1
|
Stöckl S, Taheri S, Maier V, Asid A, Toelge M, Clausen-Schaumann H, Schilling A, Grässel S. Effects of intra-articular applied rat BMSCs expressing alpha-calcitonin gene-related peptide or substance P on osteoarthritis pathogenesis in a murine surgical osteoarthritis model. Stem Cell Res Ther 2025; 16:117. [PMID: 40045368 PMCID: PMC11884178 DOI: 10.1186/s13287-025-04155-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/21/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND About 655 million persons worldwide are affected by osteoarthritis (OA). As no therapy modifies disease progression long-term, there is an immense clinical need for novel therapies. The joints are innervated by alpha calcitonin gene-related peptide (αCGRP)- and substance P (SP)-positive sensory nerve fibers. Both neuropeptides have trophic effects on target cells within the joints. The aim of this study was to examine the effects of SP- and αCGRP-expressing intra-articular (i.a.) applied rat(r)BMSC on cartilage and subchondral bone structural changes after OA induction. METHODS Mice were subjected to destabilization of the medial meniscus (DMM) surgery, followed by i.a. injections with rBMSC, transduced with lacZ, SP or αCGRP. 2, 8 and 16 weeks after DMM/Sham surgery, motion analysis and serum marker analysis were performed. Cartilage and subchondral bone properties were assessed by OA scoring, atomic force microscopy and nano-CT analysis. RESULTS OARSI scores of the medial cartilage compartments indicated induction and progression of OA after DMM surgery in all groups. Differences between the treatment groups were mostly restricted to the lateral cartilage compartments, where αCGRP caused a decrease of structural changes. DMM-rBMSC-αCGRP or -SP mice displayed decreased cartilage stiffness in the cartilage middle zone. DMM-rBMSC-αCGRP mice revealed improved mobility, whereas Sham-rBMSC-SP mice revealed reduced mobility compared to rBMSC-lacZ. With respect to condyle length, subarticular bone and ephiphyseal bone morphology, DMM-rBMSC-SP mice had more alterations indicating either a more progressed OA stage or a more severe OA pathology compared to controls. In addition, DMM-rBMSC-SP mice developed osteophytes already 8 weeks after surgery. Adiponectin serum level was increased in DMM-rBMSC-αCGRP mice, and MIP1b level in DMM-rBMSC-SP mice. Notably, pain and inflammation markers increased over time in rBMSC-SP mice while rBMSC-αCGRP mice revealed a bell-shaped curve with a peak at 8 weeks. CONCLUSIONS We conclude that i.a. injection of rBMSC in general have a beneficial effect on cartilage matrix structure, subchondral bone microarchitecture and inflammation. rBMSC-αCGRP have anabolic and possible analgesic properties and may attenuate the progression or severity of OA. In contrast, rBMSC-SP exert a more catabolic influence on knee joints of both, Sham and DMM mice, making it a potential candidate for inhibition studies.
Collapse
Affiliation(s)
- Sabine Stöckl
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology, University of Regensburg, ZMB im Biopark, 1 Am Biopark 9, 93053, Regensburg, Germany
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Regensburg, Regensburg, Germany
| | - Shahed Taheri
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medicine Göttingen, Lower Saxony, Germany
| | - Verena Maier
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), University of Applied Sciences Munich, Munich, Germany
| | - Amir Asid
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology, University of Regensburg, ZMB im Biopark, 1 Am Biopark 9, 93053, Regensburg, Germany
| | - Martina Toelge
- Department of Medical Microbiology and Hygiene, University of Regensburg, Regensburg, Germany
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), University of Applied Sciences Munich, Munich, Germany
| | - Arndt Schilling
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medicine Göttingen, Lower Saxony, Germany
| | - Susanne Grässel
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology, University of Regensburg, ZMB im Biopark, 1 Am Biopark 9, 93053, Regensburg, Germany.
| |
Collapse
|
2
|
Kontomaris SV, Malamou A, Stylianou A. Development of an accurate simplified approach for data processing in AFM indentation experiments. Micron 2025; 190:103782. [PMID: 39799615 DOI: 10.1016/j.micron.2024.103782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/15/2025]
Abstract
Atomic Force Microscopy (AFM) nanoindentation is the most effective method for determining the mechanical properties of soft biological materials and biomaterials at the nanoscale, with significant applications in many areas, including cancer diagnosis. However, a major drawback of this method is the complexity of the experimental procedure and data processing, which requires several calibration steps.To avoid this complexity, the AFM tip is usually approximated as a perfect cone. In this case, F=ch2, where F is the applied force, ℎ is the indentation depth, and c is a constant that depends on both the cone's half-angle and the material's properties. However, since AFM tips are pyramidal with a rounded tip apex (or similar to a truncated cone in some cases), the conical approximation may lead to non-negligible errors. Although equations exist that relate the applied force, indentation depth, and the sample's Young's modulus for real indenters, they are rarely used because they do not directly relate the applied force to the indentation depth (i.e., the fitting process is much more complicated compared to the conical approximation). In this paper, a new, accurate, simplified approach for data processing is proposed, based on fitting the force-indentation data to a quadratic equation of the form: F=c2h2+c1h. It is proven that the parameter c2 is independent of the tip apex properties. On the other hand, the parameter c1 depends on the material properties, the cone's half angle, and the shape and dimensions of the tip apex. Simulated force-indentation data from sphero-conical and blunted pyramidal indenters, along with real experimental data from lung tissues, are processed using the proposed approach. The key result is that Young's modulus can be accurately determined using only the c2 parameter; therefore, tip characterization can be avoided.
