natural polymers (i.e. HA and alginate) are combined with the efficiency and rapid nature of the thiol-yne click chemistry reaction to obtain biocompatible matrices with tailored properties. Our injectable click-hydrogels revealed excellent mechanical performance, long-term stability, high cytocompatibility and adequate stiffness for the targeted application. This simple approach yielded HA hydrogels with characteristics that make them suitable for applications as 3D scaffolds to support and promote soft tissue regeneration.
Collapse
Affiliation(s)
| | - Joshua E Shaw
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Maria C Arno
- School of Chemistry, University of Birmingham Edgbaston, Birmingham, B15 2TT, UK.
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK and NIHR Manchester Biomedical Research Centre, Central Manchester Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Stephen M Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| | - Andrew P Dove
- School of Chemistry, University of Birmingham Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
18
|
Osidak EO, Karalkin PA, Osidak MS, Parfenov VA, Sivogrivov DE, Pereira FDAS, Gryadunova AA, Koudan EV, Khesuani YD, Кasyanov VA, Belousov SI, Krasheninnikov SV, Grigoriev TE, Chvalun SN, Bulanova EA, Mironov VA, Domogatsky SP. Viscoll collagen solution as a novel bioink for direct 3D bioprinting. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:31. [PMID: 30830351 DOI: 10.1007/s10856-019-6233-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 02/04/2019] [Indexed: 06/09/2023]
Abstract
Collagen is one of the most promising materials for 3D bioprinting because of its distinguished biocompatibility. Cell-laden constructs made of pure collagen with or without incorporated growth supplements support engineered constructs persistence in culture and are perfectly suitable for grafting. The limiting factor for direct 3D collagen printing was poor printability of collagen solutions, especially admixed with cells or tissue spheroids. In our study, we showed that concentrated solutions of native collagen branded Viscoll were effective as bioinks with high fidelity performance. Viscoll containing 20, 30, or 40 mg/ml collagen were used for direct extrusion 3D bioprinting to form scaffolds appropriate to support spatial arrangement of tissue spheroids into rigid patterns with resolution of 0.5 mm in details. Incorporated cells demonstrated sufficient viability. Associated rheological study showed that good printability of the collagen solutions correlates with their increased storage modulus value, notably exceeding the loss modulus value. The proper combination of these physical parameters could become technological criteria for manufacturing various collagen bioinks for 3D bioprinting.
Collapse
Affiliation(s)
- Egor O Osidak
- Imtek Ltd., 3rd Cherepkovskaya 15A, Moscow, Russia.
- Gamaleya Research Institute of Epidemiology and Microbiology Federal State Budgetary Institution, Ministry of Health of the Russian Federation, Gamalei 18, Moscow, Russia.
| | - Pavel A Karalkin
- Laboratory of Biotechnological Research, 3D Bioprinting Solutions, Kashirskoe Roadway, 68/2, Moscow, Russia
- P. A. Hertsen Moscow Oncology Research Center-branch of FSBI NMRRC of the Ministry of Health of Russia, 3 2nd Botkinsky drive, Moscow, Russia
| | | | - Vladislav A Parfenov
- Laboratory of Biotechnological Research, 3D Bioprinting Solutions, Kashirskoe Roadway, 68/2, Moscow, Russia
| | | | - Frederico D A S Pereira
- Laboratory of Biotechnological Research, 3D Bioprinting Solutions, Kashirskoe Roadway, 68/2, Moscow, Russia
| | - Anna A Gryadunova
- Laboratory of Biotechnological Research, 3D Bioprinting Solutions, Kashirskoe Roadway, 68/2, Moscow, Russia
- Institute for Regenerative Medicine, Ministry of Health of the Russian Federation, Sechenov University, Trubetskaya 8/2, Moscow, Russia
| | - Elizaveta V Koudan
- Laboratory of Biotechnological Research, 3D Bioprinting Solutions, Kashirskoe Roadway, 68/2, Moscow, Russia
| | - Yusef D Khesuani
- Laboratory of Biotechnological Research, 3D Bioprinting Solutions, Kashirskoe Roadway, 68/2, Moscow, Russia
| | | | - Sergei I Belousov
- National Research Center «Kurchatov Institute», Akademika Kurchatova pl.,1, Moscow, Russia
| | | | - Timofei E Grigoriev
- National Research Center «Kurchatov Institute», Akademika Kurchatova pl.,1, Moscow, Russia
| | - Sergey N Chvalun
- National Research Center «Kurchatov Institute», Akademika Kurchatova pl.,1, Moscow, Russia
| | - Elena A Bulanova
- Laboratory of Biotechnological Research, 3D Bioprinting Solutions, Kashirskoe Roadway, 68/2, Moscow, Russia
| | - Vladimir A Mironov
- Laboratory of Biotechnological Research, 3D Bioprinting Solutions, Kashirskoe Roadway, 68/2, Moscow, Russia
- Institute for Regenerative Medicine, Ministry of Health of the Russian Federation, Sechenov University, Trubetskaya 8/2, Moscow, Russia
| | - Sergey P Domogatsky
- Imtek Ltd., 3rd Cherepkovskaya 15A, Moscow, Russia
- Russian Cardiology Research and Production Center Federal State Budgetary Institution, Ministry of Health of the Russian Federation, 3 Cherepkovskaya 15A, Moscow, Russia
| |
Collapse
|
19
|
Ronkainen A, Tanska P, Fick J, Herzog W, Korhonen R. Interrelationship of cartilage composition and chondrocyte mechanics after a partial meniscectomy in the rabbit knee joint – Experimental and numerical analysis. J Biomech 2019; 83:65-75. [DOI: 10.1016/j.jbiomech.2018.11.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 12/17/2022]
|
20
|
Guilak F, Nims RJ, Dicks A, Wu CL, Meulenbelt I. Osteoarthritis as a disease of the cartilage pericellular matrix. Matrix Biol 2018; 71-72:40-50. [PMID: 29800616 DOI: 10.1016/j.matbio.2018.05.008] [Citation(s) in RCA: 275] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/21/2018] [Accepted: 05/21/2018] [Indexed: 01/16/2023]
Abstract
Osteoarthritis is a painful joint disease characterized by progressive degeneration of the articular cartilage as well as associated changes to the subchondral bone, synovium, and surrounding joint tissues. While the effects of osteoarthritis on the cartilage extracellular matrix (ECM) have been well recognized, it is now becoming apparent that in many cases, the onset of the disease may be initially reflected in the matrix region immediately surrounding the chondrocytes, termed the pericellular matrix (PCM). Growing evidence suggests that the PCM - which along with the enclosed chondrocytes are termed the "chondron" - acts as a critical transducer or "filter" of biochemical and biomechanical signals for the chondrocyte, serving to help regulate the homeostatic balance of chondrocyte metabolic activity in response to environmental signals. Indeed, it appears that alterations in PCM properties and cell-matrix interactions, secondary to genetic, epigenetic, metabolic, or biomechanical stimuli, could in fact serve as initiating or progressive factors for osteoarthritis. Here, we discuss recent advances in the understanding of the role of the PCM, with an emphasis on the reciprocity of changes that occur in this matrix region with disease, as well as how alterations in PCM properties could serve as a driver of ECM-based diseases such as osteoarthritis. Further study of the structure, function, and composition of the PCM in normal and diseased conditions may provide new insights into the understanding of the pathogenesis of osteoarthritis, and presumably new therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Farshid Guilak
- Department of Orthopaedic Surgery, Washington University, Saint Louis, MO 63110, United States; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, United States; Department of Biomedical Engineering, Washington University, Saint Louis, MO 63110, United States.