Collapse
Affiliation(s)
- S V Kontomaris
- School of Sciences, European University Cyprus, Nicosia 2404, Cyprus; Department of Engineering and Construction, Metropolitan College, Athens 15125, Greece.
| | - A Malamou
- Radar Systems and Remote Sensing Lab of School of Electrical & Computer Engineering of National Technical University of Athens, Athens 15773 , Greece
| | - A Stylianou
- School of Sciences, European University Cyprus, Nicosia 2404, Cyprus.
| |
Collapse
|
3
|
Kondrakhova D, Unger M, Stadler H, Zakuťanská K, Tomašovičová N, Tomečková V, Horák J, Kimákova T, Komanický V. Determination diabetes mellitus disease markers in tear fluid by photothermal AFM-IR analysis. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2025; 64:102803. [PMID: 39788273 DOI: 10.1016/j.nano.2025.102803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 12/10/2024] [Accepted: 12/29/2024] [Indexed: 01/12/2025]
Abstract
The tear fluids from three healthy individuals and three patients with diabetes mellitus were examined using atomic force microscopy-infrared spectroscopy (AFM-IR) and Fourier transform infrared spectroscopy (FTIR). The dried tear samples showed different surface morphologies: the control sample had a dense network of heart-shaped dendrites, while the diabetic sample had fern-shaped dendrites. By using the AFM-IR technique we identified spatial distribution of constituents, indicating how diabetes affects the structural characteristics of dried tears. FTIR showed that the dendritic structures gradually disappeared over time due to glucose-induced lysozyme damage. The tear fluid from diabetes mellitus patients has a higher concentration of glucose, which accelerates the breakdown of lysozyme and, as a result, the quick loss of the dendritic structure. Our study shows that analysis of dry tear fluid can be promising technique for the detection of glycated proteins that reveal long lasting hyperglycemia and diabetes mellitus.
Collapse
Affiliation(s)
- Daria Kondrakhova
- Institute of Physics, Department of Condensed Matter Physics, Faculty of Science, Pavol Jozef Šafárik University in Košice, Park Angelinum 9, Košice 041 54, Slovakia
| | - Miriam Unger
- Bruker Nano Surfaces & Metrology, Östliche Rheinbrückenstrasse 49, 76187 Karlsruhe, Germany
| | - Hartmut Stadler
- Bruker Nano Surfaces & Metrology, Östliche Rheinbrückenstrasse 49, 76187 Karlsruhe, Germany
| | - Katarína Zakuťanská
- Department of Magnetism, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, Košice 040 01, Slovakia
| | - Natália Tomašovičová
- Department of Magnetism, Institute of Experimental Physics, Slovak Academy of Sciences, Watsonova 47, Košice 040 01, Slovakia
| | - Vladimíra Tomečková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Jakub Horák
- Měřicí technika Morava s.r.o., Babická 619, 664 84 Zastávka, Czech Republic
| | - Tatiana Kimákova
- Department of Public Health and Hygiene, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Šrobárová 2, Košice 041 80, Slovakia
| | - Vladimír Komanický
- Institute of Physics, Department of Condensed Matter Physics, Faculty of Science, Pavol Jozef Šafárik University in Košice, Park Angelinum 9, Košice 041 54, Slovakia.
| |
Collapse
|
4
|
Chang Z, Zhou Y, Dong L, Qiao LR, Yang H, Xu GK. Deciphering the complex mechanics of atherosclerotic plaques: A hybrid hierarchical theory-microrheology approach. Acta Biomater 2024; 189:399-412. [PMID: 39307259 DOI: 10.1016/j.actbio.2024.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/07/2024]
Abstract
Understanding the viscoelastic properties of atherosclerotic plaques at rupture-prone scales is crucial for assessing their vulnerability. Here, we develop a Hybrid Hierarchical theory-Microrheology (HHM) approach, enabling the analysis of multiscale mechanical variations and distribution changes in regional tissue viscoelasticity within plaques across different spatial scales. We disclose a universal two-stage power-law rheology in plaques, characterized by distinct power-law exponents (αshort and αlong), which serve as mechanical indexes for plaque components and assessing mechanical gradients. We further propose a self-similar hierarchical theory that effectively delineates plaque heterogeneity from the cytoplasm, cell, to tissue levels. Moreover, our proposed multi-layer perceptron model addresses the viscoelastic heterogeneity and gradients within plaques, offering a promising diagnostic strategy for identifying unstable plaques. These findings not only advance our understanding of plaque mechanics but also pave the way for innovative diagnostic approaches in cardiovascular disease management. STATEMENT OF SIGNIFICANCE: Our study pioneers a Hybrid Hierarchical theory-Microrheology (HHM) approach to dissect the intricate viscoelasticity of atherosclerotic plaques, focusing on distinct components including cap fibrosis, lipid pools, and intimal fibrosis. We unveil a universal two-stage power-law rheology capturing mechanical variations across plaque structures. The proposed hierarchical model adeptly captures viscoelasticity changes from cytoplasm, cell to tissue levels. Based on the newly proposed markers, we further develop a machine learning (ML) diagnostic model that sets precise criteria for evaluating plaque components and heterogeneity. This work not only reveals the comprehensive mechanical heterogeneity within plaques but also introduces a mechanical marker-based ML strategy for assessing plaque conditions, offering a significant leap towards understanding and diagnosing atherosclerotic risks.
Collapse
Affiliation(s)
- Zhuo Chang
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yidan Zhou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, China
| | - Le Dong
- School of Artificial Intelligence, Xidian University, Xi'an 710071, China
| | - Lin-Ru Qiao
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710000, China.
| | - Guang-Kui Xu
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, School of Aerospace Engineering, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
5
|
Kontomaris SV, Malamou A, Stylianou A. Accurate Modelling of AFM Force-Indentation Curves with Blunted Indenters at Small Indentation Depths. MICROMACHINES 2024; 15:1209. [PMID: 39459083 PMCID: PMC11509629 DOI: 10.3390/mi15101209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/24/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024]
Abstract
When testing biological samples with atomic force microscopy (AFM) nanoindentation using pyramidal indenters, Sneddon's equation is commonly used for data processing, approximating the indenter as a perfect cone. While more accurate models treat the AFM tip as a blunted cone or pyramid, these are complex and lack a direct relationship between applied force and indentation depth, complicating data analysis. This paper proposes a new equation derived from simple mathematical processes and physics-based criteria. It is accurate for small indentation depths and serves as a viable alternative to complex classical approaches. The proposed equation has been validated for ℎ < 3R (where h is the indentation depth and R is the tip radius) and confirmed through simulations with blunted conical and pyramidal indenters, as well as experiments on prostate cancer cells. It is a reliable method for experiments where the tip radius cannot be ignored, such as in shallow indentations on thin samples to avoid substrate effects.
Collapse
Affiliation(s)
- Stylianos Vasileios Kontomaris
- Cancer Mechanobiology and Applied Biophysics Group, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus;
| | - Anna Malamou
- School of Electrical and Computer Engineering, National Technical University of Athens, 15773 Athens, Greece;
| | - Andreas Stylianou
- Cancer Mechanobiology and Applied Biophysics Group, School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus;
| |
Collapse
|
6
|
Krawczyk-Wołoszyn K, Roczkowski D, Reich A, Żychowska M. Applying the Atomic Force Microscopy Technique in Medical Sciences-A Narrative Review. Biomedicines 2024; 12:2012. [PMID: 39335524 PMCID: PMC11429229 DOI: 10.3390/biomedicines12092012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/25/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Penetrating deep into the cells of the human body in real time has become increasingly possible with the implementation of modern technologies in medicine. Atomic force microscopy (AFM) enables the effective live imaging of cellular and molecular structures of biological samples (such as cells surfaces, components of biological membranes, cell nuclei, actin networks, proteins, and DNA) and provides three-dimensional surface visualization (in X-, Y-, and Z-planes). Furthermore, the AFM technique enables the study of the mechanical, electrical, and magnetic properties of cells and cell organelles and the measurements of interaction forces between biomolecules. The technique has found wide application in cancer research. With the use of AFM, it is not only possible to differentiate between healthy and cancerous cells, but also to distinguish between the stages of cancerous conditions. For many years, AFM has been an important tool for the study of neurodegenerative diseases associated with the deposition of peptide amyloid plaques. In recent years, a significant amount of research has been conducted on the application of AFM in the evaluation of connective tissue cell mechanics. This review aims to provide the spectrum of the most important applications of the AFM technique in medicine to date.
Collapse
Affiliation(s)
- Karolina Krawczyk-Wołoszyn
- Doctoral School, University of Rzeszow, 35-959 Rzeszów, Poland;
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszów, Poland;
| | - Damian Roczkowski
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszów, Poland;
| | - Adam Reich
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszów, Poland;
| | - Magdalena Żychowska
- Department of Dermatology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-959 Rzeszów, Poland;
| |
Collapse
|
7
|
Thomas-Chemin O, Séverac C, Moumen A, Martinez-Rivas A, Vieu C, Le Lann MV, Trevisiol E, Dague E. Automated Bio-AFM Generation of Large Mechanome Data Set and Their Analysis by Machine Learning to Classify Cancerous Cell Lines. ACS APPLIED MATERIALS & INTERFACES 2024; 16:44504-44517. [PMID: 39162348 DOI: 10.1021/acsami.4c09218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Mechanobiological measurements have the potential to discriminate healthy cells from pathological cells. However, a technology frequently used to measure these properties, i.e., atomic force microscopy (AFM), suffers from its low output and lack of standardization. In this work, we have optimized AFM mechanical measurement on cell populations and developed a technology combining cell patterning and AFM automation that has the potential to record data on hundreds of cells (956 cells measured for publication). On each cell, 16 force curves (FCs) and seven features/FC, constituting the mechanome, were calculated. All of the FCs were then classified using machine learning tools with a statistical approach based on a fuzzy logic algorithm, trained to discriminate between nonmalignant and cancerous cells (training base, up to 120 cells/cell line). The proof of concept was first made on prostate nonmalignant (RWPE-1) and cancerous cell lines (PC3-GFP), then on nonmalignant (Hs 895.Sk) and cancerous (Hs 895.T) skin fibroblast cell lines, and demonstrated the ability of our method to classify correctly 73% of the cells (194 cells in the database/cell line) despite the very high degree of similarity of the whole set of measurements (79-100% similarity).
Collapse
Affiliation(s)
| | - Childérick Séverac
- LAAS-CNRS, Université de Toulouse, CNRS, 31031 Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM, CNRS, EFS, ENVT, Université P. Sabatier, 31100 Toulouse, France
| | | | | | - Christophe Vieu
- LAAS-CNRS, Université de Toulouse, CNRS, 31031 Toulouse, France
| | | | - Emmanuelle Trevisiol
- LAAS-CNRS, Université de Toulouse, CNRS, 31031 Toulouse, France
- TBI, Université de Toulouse, CNRS, INRAE, INSA, 31400 Toulouse, France
| | - Etienne Dague
- LAAS-CNRS, Université de Toulouse, CNRS, 31031 Toulouse, France
| |
Collapse
|
8
|
Kan T, Li H, Hou L, Cui J, Wang Y, Sun L, Wang L, Yan M, Yu Z. Matrix stiffness aggravates osteoarthritis progression through H3K27me3 demethylation induced by mitochondrial damage. iScience 2024; 27:110507. [PMID: 39156637 PMCID: PMC11328034 DOI: 10.1016/j.isci.2024.110507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/20/2024] [Accepted: 07/11/2024] [Indexed: 08/20/2024] Open
Abstract
Abnormal epigenetics is the initial factor of the occurrence and development of osteoarthritis (OA), and abnormal mechanical load is a key pathogenic factor of OA. However, how abnormal mechanical load affects chondrocyte epigenetics is unclear. Chondrocytes reportedly respond to mechanics through the extracellular matrix (ECM), which has a role in regulating epigenetics in various diseases, and mitochondria are potential mediators of communication between mechanics and epigenetics. Therefore, it is hypothesized that the matrix mechanics of cartilage regulates their epigenetics through mitochondria and leads to OA. The matrix stiffness of OA cartilage on the stress-concentrated side increases, mitochondrial damage of chondrocyte is severe, and the chondrocyte H3K27me3 is demethylated. Moreover, mitochondrial permeability transition pore (mPTP) opens to increase the translocation of plant homeodomain finger protein 8 (Phf8) into the nucleus to catalyze H3K27me3 demethylation. This provides a new perspective for us to understand the mechanism of OA based on mechanobiology.