| | - Robert J Nims
- Department of Orthopaedic Surgery, Washington University, Saint Louis, MO 63110, United States; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, United States
| | - Amanda Dicks
- Department of Orthopaedic Surgery, Washington University, Saint Louis, MO 63110, United States; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, United States; Department of Biomedical Engineering, Washington University, Saint Louis, MO 63110, United States
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery, Washington University, Saint Louis, MO 63110, United States; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, United States
| | - Ingrid Meulenbelt
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
21
|
Lee IN, Dobre O, Richards D, Ballestrem C, Curran JM, Hunt JA, Richardson SM, Swift J, Wong LS. Photoresponsive Hydrogels with Photoswitchable Mechanical Properties Allow Time-Resolved Analysis of Cellular Responses to Matrix Stiffening. ACS APPLIED MATERIALS & INTERFACES 2018; 10:7765-7776. [PMID: 29430919 PMCID: PMC5864053 DOI: 10.1021/acsami.7b18302] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/12/2018] [Indexed: 05/03/2023]
Abstract
As cell function and phenotype can be directed by the mechanical characteristics of the surrounding matrix, hydrogels have become important platforms for cell culture systems, with properties that can be tuned by external stimuli, such as divalent cations, enzymatic treatment, and pH. However, many of these stimuli can directly affect cell behavior, making it difficult to distinguish purely mechanical signaling events. This study reports on the development of a hydrogel that incorporates photoswitchable cross-linkers, which can reversibly alter their stiffness upon irradiation with the appropriate wavelength of light. Furthermore, this study reports the response of bone-marrow-derived mesenchymal stem cells (MSCs) on these hydrogels that were stiffened systematically by irradiation with blue light. The substrates were shown to be noncytotoxic, and crucially MSCs were not affected by blue-light exposure. Time-resolved analysis of cell morphology showed characteristic cell spreading and increased aspect ratios in response to greater substrate stiffness. This hydrogel provides a platform to study mechanosignaling in cells responding to dynamic changes in stiffness, offering a new way to study mechanotransduction signaling pathways and biological processes, with implicit changes to tissue mechanics, such as development, ageing, and fibrosis.
Collapse
Affiliation(s)
- I-Ning Lee
- Manchester Institute
of Biotechnology and School of Chemistry, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
- School
of Engineering, University of Liverpool, Harrison Hughes Building, Liverpool L69 3GH, United Kingdom
| | - Oana Dobre
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - David Richards
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Judith M. Curran
- School
of Engineering, University of Liverpool, Harrison Hughes Building, Liverpool L69 3GH, United Kingdom
| | - John A. Hunt
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, United Kingdom
| | - Stephen M. Richardson
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Joe Swift
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
- Division
of Cell Matrix Biology and Regenerative Medicine, School of Biological
Sciences, Faculty of Biology, Medicine and Health, Manchester Academic
Health Science Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Lu Shin Wong
- Manchester Institute
of Biotechnology and School of Chemistry, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
22
|
Xia Y, Darling EM, Herzog W. Functional properties of chondrocytes and articular cartilage using optical imaging to scanning probe microscopy. J Orthop Res 2018; 36:620-631. [PMID: 28975657 PMCID: PMC5839958 DOI: 10.1002/jor.23757] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 09/20/2017] [Indexed: 02/04/2023]
Abstract
Mature chondrocytes in adult articular cartilage vary in number, size, and shape, depending on their depth in the tissue, location in the joint, and source species. Chondrocytes are the primary structural, functional, and metabolic unit in articular cartilage, the loss of which will induce fatigue to the extracellular matrix (ECM), eventually leading to failure of the cartilage and impairment of the joint as a whole. This brief review focuses on the functional and biomechanical studies of chondrocytes and articular cartilage, using microscopic imaging from optical microscopies to scanning probe microscopy. Three topics are covered in this review, including the functional studies of chondrons by optical imaging (unpolarized and polarized light and infrared light, two-photon excitation microscopy), the probing of chondrocytes and cartilage directly using microscale measurement techniques, and different imaging approaches that can measure chondrocyte mechanics and chondrocyte biological signaling under in situ and in vivo environments. Technical advancement in chondrocyte research during recent years has enabled new ways to study the biomechanical and functional properties of these cells and cartilage. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:620-631, 2018.
Collapse
Affiliation(s)
- Yang Xia
- Dept of Physics and Center for Biomedical Research, Oakland University, Rochester, MI 48309, USA
| | - Eric M. Darling
- Dept of Molecular Pharmacology, Physiology, and Biotechnology, School of Engineering, Dept of Orthopaedics, Center for Biomedical Engineering, Brown University, Providence, RI 02912, USA
| | - Walter Herzog
- Faculties of Kinesiology, Engineering and Medicine, University of Calgary, AB T2T 1N4, Canada
| |
Collapse
|
23
|
Theoretically proposed optimal frequency for ultrasound induced cartilage restoration. Theor Biol Med Model 2017; 14:21. [PMID: 29132387 PMCID: PMC5684760 DOI: 10.1186/s12976-017-0067-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 09/28/2017] [Indexed: 01/01/2023] Open
Abstract
Background Matching the frequency of the driving force to that of the system’s natural frequency of vibration results in greater amplitude response. Thus we hypothesize that applying ultrasound at the chondrocyte’s resonant frequency will result in greater deformation than applying similar ultrasound power at a frequency outside of the resonant bandwidth. Based on this resonant hypothesis, our group previously confirmed theoretically and experimentally that ultrasound stimulation of suspended chondrocytes at resonance (5 MHz) maximized gene expression of load inducible genes. However, this study was based on suspended chondrocytes. The resonant frequency of a chondrocyte does not only depend on the cell mass and intracellular stiffness, but also on the mechanical properties of the surrounding medium. An in vivo chondrocyte’s environment differs whether it be a blood clot (following microfracture), a hydrogel or the pericellular and extracellular matrices of the natural cartilage. All have distinct structures and compositions leading to different resonant frequencies. In this study, we present two theoretical models, the first model to understand the effects of the resonant frequency on the cellular deformation and the second to identify the optimal frequency range for clinical applications of ultrasound to enhance cartilage restoration. Results We showed that applying low-intensity ultrasound at the resonant frequency induced deformation equivalent to that experimentally calculated in previous studies at higher intensities and a 1 MHz frequency. Additionally, the resonant frequency of an in vivo chondrocyte in healthy conditions, osteoarthritic conditions, embedded in a blood clot and embedded in fibrin ranges from 3.5 − 4.8 MHz. Conclusion The main finding of this study is the theoretically proposed optimal frequency for clinical applications of therapeutic ultrasound induced cartilage restoration is 3.5 − 4.8 MHz (the resonant frequencies of in vivo chondrocytes). Application of ultrasound in this frequency range will maximize desired bioeffects.
Collapse
|
24
|
Angel PM, Narmoneva DA, Sewell-Loftin MK, Munjal C, Dupuis L, Landis BJ, Jegga A, Kern CB, Merryman WD, Baldwin HS, Bressan GM, Hinton RB. Proteomic Alterations Associated with Biomechanical Dysfunction are Early Processes in the Emilin1 Deficient Mouse Model of Aortic Valve Disease. Ann Biomed Eng 2017; 45:2548-2562. [PMID: 28812215 DOI: 10.1007/s10439-017-1899-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 08/08/2017] [Indexed: 12/13/2022]
Abstract
Aortic valve (AV) disease involves stiffening of the AV cusp with progression characterized by inflammation, fibrosis, and calcification. Here, we examine the relationship between biomechanical valve function and proteomic changes before and after the development of AV pathology in the Emilin1-/- mouse model of latent AV disease. Biomechanical studies were performed to quantify tissue stiffness at the macro (micropipette) and micro (atomic force microscopy (AFM)) levels. Micropipette studies showed that the Emilin1-/- AV annulus and cusp regions demonstrated increased stiffness only after the onset of AV disease. AFM studies showed that the Emilin1-/- cusp stiffens before the onset of AV disease and worsens with the onset of disease. Proteomes from AV cusps were investigated to identify protein functions, pathways, and interaction network alterations that occur with age- and genotype-related valve stiffening. Protein alterations due to Emilin1 deficiency, including changes in pathways and functions, preceded biomechanical aberrations, resulting in marked depletion of extracellular matrix (ECM) proteins interacting with TGFB1, including latent transforming growth factor beta 3 (LTBP3), fibulin 5 (FBLN5), and cartilage intermediate layer protein 1 (CILP1). This study identifies proteomic dysregulation is associated with biomechanical dysfunction as early pathogenic processes in the Emilin1-/- model of AV disease.