Collapse
Affiliation(s)
- Tianyou Kan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - Hanjun Li
- Renji-Med X Clinical Stem Cell Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lingli Hou
- Shanghai Institute of Precision Medicine, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Junqi Cui
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yao Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Lin Sun
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Liao Wang
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mengning Yan
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhifeng Yu
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
9
|
Dönges L, Damle A, Mainardi A, Bock T, Schönenberger M, Martin I, Barbero A. Engineered human osteoarthritic cartilage organoids. Biomaterials 2024; 308:122549. [PMID: 38554643 DOI: 10.1016/j.biomaterials.2024.122549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
The availability of human cell-based models capturing molecular processes of cartilage degeneration can facilitate development of disease-modifying therapies for osteoarthritis [1], a currently unmet clinical need. Here, by imposing specific inflammatory challenges upon mesenchymal stromal cells at a defined stage of chondrogenesis, we engineered a human organotypic model which recapitulates main OA pathological traits such as chondrocyte hypertrophy, cartilage matrix mineralization, enhanced catabolism and mechanical stiffening. To exemplify the utility of the model, we exposed the engineered OA cartilage organoids to factors known to attenuate pathological features, including IL-1Ra, and carried out mass spectrometry-based proteomics. We identified that IL-1Ra strongly reduced production of the transcription factor CCAAT/enhancer-binding protein beta [2] and demonstrated that inhibition of the C/EBPβ-activating kinases could revert the degradative processes. Human OA cartilage organoids thus represent a relevant tool towards the discovery of new molecular drivers of cartilage degeneration and the assessment of therapeutics targeting associated pathways.
Collapse
Affiliation(s)
- Laura Dönges
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Atharva Damle
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Andrea Mainardi
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Thomas Bock
- Proteomics Core Facility, Biozentrum University of Basel, 4056, Basel, Switzerland
| | - Monica Schönenberger
- Nano Imaging Lab, Swiss Nanoscience Institute, University of Basel, 4056, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland.
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| |
Collapse
|
10
|
Lima I, Silva A, Sousa F, Ferreira W, Freire R, de Oliveira C, de Sousa J. Measuring the viscoelastic relaxation function of cells with a time-dependent interpretation of the Hertz-Sneddon indentation model. Heliyon 2024; 10:e30623. [PMID: 38770291 PMCID: PMC11103437 DOI: 10.1016/j.heliyon.2024.e30623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 05/22/2024] Open
Abstract
The Hertz-Sneddon elastic indentation model is widely adopted in the biomechanical investigation of living cells and other soft materials using atomic force microscopy despite the explicit viscoelastic nature of these materials. In this work, we demonstrate that an exact analytical viscoelastic force model for power-law materials, can be interpreted as a time-dependent Hertz-Sneddon-like model. Characterizing fibroblasts (L929) and osteoblasts (OFCOLII) demonstrates the model's accuracy. Our results show that the difference between Young's modulus E Y obtained by fitting force curves with the Hertz-Sneddon model and the effective Young's modulus derived from the viscoelastic force model is less than 3%, even when cells are probed at large forces where nonlinear deformation effects become significant. We also propose a measurement protocol that involves probing samples at different indentation speeds and forces, enabling the construction of the average viscoelastic relaxation function of samples by conveniently fitting the force curves with the Hertz-Sneddon model.
Collapse
Affiliation(s)
- I.V.M. Lima
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - A.V.S. Silva
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
- Instituto Federal do Rio Grande do Norte, Pau dos Ferros, 59900-000, Rio Grande do Norte, Brazil
| | - F.D. Sousa
- Núcleo de Biologia Experimental, Universidade de Fortaleza, Fortaleza, 60811-905, Ceará, Brazil
| | - W.P. Ferreira
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - R.S. Freire
- Central Analítica, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - C.L.N. de Oliveira
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - J.S. de Sousa
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| |
Collapse
|
11
|
Fu B, Shen J, Zou X, Sun N, Zhang Z, Liu Z, Zeng C, Liu H, Huang W. Matrix stiffening promotes chondrocyte senescence and the osteoarthritis development through downregulating HDAC3. Bone Res 2024; 12:32. [PMID: 38789434 PMCID: PMC11126418 DOI: 10.1038/s41413-024-00333-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/13/2024] [Accepted: 04/01/2024] [Indexed: 05/26/2024] Open
Abstract
Extracellular matrix (ECM) stiffening is a typical characteristic of cartilage aging, which is a quintessential feature of knee osteoarthritis (KOA). However, little is known about how ECM stiffening affects chondrocytes and other molecules downstream. This study mimicked the physiological and pathological stiffness of human cartilage using polydimethylsiloxane (PDMS) substrates. It demonstrated that epigenetic Parkin regulation by histone deacetylase 3 (HDAC3) represents a new mechanosensitive mechanism by which the stiffness matrix affected chondrocyte physiology. We found that ECM stiffening accelerated cultured chondrocyte senescence in vitro, while the stiffness ECM downregulated HDAC3, prompting Parkin acetylation to activate excessive mitophagy and accelerating chondrocyte senescence and osteoarthritis (OA) in mice. Contrarily, intra-articular injection with an HDAC3-expressing adeno-associated virus restored the young phenotype of the aged chondrocytes stimulated by ECM stiffening and alleviated OA in mice. The findings indicated that changes in the mechanical ECM properties initiated pathogenic mechanotransduction signals, promoted the Parkin acetylation and hyperactivated mitophagy, and damaged chondrocyte health. These results may provide new insights into chondrocyte regulation by the mechanical properties of ECM, suggesting that the modification of the physical ECM properties may be a potential OA treatment strategy.
Collapse
Affiliation(s)
- Bowen Fu
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510145, Guangdong, China
- Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Jianlin Shen
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China
- Central Laboratory, Affiliated Hospital of Putian University, Putian, 351100, Fujian, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Nian Sun
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510145, Guangdong, China
- Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Ze Zhang
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangzhou Blood Center, Guangzhou, 510095, Guangdong, China
| | - Zengping Liu
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Guangzhou Blood Center, Guangzhou, 510095, Guangdong, China
| | - Canjun Zeng
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510145, Guangdong, China
- Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China
| | - Huan Liu
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Wenhua Huang
- Guangdong Medical Innovation Platform for Translation of 3D Printing Application, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China.
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510145, Guangdong, China.
- Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
12
|
Hartmann B, Fleischhauer L, Nicolau M, Jensen THL, Taran FA, Clausen-Schaumann H, Reuten R. Profiling native pulmonary basement membrane stiffness using atomic force microscopy. Nat Protoc 2024; 19:1498-1528. [PMID: 38429517 DOI: 10.1038/s41596-024-00955-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 11/27/2023] [Indexed: 03/03/2024]
Abstract
Mammalian cells sense and react to the mechanics of their immediate microenvironment. Therefore, the characterization of the biomechanical properties of tissues with high spatial resolution provides valuable insights into a broad variety of developmental, homeostatic and pathological processes within living organisms. The biomechanical properties of the basement membrane (BM), an extracellular matrix (ECM) substructure measuring only ∼100-400 nm across, are, among other things, pivotal to tumor progression and metastasis formation. Although the precise assignment of the Young's modulus E of such a thin ECM substructure especially in between two cell layers is still challenging, biomechanical data of the BM can provide information of eminent diagnostic potential. Here we present a detailed protocol to quantify the elastic modulus of the BM in murine and human lung tissue, which is one of the major organs prone to metastasis. This protocol describes a streamlined workflow to determine the Young's modulus E of the BM between the endothelial and epithelial cell layers shaping the alveolar wall in lung tissues using atomic force microscopy (AFM). Our step-by-step protocol provides instructions for murine and human lung tissue extraction, inflation of these tissues with cryogenic cutting medium, freezing and cryosectioning of the tissue samples, and AFM force-map recording. In addition, it guides the reader through a semi-automatic data analysis procedure to identify the pulmonary BM and extract its Young's modulus E using an in-house tailored user-friendly AFM data analysis software, the Center for Applied Tissue Engineering and Regenerative Medicine processing toolbox, which enables automatic loading of the recorded force maps, conversion of the force versus piezo-extension curves to force versus indentation curves, calculation of Young's moduli and generation of Young's modulus maps, where the pulmonary BM can be identified using a semi-automatic spatial filtering tool. The entire protocol takes 1-2 d.
Collapse
Affiliation(s)
- Bastian Hartmann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Lutz Fleischhauer
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany
- Center for Nanoscience, Munich, Germany
| | - Monica Nicolau
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Thomas Hartvig Lindkær Jensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Pathology, Rigshospitalet, Copenhagen, Denmark
| | - Florin-Andrei Taran
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Hauke Clausen-Schaumann
- Munich University of Applied Sciences, Center for Applied Tissue Engineering and Regenerative Medicine - CANTER, Munich, Germany.
- Center for Nanoscience, Munich, Germany.
| | - Raphael Reuten
- Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany.
- Department of Obstetrics and Gynecology, Medical Center, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
13
|
Wong CA, Fraticelli Guzmán NS, Read AT, Hedberg-Buenz A, Anderson MG, Feola AJ, Sulchek T, Ethier CR. A method for analyzing AFM force mapping data obtained from soft tissue cryosections. J Biomech 2024; 168:112113. [PMID: 38648717 PMCID: PMC11128031 DOI: 10.1016/j.jbiomech.2024.112113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/23/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
Atomic force microscopy (AFM) is a valuable tool for assessing mechanical properties of biological samples, but interpretations of measurements on whole tissues can be difficult due to the tissue's highly heterogeneous nature. To overcome such difficulties and obtain more robust estimates of tissue mechanical properties, we describe an AFM force mapping and data analysis pipeline to characterize the mechanical properties of cryosectioned soft tissues. We assessed this approach on mouse optic nerve head and rat trabecular meshwork, cornea, and sclera. Our data show that the use of repeated measurements, outlier exclusion, and log-normal data transformation increases confidence in AFM mechanical measurements, and we propose that this methodology can be broadly applied to measuring soft tissue properties from cryosections.
Collapse
Affiliation(s)
- Cydney A Wong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | | - A Thomas Read
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Adam Hedberg-Buenz
- Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA
| | - Michael G Anderson
- Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA
| | - Andrew J Feola
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Department of Ophthalmology, Emory University, Atlanta, GA; Center for Visual & Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
| | - Todd Sulchek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - C Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Department of Ophthalmology, Emory University, Atlanta, GA.
| |
Collapse
|
14
|
Magerle R, Zech P, Dehnert M, Bendixen A, Otto A. Rate-independent hysteretic energy dissipation in collagen fibrils. SOFT MATTER 2024; 20:2831-2839. [PMID: 38456340 DOI: 10.1039/d3sm01625k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Nanoindentation cycles measured with an atomic force microscope on hydrated collagen fibrils exhibit a rate-independent hysteresis with return point memory. This previously unknown energy dissipation mechanism describes in unified form elastoplastic indentation, capillary adhesion, and surface leveling at indentation velocities smaller than 1 μm s-1, where viscous friction is negligible. A generic hysteresis model, based on force-distance data measured during one large approach-retract cycle, predicts the force (output) and the dissipated energy for arbitrary indentation trajectories (input). While both quantities are rate independent, they do depend nonlinearly on indentation history and on indentation amplitude.