Collapse
Affiliation(s)
- P M Angel
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - D A Narmoneva
- Division of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - M K Sewell-Loftin
- Division of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - C Munjal
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, MLC 7020, Cincinnati, OH, 45229, USA
| | - L Dupuis
- Department of Regenerative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - B J Landis
- Division of Pediatric Cardiology, Indiana University, Indianapolis, IN, USA
| | - A Jegga
- Division of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - C B Kern
- Department of Regenerative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - W D Merryman
- Division of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - H S Baldwin
- Division of Pediatric Cardiology, Vanderbilt University, Nashville, TN, USA
| | - G M Bressan
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Robert B Hinton
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, MLC 7020, Cincinnati, OH, 45229, USA.
| |
Collapse
|
25
|
Ivanovska IL, Swift J, Spinler K, Dingal D, Cho S, Discher DE. Cross-linked matrix rigidity and soluble retinoids synergize in nuclear lamina regulation of stem cell differentiation. Mol Biol Cell 2017; 28:2010-2022. [PMID: 28566555 PMCID: PMC5541850 DOI: 10.1091/mbc.e17-01-0010] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/09/2017] [Accepted: 05/26/2017] [Indexed: 12/30/2022] Open
Abstract
A nanofilm of cross-linked collagen-I is equivalent to a relatively stiff matrix, which stiffens the nucleus, correlating broadly with lamin-A (including mutant progerin), retinoic acid transcription factor level and activity, and osteoinduction. In vitro results are supported by studies of ectopic bone formation in vivo. Synergistic cues from extracellular matrix and soluble factors are often obscure in differentiation. Here the rigidity of cross-linked collagen synergizes with retinoids in the osteogenesis of human marrow mesenchymal stem cells (MSCs). Collagen nanofilms serve as a model matrix that MSCs can easily deform unless the film is enzymatically cross-linked, which promotes the spreading of cells and the stiffening of nuclei as both actomyosin assembly and nucleoskeletal lamin-A increase. Expression of lamin-A is known to be controlled by retinoic acid receptor (RAR) transcription factors, but soft matrix prevents any response to any retinoids. Rigid matrix is needed to induce rapid nuclear accumulation of the RARG isoform and for RARG-specific antagonist to increase or maintain expression of lamin-A as well as for RARG-agonist to repress expression. A progerin allele of lamin-A is regulated in the same manner in iPSC-derived MSCs. Rigid matrices are further required for eventual expression of osteogenic markers, and RARG-antagonist strongly drives lamin-A–dependent osteogenesis on rigid substrates, with pretreated xenografts calcifying in vivo to a similar extent as native bone. Proteomics-detected targets of mechanosensitive lamin-A and retinoids underscore the convergent synergy of insoluble and soluble cues in differentiation.
Collapse
Affiliation(s)
- Irena L Ivanovska
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Joe Swift
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Kyle Spinler
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Dave Dingal
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Sangkyun Cho
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Dennis E Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
26
|
Salinas D, Minor CA, Carlson RP, McCutchen CN, Mumey BM, June RK. Combining Targeted Metabolomic Data with a Model of Glucose Metabolism: Toward Progress in Chondrocyte Mechanotransduction. PLoS One 2017; 12:e0168326. [PMID: 28056047 PMCID: PMC5215894 DOI: 10.1371/journal.pone.0168326] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/30/2016] [Indexed: 11/19/2022] Open
Abstract
Osteoarthritis is a debilitating disease likely involving altered metabolism of the chondrocytes in articular cartilage. Chondrocytes can respond metabolically to mechanical loads via cellular mechanotransduction, and metabolic changes are significant because they produce the precursors to the tissue matrix necessary for cartilage health. However, a comprehensive understanding of how energy metabolism changes with loading remains elusive. To improve our understanding of chondrocyte mechanotransduction, we developed a computational model to calculate the rate of reactions (i.e. flux) across multiple components of central energy metabolism based on experimental data. We calculated average reaction flux profiles of central metabolism for SW1353 human chondrocytes subjected to dynamic compression for 30 minutes. The profiles were obtained solving a bounded variable linear least squares problem, representing the stoichiometry of human central energy metabolism. Compression synchronized chondrocyte energy metabolism. These data are consistent with dynamic compression inducing early time changes in central energy metabolism geared towards more active protein synthesis. Furthermore, this analysis demonstrates the utility of combining targeted metabolomic data with a computational model to enable rapid analysis of cellular energy utilization.
Collapse
Affiliation(s)
- Daniel Salinas
- Computer Science, Montana State University, Bozeman, MT United States of America
| | - Cody A. Minor
- Mathematics, Montana State University, Bozeman, MT United States of America
| | - Ross P. Carlson
- Chemical & Biological Engineering, Montana State University, Bozeman, MT United States of America
| | - Carley N. McCutchen
- Mechanical & Industrial Engineering, Montana State University, Bozeman, MT United States of America
| | - Brendan M. Mumey
- Computer Science, Montana State University, Bozeman, MT United States of America
| | - Ronald K. June
- Mechanical & Industrial Engineering, Montana State University, Bozeman, MT United States of America
- Department of Cell Biology & Neurosciences, Montana State University, Bozeman, MT United States of America
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA United States of America
- * E-mail:
| |
Collapse
|
27
|
Zhang Q, Yu Y, Zhao H. The effect of matrix stiffness on biomechanical properties of chondrocytes. Acta Biochim Biophys Sin (Shanghai) 2016; 48:958-965. [PMID: 27590061 DOI: 10.1093/abbs/gmw087] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 06/16/2016] [Indexed: 01/14/2023] Open
Abstract
The behavior of chondrocytes is regulated by multiple mechanical microenvironmental cues. During development and degenerative disease of articular cartilage, as an external signal, the extracellular matrix stiffness of chondrocytes changes significantly, but whether and how this biophysical cue affects biomechanical properties of chondrocytes remain elusive. In the present study, we designed supporting-biomaterials as mimics of native pericellular matrix to study the effect of matrix stiffness on chondrocyte morphology and F-actin distribution. Furthermore, the active mechanical behavior of chondrocytes during sensing and responding to different matrix stiffness was quantitatively investigated using atom force microscope technique and theoretical model. Our results indicated that stiffer matrix tends to increase the cell spreading area, the percentage of irregular cell shape distribution and mechanical parameters including elastic modulus (Eelastic), instantaneous modulus (E0), relaxed modulus (ER) and apparent viscosity (μ) of chondrocytes. Knowledge of matrix stiffness-dependent biomechanical behaviors of chondrocytes has important implications for optimizing matrix material and advancing chondrocyte-based applications for functional tissue engineering.