Collapse
Affiliation(s)
- Robert Magerle
- Fakultät für Naturwissenschaften, Technische Universität Chemnitz, 09107 Chemnitz, Germany.
| | - Paul Zech
- Fakultät für Naturwissenschaften, Technische Universität Chemnitz, 09107 Chemnitz, Germany.
| | - Martin Dehnert
- Fakultät für Naturwissenschaften, Technische Universität Chemnitz, 09107 Chemnitz, Germany.
| | - Alexandra Bendixen
- Fakultät für Naturwissenschaften, Technische Universität Chemnitz, 09107 Chemnitz, Germany.
| | - Andreas Otto
- Fakultät für Naturwissenschaften, Technische Universität Chemnitz, 09107 Chemnitz, Germany.
| |
Collapse
|
15
|
Govindaraj K, Meteling M, van Rooij J, Becker M, van Wijnen AJ, van den Beucken JJJP, Ramos YFM, van Meurs J, Post JN, Leijten J. Osmolarity-Induced Altered Intracellular Molecular Crowding Drives Osteoarthritis Pathology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306722. [PMID: 38213111 PMCID: PMC10953583 DOI: 10.1002/advs.202306722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/08/2023] [Indexed: 01/13/2024]
Abstract
Osteoarthritis (OA) is a multifactorial degenerative joint disease of which the underlying mechanisms are yet to be fully understood. At the molecular level, multiple factors including altered signaling pathways, epigenetics, metabolic imbalance, extracellular matrix degradation, production of matrix metalloproteinases, and inflammatory cytokines, are known to play a detrimental role in OA. However, these factors do not initiate OA, but are mediators or consequences of the disease, while many other factors causing the etiology of OA are still unknown. Here, it is revealed that microenvironmental osmolarity can induce and reverse osteoarthritis-related behavior of chondrocytes via altered intracellular molecular crowding, which represents a previously unknown mechanism underlying OA pathophysiology. Decreased intracellular crowding is associated with increased sensitivity to proinflammatory triggers and decreased responsiveness to anabolic stimuli. OA-induced lowered intracellular molecular crowding could be renormalized via exposure to higher extracellular osmolarity such as those found in healthy joints, which reverse OA chondrocyte's sensitivity to catabolic stimuli as well as its glycolytic metabolism.
Collapse
Affiliation(s)
- Kannan Govindaraj
- Department of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Marieke Meteling
- Department of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Jeroen van Rooij
- Department of Internal MedicineErasmus MCDr. Molewaterplein 40Rotterdam3015GDThe Netherlands
| | - Malin Becker
- Department of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | | | | | - Yolande F. M. Ramos
- Department of Biomedical Data SciencesSection Molecular EpidemiologyLUMCEinthovenweg 20Leiden2333 ZCThe Netherlands
| | - Joyce van Meurs
- Department of Internal MedicineErasmus MCDr. Molewaterplein 40Rotterdam3015GDThe Netherlands
- Department of Orthopedics & Sports MedicineErasmus MCDr. Molewaterplein 40Rotterdam3015GDThe Netherlands
| | - Janine N. Post
- Department of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| | - Jeroen Leijten
- Department of Developmental BioengineeringFaculty of Science and Technology, Technical Medical CentreUniversity of TwenteDrienerlolaan 5Enschede7522NBThe Netherlands
| |
Collapse
|
16
|
Stolz M. The Revolution in Breast Cancer Diagnostics: From Visual Inspection of Histopathology Slides to Using Desktop Tissue Analysers for Automated Nanomechanical Profiling of Tumours. Bioengineering (Basel) 2024; 11:237. [PMID: 38534510 DOI: 10.3390/bioengineering11030237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
We aim to develop new portable desktop tissue analysers (DTAs) to provide fast, low-cost, and precise test results for fast nanomechanical profiling of tumours. This paper will explain the reasoning for choosing indentation-type atomic force microscopy (IT-AFM) to reveal the functional details of cancer. Determining the subtype, cancer stage, and prognosis will be possible, which aids in choosing the best treatment. DTAs are based on fast IT-AFM at the size of a small box that can be made for a low budget compared to other clinical imaging tools. The DTAs can work in remote areas and all parts of the world. There are a number of direct benefits: First, it is no longer needed to wait a week for the pathology report as the test will only take 10 min. Second, it avoids the complicated steps of making histopathology slides and saves costs of labour. Third, computers and robots are more consistent, more reliable, and more economical than human workers which may result in fewer diagnostic errors. Fourth, the IT-AFM analysis is capable of distinguishing between various cancer subtypes. Fifth, the IT-AFM analysis could reveal new insights about why immunotherapy fails. Sixth, IT-AFM may provide new insights into the neoadjuvant treatment response. Seventh, the healthcare system saves money by reducing diagnostic backlogs. Eighth, the results are stored on a central server and can be accessed to develop strategies to prevent cancer. To bring the IT-AFM technology from the bench to the operation theatre, a fast IT-AFM sensor needs to be developed and integrated into the DTAs.
Collapse
Affiliation(s)
- Martin Stolz
- National Centre for Advanced Tribology at Southampton, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
17
|
Fan X, Sun AR, Young RSE, Afara IO, Hamilton BR, Ong LJY, Crawford R, Prasadam I. Spatial analysis of the osteoarthritis microenvironment: techniques, insights, and applications. Bone Res 2024; 12:7. [PMID: 38311627 PMCID: PMC10838951 DOI: 10.1038/s41413-023-00304-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 02/06/2024] Open
Abstract
Osteoarthritis (OA) is a debilitating degenerative disease affecting multiple joint tissues, including cartilage, bone, synovium, and adipose tissues. OA presents diverse clinical phenotypes and distinct molecular endotypes, including inflammatory, metabolic, mechanical, genetic, and synovial variants. Consequently, innovative technologies are needed to support the development of effective diagnostic and precision therapeutic approaches. Traditional analysis of bulk OA tissue extracts has limitations due to technical constraints, causing challenges in the differentiation between various physiological and pathological phenotypes in joint tissues. This issue has led to standardization difficulties and hindered the success of clinical trials. Gaining insights into the spatial variations of the cellular and molecular structures in OA tissues, encompassing DNA, RNA, metabolites, and proteins, as well as their chemical properties, elemental composition, and mechanical attributes, can contribute to a more comprehensive understanding of the disease subtypes. Spatially resolved biology enables biologists to investigate cells within the context of their tissue microenvironment, providing a more holistic view of cellular function. Recent advances in innovative spatial biology techniques now allow intact tissue sections to be examined using various -omics lenses, such as genomics, transcriptomics, proteomics, and metabolomics, with spatial data. This fusion of approaches provides researchers with critical insights into the molecular composition and functions of the cells and tissues at precise spatial coordinates. Furthermore, advanced imaging techniques, including high-resolution microscopy, hyperspectral imaging, and mass spectrometry imaging, enable the visualization and analysis of the spatial distribution of biomolecules, cells, and tissues. Linking these molecular imaging outputs to conventional tissue histology can facilitate a more comprehensive characterization of disease phenotypes. This review summarizes the recent advancements in the molecular imaging modalities and methodologies for in-depth spatial analysis. It explores their applications, challenges, and potential opportunities in the field of OA. Additionally, this review provides a perspective on the potential research directions for these contemporary approaches that can meet the requirements of clinical diagnoses and the establishment of therapeutic targets for OA.
Collapse
Affiliation(s)
- Xiwei Fan
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Antonia Rujia Sun
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Reuben S E Young
- Central Analytical Research Facility, Queensland University of Technology, Brisbane, QLD, Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW, Australia
| | - Isaac O Afara
- Department of Technical Physics, University of Eastern Finland, Kuopio, Finland
- School of Electrical Engineering and Computer Science, Faculty of Engineering, Architecture and Information Technology, University of Queensland, Brisbane, QLD, Australia
| | - Brett R Hamilton
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD, Australia
| | - Louis Jun Ye Ong
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ross Crawford
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
- The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Indira Prasadam
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia.
- School of Mechanical, Medical & Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
18
|
Iijima H, Zhang F, Ambrosio F, Matsui Y. Network-based cytokine inference implicates Oncostatin M as a driver of an inflammation phenotype in knee osteoarthritis. Aging Cell 2024; 23:e14043. [PMID: 38111237 PMCID: PMC10861212 DOI: 10.1111/acel.14043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/28/2023] [Accepted: 11/01/2023] [Indexed: 12/20/2023] Open
Abstract
Inflammatory cytokines released by synovium after trauma disturb the gene regulatory network and have been implicated in the pathophysiology of osteoarthritis. A mechanistic understanding of how aging perturbs this process can help identify novel interventions. Here, we introduced network paradigms to simulate cytokine-mediated pathological communication between the synovium and cartilage. Cartilage-specific network analysis of injured young and aged murine knees revealed aberrant matrix remodeling as a transcriptomic response unique to aged knees displaying accelerated cartilage degradation. Next, network-based cytokine inference with pharmacological manipulation uncovered IL6 family member, Oncostatin M (OSM), as a driver of the aberrant matrix remodeling. By implementing a phenotypic drug discovery approach, we identified that the activation of OSM recapitulated an "inflammatory" phenotype of knee osteoarthritis and highlighted high-value targets for drug development and repurposing. These findings offer translational opportunities targeting the inflammation-driven osteoarthritis phenotype.
Collapse
Affiliation(s)
- Hirotaka Iijima
- Discovery Center for Musculoskeletal RecoverySchoen Adams Research Institute at SpauldingCharlestownMassachusettsUSA
- Department of Physical Medicine & RehabilitationHarvard Medical SchoolBostonMassachusettsUSA
- Department of Physical Medicine & RehabilitationSpaulding Rehabilitation HospitalCharlestownMassachusettsUSA
- Institute for Advanced ResearchNagoya UniversityNagoyaJapan
- Biomedical and Health Informatics Unit, Graduate School of MedicineNagoya UniversityNagoyaJapan
| | - Fan Zhang
- Department of Medicine Division of RheumatologyUniversity of Colorado School of MedicineAuroraColoradoUSA
- Department of Biomedical Informatics Center for Health AIUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Fabrisia Ambrosio
- Discovery Center for Musculoskeletal RecoverySchoen Adams Research Institute at SpauldingCharlestownMassachusettsUSA
- Department of Physical Medicine & RehabilitationHarvard Medical SchoolBostonMassachusettsUSA
- Department of Physical Medicine & RehabilitationSpaulding Rehabilitation HospitalCharlestownMassachusettsUSA
| | - Yusuke Matsui
- Biomedical and Health Informatics Unit, Graduate School of MedicineNagoya UniversityNagoyaJapan
- Institute for Glyco‐core Research, Tokai National Higher Education and Research SystemNagoya UniversityNagoyaJapan
| |
Collapse
|
19
|
Song J, Zeng X, Li C, Yin H, Mao S, Ren D. Alteration in cartilage matrix stiffness as an indicator and modulator of osteoarthritis. Biosci Rep 2024; 44:BSR20231730. [PMID: 38014522 PMCID: PMC10794814 DOI: 10.1042/bsr20231730] [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: 10/11/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023] Open
Abstract
Osteoarthritis (OA) is characterized by cartilage degeneration and destruction, leading to joint ankylosis and disability. The major challenge in diagnosing OA at early stage is not only lack of clinical symptoms but also the insufficient histological and immunohistochemical signs. Alteration in cartilage stiffness during OA progression, especially at OA initiation, has been confirmed by growing evidences. Moreover, the stiffness of cartilage extracellular matrix (ECM), pericellular matrix (PCM) and chondrocytes during OA development are dynamically changed in unique and distinct fashions, revealing possibly inconsistent conclusions when detecting cartilage matrix stiffness at different locations and scales. In addition, it will be discussed regarding the mechanisms through which OA-related cartilage degenerations exhibit stiffened or softened matrix, highlighting some critical events that generally incurred to cartilage stiffness alteration, as well as some typical molecules that participated in constituting the mechanical properties of cartilage. Finally, in vitro culturing chondrocytes in various stiffness-tunable scaffolds provided a reliable method to explore the matrix stiffness-dependent modulation of chondrocyte metabolism, which offers valuable information on optimizing implant scaffolds to maximally promote cartilage repair and regeneration during OA. Overall, this review systematically and comprehensively elucidated the current progresses in the relationship between cartilage stiffness alteration and OA progression. We hope that deeper attention and understanding in this researching field will not only develop more innovative methods in OA early detection and diagnose but also provide promising ideas in OA therapy and prognosis.