Collapse
Affiliation(s)
- Quanyou Zhang
- Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China College of Mechanics, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yang Yu
- Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
| | - Hucheng Zhao
- Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
28
|
Ronkainen A, Fick J, Herzog W, Korhonen R. Site-specific cell-tissue interactions in rabbit knee joint articular cartilage. J Biomech 2016; 49:2882-2890. [DOI: 10.1016/j.jbiomech.2016.06.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/03/2016] [Accepted: 06/24/2016] [Indexed: 10/21/2022]
|
29
|
Extracellular matrix stiffness dictates Wnt expression through integrin pathway. Sci Rep 2016; 6:20395. [PMID: 26854061 PMCID: PMC4745056 DOI: 10.1038/srep20395] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 01/04/2016] [Indexed: 12/23/2022] Open
Abstract
It is well established that extracellular matrix (ECM) stiffness plays a significant role in regulating the phenotypes and behaviors of many cell types. However, the mechanism underlying the sensing of mechanical cues and subsequent elasticity-triggered pathways remains largely unknown. We observed that stiff ECM significantly enhanced the expression level of several members of the Wnt/β-catenin pathway in both bone marrow mesenchymal stem cells and primary chondrocytes. The activation of β-catenin by stiff ECM is not dependent on Wnt signals but is elevated by the activation of integrin/ focal adhesion kinase (FAK) pathway. The accumulated β-catenin then bound to the wnt1 promoter region to up-regulate the gene transcription, thus constituting a positive feedback of the Wnt/β-catenin pathway. With the amplifying effect of positive feedback, this integrin-activated β-catenin/Wnt pathway plays significant roles in mediating the enhancement of Wnt signal on stiff ECM and contributes to the regulation of mesenchymal stem cell differentiation and primary chondrocyte phenotype maintenance. The present integrin-regulated Wnt1 expression and signaling contributes to the understanding of the molecular mechanisms underlying the regulation of cell behaviors by ECM elasticity.
Collapse
|
30
|
Guo H, Torzilli PA. Shape of chondrocytes within articular cartilage affects the solid but not the fluid microenvironment under unconfined compression. Acta Biomater 2016; 29:170-179. [PMID: 26525115 DOI: 10.1016/j.actbio.2015.10.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/14/2015] [Accepted: 10/20/2015] [Indexed: 10/22/2022]
Abstract
Metabolic activity of the chondrocytes in articular cartilage is strongly related to their zone-specific shape and the composition and mechanical properties of their surrounding extracellular matrix (ECM). However the mechanisms by which cell shape influences the response of the ECM microenvironment to mechanical loading is yet to be elucidated. This relationship was studied using a biphasic multiscale finite element model of different shaped chondrocytes in the superficial and deep zones of the ECM during unconfined stress relaxation. For chondrocytes in the superficial zone, increasing the cell's initial aspect ratio (length/height) increased the deformation and solid stresses of the chondrocyte and pericellular matrix (PCM) during the loading phase; for chondrocytes in the deep zone the effect of the cell shape on the solid microenvironment was time and variable dependent. However, for superficial and deep zone chondrocytes the cell shape did not affect the fluid pressure and fluid shear stress. These results suggest that mechanotransduction of chondrocytes in articular cartilage may be regulated through the solid phase rather than the fluid phase, and that high stresses and deformations in the solid microenvironment in the superficial zone may be essential for the zone-specific biosynthetic activity of the chondrocyte. The biphasic multiscale computational analysis suggests that maintaining the cell shape is critical for regulating the microenvironment and metabolic activity of the chondrocyte in tissue engineering constructs. STATEMENT OF SIGNIFICANCE We investigated the effect of chondrocyte shape on the cellular microenvironment using a biphasic multiscale finite element analysis. Our study showed that cell shapes affects the solid but not the fluid microenvironment of the chondrocyte, and that maintaining the cell shape is critical for regulating the microenvironment and metabolic activity of the chondrocyte in native cartilage and tissue engineering constructs. As far as we know, this is the first study on the mechanotransduction mechanisms by which cell shape influences the response of the microenvironment to mechanical loading. This study is important for understanding cell mechanobiology, not only for regulation of cell phenotype in tissue engineered constructs but, as important, for understanding changes in normal chondrocyte function after post-traumatic injury and in the initiation and progression of osteoarthritis.
Collapse
|
31
|
Akhmanova M, Osidak E, Domogatsky S, Rodin S, Domogatskaya A. Physical, Spatial, and Molecular Aspects of Extracellular Matrix of In Vivo Niches and Artificial Scaffolds Relevant to Stem Cells Research. Stem Cells Int 2015; 2015:167025. [PMID: 26351461 PMCID: PMC4553184 DOI: 10.1155/2015/167025] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/07/2015] [Accepted: 06/24/2015] [Indexed: 12/27/2022] Open
Abstract
Extracellular matrix can influence stem cell choices, such as self-renewal, quiescence, migration, proliferation, phenotype maintenance, differentiation, or apoptosis. Three aspects of extracellular matrix were extensively studied during the last decade: physical properties, spatial presentation of adhesive epitopes, and molecular complexity. Over 15 different parameters have been shown to influence stem cell choices. Physical aspects include stiffness (or elasticity), viscoelasticity, pore size, porosity, amplitude and frequency of static and dynamic deformations applied to the matrix. Spatial aspects include scaffold dimensionality (2D or 3D) and thickness; cell polarity; area, shape, and microscale topography of cell adhesion surface; epitope concentration, epitope clustering characteristics (number of epitopes per cluster, spacing between epitopes within cluster, spacing between separate clusters, cluster patterns, and level of disorder in epitope arrangement), and nanotopography. Biochemical characteristics of natural extracellular matrix molecules regard diversity and structural complexity of matrix molecules, affinity and specificity of epitope interaction with cell receptors, role of non-affinity domains, complexity of supramolecular organization, and co-signaling by growth factors or matrix epitopes. Synergy between several matrix aspects enables stem cells to retain their function in vivo and may be a key to generation of long-term, robust, and effective in vitro stem cell culture systems.
Collapse
Affiliation(s)
| | - Egor Osidak
- Imtek Limited, 3 Cherepkovskaya 15, Moscow 21552, Russia
- Gamaleya Research Institute of Epidemiology and Microbiology Federal State Budgetary Institution, Ministry of Health of the Russian Federation, Gamalei 18, Moscow 123098, Russia
| | - Sergey Domogatsky
- Imtek Limited, 3 Cherepkovskaya 15, Moscow 21552, Russia
- Russian Cardiology Research and Production Center Federal State Budgetary Institution, Ministry of Health of the Russian Federation, 3 Cherepkovskaya 15, Moscow 21552, Russia
| | - Sergey Rodin
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Anna Domogatskaya
- Division of Matrix Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden
| |
Collapse
|
32
|
Tanska P, Mononen ME, Korhonen RK. A multi-scale finite element model for investigation of chondrocyte mechanics in normal and medial meniscectomy human knee joint during walking. J Biomech 2015; 48:1397-406. [PMID: 25795269 DOI: 10.1016/j.jbiomech.2015.02.043] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 02/17/2015] [Indexed: 10/23/2022]
Abstract
Mechanical signals experienced by chondrocytes (articular cartilage cells) modulate cell synthesis and cartilage health. Multi-scale modeling can be used to study how forces are transferred from joint surfaces through tissues to chondrocytes. Therefore, estimation of chondrocyte behavior during certain physical activities, such as walking, could provide information about how cells respond to normal and abnormal loading in joints. In this study, a 3D multi-scale model was developed for evaluating chondrocyte and surrounding peri- and extracellular matrix responses during gait loading within healthy and medial meniscectomy knee joints. The knee joint geometry was based on MRI, whereas the input used for gait loading was obtained from the literature. Femoral and tibial cartilages were modeled as fibril-reinforced poroviscoelastic materials, whereas menisci were considered as transversely isotropic. Fluid pressures in the chondrocyte and cartilage tissue increased up to 2MPa (an increase of 30%) in the meniscectomy joint compared to the normal, healthy joint. The elevated level of fluid pressure was observed during the entire stance phase of gait. A medial meniscectomy caused substantially larger (up to 60%) changes in maximum principal strains in the chondrocyte compared to those in the peri- or extracellular matrices. Chondrocyte volume or morphology did not change substantially due to a medial meniscectomy. Current findings suggest that during walking chondrocyte deformations are not substantially altered due to a medial meniscectomy, while abnormal joint loading exposes chondrocytes to elevated levels of fluid pressure and maximum principal strains (compared to strains in the peri- or extracellular matrices). These might contribute to cell viability and the onset of osteoarthritis.
Collapse
Affiliation(s)
- Petri Tanska
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland; Diagnostic Imaging Centre, Kuopio University Hospital, Kuopio, Finland.
| | - Mika E Mononen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Rami K Korhonen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland; Diagnostic Imaging Centre, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
33
|
Wilusz RE, Sanchez-Adams J, Guilak F. The structure and function of the pericellular matrix of articular cartilage. Matrix Biol 2014; 39:25-32. [PMID: 25172825 DOI: 10.1016/j.matbio.2014.08.009] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chondrocytes in articular cartilage are surrounded by a narrow pericellular matrix (PCM) that is both biochemically and biomechanically distinct from the extracellular matrix (ECM) of the tissue. While the PCM was first observed nearly a century ago, its role is still under investigation. In support of early hypotheses regarding its function, increasing evidence indicates that the PCM serves as a transducer of biochemical and biomechanical signals to the chondrocyte. Work over the past two decades has established that the PCM in adult tissue is defined biochemically by several molecular components, including type VI collagen and perlecan. On the other hand, the biomechanical properties of this structure have only recently been measured. Techniques such as micropipette aspiration, in situ imaging, computational modeling, and atomic force microscopy have determined that the PCM exhibits distinct mechanical properties as compared to the ECM, and that these properties are influenced by specific PCM components as well as disease state. Importantly, the unique relationships among the mechanical properties of the chondrocyte, PCM, and ECM in different zones of cartilage suggest that this region significantly influences the stress-strain environment of the chondrocyte. In this review, we discuss recent advances in the measurement of PCM mechanical properties and structure that further increase our understanding of PCM function. Taken together, these studies suggest that the PCM plays a critical role in controlling the mechanical environment and mechanobiology of cells in cartilage and other cartilaginous tissues, such as the meniscus or intervertebral disc.
Collapse
Affiliation(s)
- Rebecca E Wilusz
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC, USA; Department of Biomedical Engineering, Duke University Medical Center, Durham, NC, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Johannah Sanchez-Adams
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC, USA; Department of Biomedical Engineering, Duke University Medical Center, Durham, NC, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC, USA; Department of Biomedical Engineering, Duke University Medical Center, Durham, NC, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
34
|
Krishnamurthy VK, Evans AN, Wansapura JP, Osinska H, Maddy KE, Biechler SV, Narmoneva DA, Goodwin RL, Hinton RB. Asymmetric cell-matrix and biomechanical abnormalities in elastin insufficiency induced aortopathy. Ann Biomed Eng 2014; 42:2014-28. [PMID: 25099772 DOI: 10.1007/s10439-014-1072-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/14/2014] [Indexed: 01/28/2023]
Abstract
Aortopathy is characterized by vascular smooth muscle cell (VSMC) abnormalities and elastic fiber fragmentation. Elastin insufficient (Eln (+/-)) mice demonstrate latent aortopathy similar to human disease. We hypothesized that aortopathy manifests primarily in the aorto-pulmonary septal (APS) side of the thoracic aorta due to asymmetric cardiac neural crest (CNC) distribution. Anatomic (aortic root vs. ascending aorta) and molecular (APS vs. non-APS) regions of proximal aorta tissue were examined in adult and aged wild type (WT) and mutant (Eln (+/-)) mice. CNC, VSMCs, elastic fiber architecture, proteoglycan expression, morphometrics and biomechanical properties were examined using histology, 3D reconstruction, micropipette aspiration and in vivo magnetic resonance imaging (MRI). In the APS side of Eln (+/-) aorta, Sonic Hedgehog (SHH) is decreased while SM22 is increased. Elastic fiber architecture abnormalities are present in the Eln (+/-) aortic root and APS ascending aorta, and biglycan is increased in the aortic root while aggrecan is increased in the APS aorta. The Eln (+/-) ascending aorta is stiffer than the aortic root, the APS side is thicker and stiffer than the non-APS side, and significant differences in the individual aortic root sinuses are observed. Asymmetric structure-function abnormalities implicate regional CNC dysregulation in the development and progression of aortopathy.
Collapse
Affiliation(s)
- Varun K Krishnamurthy
- Division of Cardiology, the Heart Institute, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, MLC 7020, Cincinnati, OH, 45229, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Dingal PCDP, Discher DE. Material control of stem cell differentiation: challenges in nano-characterization. Curr Opin Biotechnol 2014; 28:46-50. [DOI: 10.1016/j.copbio.2013.11.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 10/31/2013] [Accepted: 11/01/2013] [Indexed: 11/25/2022]
|
36
|
de Vries SAH, van Turnhout MC, Oomens CWJ, Erdemir A, Ito K, van Donkelaar CC. Deformation thresholds for chondrocyte death and the protective effect of the pericellular matrix. Tissue Eng Part A 2014; 20:1870-6. [PMID: 24438476 DOI: 10.1089/ten.tea.2013.0436] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In cartilage tissue engineering studies, the stimulatory effect of mechanical perturbation declines after the first 2 weeks of culture. Similarly, it is known that chondrocyte-agarose constructs should not be loaded within the first days after seeding, to prevent considerable cell death, suggesting a mechanical threshold. This study aims to establish a relationship between chondrocyte deformation and death, and to evaluate the protective effect of the pericellular matrix (PCM) that is formed in 3D cultures. Chondrocyte viability was monitored every hour for 24 h after applying a strain range of 0% to 25% to agarose constructs containing chondrocytes, cultured for 1, 3, 5, 7, or 10 days. At these culture time points, the PCM thickness and chondrocyte deformation were assessed by means of histology and assayed for biochemical contents. Inverse finite element (FE) simulations were used to evaluate the change of mechanical properties of the chondrocyte and PCM over the 10-day culture duration. Chondrocyte death was demonstrated to be dependent on both the magnitude and duration of straining. The highest cell death was observed at day 1 (43%), reducing over culture duration (15% at day 3 and 2.5% at day 10). Cell deformation at 25% compression decreased significantly over culture duration (aspect ratio of 2.24±0.67 at day 1 and 1.45±0.24 at day 3) and with increased matrix production. Inverse FE simulations showed an increasing PCM Young's modulus of 45 kPa at day 3 to 162 kPa at day 10. The current results provide evidence for a mechanical threshold for chondrocyte death and for the protective effect of the PCM. As such, these insights may help in establishing mechanical loading protocols for cartilage tissue engineering studies.
Collapse
Affiliation(s)
- Stefan A H de Vries
- 1 Department of Biomedical Engineering, Eindhoven University of Technology , Eindhoven, The Netherlands
| | | | | | | | | | | |
Collapse
|
37
|
Guo H, Maher SA, Torzilli PA. A biphasic multiscale study of the mechanical microenvironment of chondrocytes within articular cartilage under unconfined compression. J Biomech 2014; 47:2721-9. [PMID: 24882738 DOI: 10.1016/j.jbiomech.2014.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 04/01/2014] [Accepted: 05/03/2014] [Indexed: 10/25/2022]
Abstract
Computational analyses have been used to study the biomechanical microenvironment of the chondrocyte that cannot be assessed by in vitro experimental studies; yet all computational studies thus far have focused on the effect of zonal location (superficial, middle, and deep) on the mechanical microenvironment of chondrocytes. The aim of this paper was to study the effect of both zonal and radial locations on the biomechanical microenvironment of chondrocytes in inhomogeneous cartilage under unconfined stress relaxation. A biphasic multiscale approach was employed and nine chondrocytes in different locations were studied. Hyperelastic biphasic theory and depth-dependent aggregate modulus and permeability of articular cartilage were included in the models. It was found that both zonal and radial locations affected the biomechanical stresses and strains of the chondrocytes. Chondrocytes in the mid-radial location had increased volume during the early stage of the loading process. Maximum principal shear stress at the interface between the chondrocyte and the extracellular matrix (ECM) increased with depth, yet that at the ECM-pericellular matrix (PCM) interface had an inverse trend. Fluid pressure decreased with depth, while the fluid pressure difference between the top and bottom boundaries of the microscale model increased with depth. Regardless of location, fluid was exchanged between the chondrocyte, PCM, and ECM. These findings suggested that even under simple compressive loading conditions, the biomechanical microenvironment of the chondrocytes, PCM and ECM was spatially dependent. The current study provides new insight on chondrocyte biomechanics.
Collapse
Affiliation(s)
- Hongqiang Guo
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA.
| | - Suzanne A Maher
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| | - Peter A Torzilli
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
| |
Collapse
|
38
|
Wilusz RE, Zauscher S, Guilak F. Micromechanical mapping of early osteoarthritic changes in the pericellular matrix of human articular cartilage. Osteoarthritis Cartilage 2013; 21:1895-903. [PMID: 24025318 PMCID: PMC3856176 DOI: 10.1016/j.joca.2013.08.026] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Revised: 08/26/2013] [Accepted: 08/29/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a degenerative joint disease characterized by the progressive loss of articular cartilage. While macroscale degradation of the cartilage extracellular matrix (ECM) has been extensively studied, microscale changes in the chondrocyte pericellular matrix (PCM) and immediate microenvironment with OA are not fully understood. The objective of this study was to quantify osteoarthritic changes in the micromechanical properties of the ECM and PCM of human articular cartilage in situ using atomic force microscopy (AFM). METHOD AFM elastic mapping was performed on cryosections of human cartilage harvested from both condyles of macroscopically normal and osteoarthritic knee joints. This method was used to test the hypotheses that both ECM and PCM regions exhibit a loss of mechanical properties with OA and that the size of the PCM is enlarged in OA cartilage as compared to normal tissue. RESULTS Significant decreases were observed in both ECM and PCM moduli of 45% and 30%, respectively, on the medial condyle of OA knee joints as compared to cartilage from macroscopically normal joints. Enlargement of the PCM, as measured biomechanically, was also observed in medial condyle OA cartilage, reflecting the underlying distribution of type VI collagen in the region. No significant differences were observed in elastic moduli or their spatial distribution on the lateral condyle between normal and OA joints. CONCLUSION Our findings provide new evidence of significant site-specific degenerative changes in the chondrocyte micromechanical environment with OA.
Collapse
Affiliation(s)
- Rebecca E. Wilusz
- Department of Orthopaedic Surgery, Duke University Medical Center
- Department of Biomedical Engineering, Duke University
| | - Stefan Zauscher
- Department of Mechanical Engineering and Materials Science, Duke University
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Duke University Medical Center
- Department of Biomedical Engineering, Duke University
- Department of Mechanical Engineering and Materials Science, Duke University
| |
Collapse
|
39
|
Wilusz RE, Guilak F. High resistance of the mechanical properties of the chondrocyte pericellular matrix to proteoglycan digestion by chondroitinase, aggrecanase, or hyaluronidase. J Mech Behav Biomed Mater 2013; 38:183-97. [PMID: 24156881 DOI: 10.1016/j.jmbbm.2013.09.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 09/09/2013] [Accepted: 09/20/2013] [Indexed: 11/18/2022]
Abstract
In articular cartilage, the extracellular matrix (ECM) and chondrocyte-associated pericellular matrix (PCM) are characterized by a high concentration of proteoglycans (PGs) and their associated glycosaminoglycans (GAGs). These molecules serve important biochemical, structural, and biomechanical roles in the tissue and differences in their regional distributions suggest that different GAG/PG species contribute to the specific biomechanical properties of the ECM and PCM. The objective of this study was to investigate region-specific contributions of aggrecan, chondroitin and dermatan sulfate, and hyaluronan to the micromechanical properties of articular cartilage PCM and ECM in situ. Cryosections of porcine cartilage underwent digestion with ADAMTS-4, chondroitinase ABC, bacterial hyaluronidase or human leukocyte elastase. Guided by immunofluorescence for type VI collagen, AFM stiffness mapping was used to evaluate the elastic properties of matched PCM and ECM regions in paired control and digested cartilage sections. These methods were used to test the hypotheses that specific enzymatic digestion of GAGs or PGs would reduce both PCM and ECM elastic moduli. Elastase, which digests a number of PGs, some types of collagen, and non-collagenous proteins, was used as a positive control. ECM elastic moduli were significantly reduced by all enzyme treatments. However, PCM micromechanical properties were unaffected by enzymatic digestion of aggrecan, chondroitin/dermatan sulfate, and hyaluronan but were significantly reduced by 24% following elastase digestion. Our results provide new evidence for high resistance of PCM micromechanical properties to PG digestion and suggest a potential role for elastase in the degradation of the ECM and PCM.
Collapse
Affiliation(s)
- Rebecca E Wilusz
- Department of Orthopaedic Surgery, Duke University Medical Center, USA; Department of Biomedical Engineering, Duke University, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Duke University Medical Center, USA; Department of Biomedical Engineering, Duke University, USA.
| |
Collapse
|
40
|
Chen C, Tambe DT, Deng L, Yang L. Biomechanical properties and mechanobiology of the articular chondrocyte. Am J Physiol Cell Physiol 2013; 305:C1202-8. [PMID: 24067919 DOI: 10.1152/ajpcell.00242.2013] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To withstand physiological loading over a lifetime, human synovial joints are covered and protected by articular cartilage, a layer of low-friction, load-bearing tissue. The unique mechanical function of articular cartilage largely depends on the composition and structural integrity of the cartilage matrix. The matrix is produced by highly specialized resident cells called chondrocytes. Under physiological loading, chondrocytes maintain the balance between degradation and synthesis of matrix macromolecules. Under excessive loading or injury, however, degradation exceeds synthesis, causing joint degeneration and, eventually, osteoarthritis (OA). Hence, the mechanoresponses of chondrocytes play an important role in the development of OA. Despite its clear importance, the mechanobiology of articular chondrocytes is not well understood. To summarize our current understanding, here we review studies of the effect of mechanical forces on mechanical and biological properties of articular chondrocytes. First, we present the viscoelastic properties of the cell nucleus, chondrocyte, pericellular matrix, and chondron. Then we discuss how these properties change in OA. Finally, we discuss the responses of normal and osteoarthritic chondrocytes to a variety of mechanical stimuli. Studies reviewed here may provide novel insights into the pathogenesis of OA and may help in development of effective biophysical treatment.
Collapse
Affiliation(s)
- Cheng Chen
- Center for Joint Surgery, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | |
Collapse
|
41
|
Swift J, Ivanovska IL, Buxboim A, Harada T, Dingal PCDP, Pinter J, Pajerowski JD, Spinler KR, Shin JW, Tewari M, Rehfeldt F, Speicher DW, Discher DE. Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 2013; 341:1240104. [PMID: 23990565 DOI: 10.1126/science.1240104] [Citation(s) in RCA: 1327] [Impact Index Per Article: 120.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tissues can be soft like fat, which bears little stress, or stiff like bone, which sustains high stress, but whether there is a systematic relationship between tissue mechanics and differentiation is unknown. Here, proteomics analyses revealed that levels of the nucleoskeletal protein lamin-A scaled with tissue elasticity, E, as did levels of collagens in the extracellular matrix that determine E. Stem cell differentiation into fat on soft matrix was enhanced by low lamin-A levels, whereas differentiation into bone on stiff matrix was enhanced by high lamin-A levels. Matrix stiffness directly influenced lamin-A protein levels, and, although lamin-A transcription was regulated by the vitamin A/retinoic acid (RA) pathway with broad roles in development, nuclear entry of RA receptors was modulated by lamin-A protein. Tissue stiffness and stress thus increase lamin-A levels, which stabilize the nucleus while also contributing to lineage determination.
Collapse
Affiliation(s)
- Joe Swift
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Khani MM, Tafazzoli-Shadpour M, Rostami M, Peirovi H, Janmaleki M. Evaluation of mechanical properties of human mesenchymal stem cells during differentiation to smooth muscle cells. Ann Biomed Eng 2013; 42:1373-80. [PMID: 23949657 DOI: 10.1007/s10439-013-0889-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/06/2013] [Indexed: 01/21/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are multipotent cells appropriate for a variety of tissue engineering and cell therapy applications. Mechanical properties of hMSCs during differentiation are associated with their particular metabolic activity and regulate cell function due to alternations in cytoskeleton and structural elements. The objective of this study is to evaluate elastic and viscoelastic properties of hMSCs during long term cultivation in control and transforming growth factor-β1 treatment groups using micropipette aspiration technique. The mean Young's modulus (E) of the control samples remained nearly unchanged during 6 days of cultivation, but that of the test samples showed an initial reduction compared to its relevant control sample after 2 days of treatment by biological growth factor, followed by a significant rise after 4 and 6 days. The viscoelastic creep tests showed that both instantaneous and equilibrium moduli significantly increased with the treatment time and reached to maximum values of 622.9 ± 114.2 and 144.3 ± 11.6 Pa at the sixth day, respectively, while increase in apparent viscosity was not statistically significant. Such change of mechanical properties of hMSCs during specific lineage commitment contributes to regenerative medicine as well as stem-cell-based therapy in which biophysical signals regulate stem cell fate.
Collapse
Affiliation(s)
- Mohammad-Mehdi Khani
- Faculty of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | | | | | | | | |
Collapse
|
43
|
Dowling EP, Ronan W, McGarry JP. Computational investigation of in situ chondrocyte deformation and actin cytoskeleton remodelling under physiological loading. Acta Biomater 2013; 9:5943-55. [PMID: 23271042 DOI: 10.1016/j.actbio.2012.12.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 12/14/2012] [Accepted: 12/17/2012] [Indexed: 12/22/2022]
Abstract
Previous experimental studies have determined local strain fields for both healthy and degenerate cartilage tissue during mechanical loading. However, the biomechanical response of chondrocytes in situ, in particular the response of the actin cytoskeleton to physiological loading conditions, is poorly understood. In the current study a three-dimensional (3-D) representative volume element (RVE) for cartilage tissue is created, comprising a chondrocyte surrounded by a pericellular matrix and embedded in an extracellular matrix. A 3-D active modelling framework incorporating actin cytoskeleton remodelling and contractility is implemented to predict the biomechanical behaviour of chondrocytes. Physiological and abnormal strain fields, based on the experimental study of Wong and Sah (J. Orthop. Res. 2010; 28: 1554-1561), are applied to the RVE. Simulations demonstrate that the presence of a focal defect significantly affects cellular deformation, increases the stress experienced by the nucleus, and alters the distribution of the actin cytoskeleton. It is demonstrated that during dynamic loading cyclic tension reduction in the cytoplasm causes continuous dissociation of the actin cytoskeleton. In contrast, during static loading significant changes in cytoplasm tension are not predicted and hence the rate of dissociation of the actin cytoskeleton is reduced. It is demonstrated that chondrocyte behaviour is affected by the stiffness of the pericellular matrix, and also by the anisotropy of the extracellular matrix. The findings of the current study are of particular importance in understanding the biomechanics underlying experimental observations such as actin cytoskeleton dissociation during the dynamic loading of chondrocytes.
Collapse
Affiliation(s)
- Enda P Dowling
- Mechanical and Biomedical Engineering, National University of Ireland-Galway, Galway, Ireland
| | | | | |
Collapse
|
44
|
|
45
|
Madden R, Han SK, Herzog W. Chondrocyte deformation under extreme tissue strain in two regions of the rabbit knee joint. J Biomech 2012; 46:554-60. [PMID: 23089458 DOI: 10.1016/j.jbiomech.2012.09.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 09/15/2012] [Accepted: 09/20/2012] [Indexed: 11/18/2022]
Abstract
Articular cartilage and its native cells-chondrocytes-are exposed to a wide range of mechanical loading. Chondrocytes are responsible for maintaining the cartilage matrix, yet relatively little is known regarding their behavior under a complete range of mechanical loads or how cell mechanics are affected by region within the joint. The purpose of this study was to investigate chondrocyte deformations in situ under tissue loads ranging from physiological to extreme (0-80% nominal strain) in two regions of the rabbit knee joint (femoral condyles and patellae). Local matrix strains and cell compressive strains increased with increasing loads. At low loads the extracellular matrix (ECM) strains in the superficial zone were greater than the applied tissue strains, while at extreme loads, the local ECM strains were smaller than the applied strains. Cell compressive strains were always smaller than the applied tissue strains and, in our intact, in situ preparation, were substantially smaller than those previously found in hemi-cylindrical explants. This resulted in markedly different steady-state cell volume changes in the current study compared to those working with cartilage explants. Additionally, cells from different regions in the knee exhibited striking differences in deformation behavior under load. The current results suggest: (i) that the local extracellular and pericellular matrix environment is intimately linked to chondrocyte mechanobiology, protecting chondrocytes from potentially damaging strains at high tissue loads; and (ii) that cell mechanics are a function of applied load and local cartilage tissue structure.
Collapse
Affiliation(s)
- Ryan Madden
- Department of Biomedical Engineering, University of Calgary, Canada.
| | | | | |
Collapse
|
46
|
McLeod MA, Wilusz RE, Guilak F. Depth-dependent anisotropy of the micromechanical properties of the extracellular and pericellular matrices of articular cartilage evaluated via atomic force microscopy. J Biomech 2012; 46:586-92. [PMID: 23062866 DOI: 10.1016/j.jbiomech.2012.09.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2012] [Revised: 08/21/2012] [Accepted: 09/07/2012] [Indexed: 01/30/2023]
Abstract
The extracellular matrix (ECM) of articular cartilage is structurally and mechanically inhomogeneous and anisotropic, exhibiting variations in composition, collagen fiber architecture, and pericellular matrix (PCM) morphology among the different zones (superficial, middle, and deep). Joint loading exposes chondrocytes to a complex biomechanical environment, as the microscale mechanical environment of the chondrocyte depends on the relative properties of its PCM and local ECM. ECM anisotropy and chondrocyte deformation are influenced by the split-line direction, the preferred collagen fiber orientation parallel to the articular surface. While previous studies have demonstrated that cartilage macroscale properties vary with depth and the direction of loading relative to the split-line direction, the potential anisotropic behavior of the ECM and PCM at the microscale has yet to be examined. The goal of this study was to characterize the depth and directional dependence of the microscale biomechanical properties of porcine cartilage ECM and PCM in situ. Cartilage was cryosectioned to generate samples oriented parallel and perpendicular to the split-line direction and normal to the articular surface. Atomic force microscopy (AFM)-based stiffness mapping was utilized to measure ECM and PCM microscale elastic properties in all three directions within each zone. Distinct anisotropy in ECM elastic moduli was observed in the superficial and deep zones, while the middle zone exhibited subtle anisotropy. PCM elastic moduli exhibited zonal uniformity with depth and directional dependence when pooled across the zones. These findings provide new evidence for mechanical inhomogeneity and anisotropy at the microscale in articular cartilage.
Collapse
Affiliation(s)
- Morgan A McLeod
- Departments of Orthopaedic Surgery and Biomedical Engineering, Duke University, Durham, NC, USA
| | | | | |
Collapse
|
47
|
Wilusz RE, DeFrate LE, Guilak F. Immunofluorescence-guided atomic force microscopy to measure the micromechanical properties of the pericellular matrix of porcine articular cartilage. J R Soc Interface 2012; 9:2997-3007. [PMID: 22675162 DOI: 10.1098/rsif.2012.0314] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The pericellular matrix (PCM) is a narrow region that is rich in type VI collagen that surrounds each chondrocyte within the extracellular matrix (ECM) of articular cartilage. Previous studies have demonstrated that the chondrocyte micromechanical environment depends on the relative properties of the chondrocyte, its PCM and the ECM. The objective of this study was to measure the influence of type VI collagen on site-specific micromechanical properties of cartilage in situ by combining atomic force microscopy stiffness mapping with immunofluorescence imaging of PCM and ECM regions in cryo-sectioned tissue samples. This method was used to test the hypotheses that PCM biomechanical properties correlate with the presence of type VI collagen and are uniform with depth from the articular surface. Control experiments verified that immunolabelling did not affect the properties of the ECM or PCM. PCM biomechanical properties correlated with the presence of type VI collagen, and matrix regions lacking type VI collagen immediately adjacent to the PCM exhibited higher elastic moduli than regions positive for type VI collagen. PCM elastic moduli were similar in all three zones. Our findings provide further support for type VI collagen in defining the chondrocyte PCM and contributing to its biological and biomechanical properties.
Collapse
Affiliation(s)
- Rebecca E Wilusz
- Department of Orthopaedic Surgery, Duke University Medical Center, Box 3093, Durham, NC 27710, USA
| | | | | |
Collapse
|
48
|
López E, Madero L, López-Pascual J, Latterich M. Clinical proteomics and OMICS clues useful in translational medicine research. Proteome Sci 2012; 10:35. [PMID: 22642823 PMCID: PMC3536680 DOI: 10.1186/1477-5956-10-35] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 05/04/2012] [Indexed: 12/21/2022] Open
Abstract
Since the advent of the new proteomics era more than a decade ago, large-scale studies of protein profiling have been used to identify distinctive molecular signatures in a wide array of biological systems, spanning areas of basic biological research, clinical diagnostics, and biomarker discovery directed toward therapeutic applications. Recent advances in protein separation and identification techniques have significantly improved proteomic approaches, leading to enhancement of the depth and breadth of proteome coverage. Proteomic signatures, specific for multiple diseases, including cancer and pre-invasive lesions, are emerging. This article combines, in a simple manner, relevant proteomic and OMICS clues used in the discovery and development of diagnostic and prognostic biomarkers that are applicable to all clinical fields, thus helping to improve applications of clinical proteomic strategies for translational medicine research.
Collapse
Affiliation(s)
- Elena López
- Centro de Investigación i + 12, Hospital 12 de Octubre, Av, De Córdoba s/n, 28040, Madrid, Spain.
| | | | | | | |
Collapse
|
49
|
Sibole SC, Erdemir A. Chondrocyte deformations as a function of tibiofemoral joint loading predicted by a generalized high-throughput pipeline of multi-scale simulations. PLoS One 2012; 7:e37538. [PMID: 22649535 PMCID: PMC3359292 DOI: 10.1371/journal.pone.0037538] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 04/23/2012] [Indexed: 01/08/2023] Open
Abstract
Cells of the musculoskeletal system are known to respond to mechanical loading and chondrocytes within the cartilage are not an exception. However, understanding how joint level loads relate to cell level deformations, e.g. in the cartilage, is not a straightforward task. In this study, a multi-scale analysis pipeline was implemented to post-process the results of a macro-scale finite element (FE) tibiofemoral joint model to provide joint mechanics based displacement boundary conditions to micro-scale cellular FE models of the cartilage, for the purpose of characterizing chondrocyte deformations in relation to tibiofemoral joint loading. It was possible to identify the load distribution within the knee among its tissue structures and ultimately within the cartilage among its extracellular matrix, pericellular environment and resident chondrocytes. Various cellular deformation metrics (aspect ratio change, volumetric strain, cellular effective strain and maximum shear strain) were calculated. To illustrate further utility of this multi-scale modeling pipeline, two micro-scale cartilage constructs were considered: an idealized single cell at the centroid of a 100×100×100 μm block commonly used in past research studies, and an anatomically based (11 cell model of the same volume) representation of the middle zone of tibiofemoral cartilage. In both cases, chondrocytes experienced amplified deformations compared to those at the macro-scale, predicted by simulating one body weight compressive loading on the tibiofemoral joint. In the 11 cell case, all cells experienced less deformation than the single cell case, and also exhibited a larger variance in deformation compared to other cells residing in the same block. The coupling method proved to be highly scalable due to micro-scale model independence that allowed for exploitation of distributed memory computing architecture. The method's generalized nature also allows for substitution of any macro-scale and/or micro-scale model providing application for other multi-scale continuum mechanics problems.
Collapse
Affiliation(s)
- Scott C. Sibole
- Computational Biomodeling (CoBi) Core and Department of Biomedical Engineering Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ahmet Erdemir
- Computational Biomodeling (CoBi) Core and Department of Biomedical Engineering Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| |
Collapse
|
50
|
Krishnamurthy VK, Opoka AM, Kern CB, Guilak F, Narmoneva DA, Hinton RB. Maladaptive matrix remodeling and regional biomechanical dysfunction in a mouse model of aortic valve disease. Matrix Biol 2012; 31:197-205. [PMID: 22265892 DOI: 10.1016/j.matbio.2012.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 12/21/2011] [Accepted: 01/03/2012] [Indexed: 12/12/2022]
Abstract
Aortic valve disease (AVD) occurs in 2.5% of the general population and often requires surgical intervention. Aortic valve malformation (AVM) underlies the majority of cases, suggesting a developmental etiology. Elastin haploinsufficiency results in complex cardiovascular problems, and 20-45% of patients have AVM and/or AVD. Elastin insufficient (Eln+/-) mice demonstrate AVM and latent AVD due to abnormalities in the valve annulus region. The objective of this study was to examine extracellular matrix (ECM) remodeling and biomechanical properties in regional aortic valve tissue and determine the impact of early AVM on late AVD in the Eln+/- mouse model. Aortic valve ECM composition and remodeling from juvenile, adult, and aged stages were evaluated in Eln+/- mice using histology, ELISA, immunohistochemistry and gelatin zymography. Aortic valve tissue biomechanical properties were determined using micropipette aspiration. Cartilage-like nodules were demonstrated within the valve annulus region at all stages identifying a developmental abnormality preceding AVD. Interestingly, maladaptive ECM remodeling was observed in early AVM without AVD and worsened with late AVD, as evidenced by increased MMP-2 and MMP-9 expression and activity, as well as abnormalities in ADAMTS-mediated versican processing. Cleaved versican was increased in the valve annulus region of aged Eln+/- mice, and this abnormality correlated temporally with adverse alterations in valve tissue biomechanical properties and the manifestation of AVD. These findings identify maladaptive ECM remodeling in functional AVM as an early disease process with a progressive natural history, similar to that seen in human AVD, emphasizing the importance of the annulus region in pathogenesis. Combining molecular and engineering approaches provides complementary mechanistic insights that may be informative in the search for new therapeutic targets and durable valve bioprostheses.
Collapse
Affiliation(s)
- Varun K Krishnamurthy
- Division of Cardiology, the Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | |
Collapse
|