Collapse
Affiliation(s)
- Jing Song
- Qingdao University Affiliated Qingdao Women and Children’s Hospital, Department of Stomatology Medical Center, Qingdao University, Qingdao, Shandong, CN, China
| | - Xuemin Zeng
- The Affiliated Hospital of Qingdao University, Department of Stomatology Medical Center, Qingdao University, Qingdao, Shandong, CN, China
| | - Chenzhi Li
- The Affiliated Hospital of Qingdao University, Department of Stomatology Medical Center, Qingdao University, Qingdao, Shandong, CN, China
| | - Hongyan Yin
- Institute of Hybrid Materials, College of Materials Science and Engineering, Qingdao University, Qingdao, Shandong, CN, China
| | - Sui Mao
- Institute of Hybrid Materials, College of Materials Science and Engineering, Qingdao University, Qingdao, Shandong, CN, China
| | - Dapeng Ren
- The Affiliated Hospital of Qingdao University, Department of Stomatology Medical Center, Qingdao University, Qingdao, Shandong, CN, China
| |
Collapse
|
20
|
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
|
21
|
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: 6] [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
|
22
|
Zhou C, Yang Y, Duan M, Chen C, Pi C, Zhang D, Liu X, Xie J. Biomimetic Fibers Based on Equidistant Micropillar Arrays Determines Chondrocyte Fate via Mechanoadaptability. Adv Healthc Mater 2023; 12:e2301685. [PMID: 37596884 DOI: 10.1002/adhm.202301685] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/02/2023] [Indexed: 08/20/2023]
Abstract
It is recognized that the changes in the physical properties of extracellular matrix (ECM) result in fine-tuned cell responses including cell morphology, proliferation and differentiation. In this study, a novel patterned equidistant micropillar substrate based on polydimethylsiloxane (PDMS) is designed to mimic the collagen fiber-like network of the cartilage matrix. By changing the component of the curing agent to an oligomeric base, micropillar substrates with the same topology but different stiffnesses are obtained and it is found that chondrocytes seeded onto the soft micropillar substrate maintain their phenotype by gathering type II collagen and aggrecan more effectively than those seeded onto the stiff micropillar substrate. Moreover, chondrocytes sense and respond to micropillar substrates with different stiffnesses by altering the ECM-cytoskeleton-focal adhesion axis. Further, it is found that the soft substrate-preserved chondrocyte phenotype is dependent on the activation of Wnt/β-catenin signaling. Finally, it is indicated that the changes in osteoid-like region formation and cartilage phenotype loss in the stiffened sclerotic area of osteoarthritis cartilage to validate the changes triggered by micropillar substrates with different stiffnesses. This study provides the cell behavior changes that are more similar to those of real chondrocytes at tissue level during the transition from a normal state to a state of osteoarthritis.
Collapse
Affiliation(s)
- Chenchen Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Yueyi Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Mengmeng Duan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Cheng Chen
- College of Medical Informatics, Chongqing Medical University, Chongqing, 400016, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610064, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610064, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
23
|
Wong CA, Fraticelli Guzmán NS, Read AT, Hedberg-Buenz A, Anderson MG, Feola AJ, Sulchek T, Ethier CR. A Method for Analyzing AFM Force Mapping Data Obtained from Soft Tissue Cryosections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566263. [PMID: 38014311 PMCID: PMC10680563 DOI: 10.1101/2023.11.08.566263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Atomic force microscopy (AFM) is a valuable tool for assessing mechanical properties of biological samples, but interpretations of measurements on whole tissues can be difficult due to the tissue's highly heterogeneous nature. To overcome such difficulties and obtain more robust estimates of tissue mechanical properties, we describe an AFM force mapping and data analysis pipeline to characterize the mechanical properties of cryosectioned soft tissues. We assessed this approach on mouse optic nerve head and rat trabecular meshwork, cornea, and sclera. Our data show that the use of repeated measurements, outlier exclusion, and log-normal data transformation increases confidence in AFM mechanical measurements, and we propose that this methodology can be broadly applied to measuring soft tissue properties from cryosections.
Collapse
Affiliation(s)
- Cydney A Wong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | | | - A Thomas Read
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Adam Hedberg-Buenz
- Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA
| | - Michael G Anderson
- Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA
| | - Andrew J Feola
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Ophthalmology, Emory University, Atlanta, GA
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Atlanta GA
| | - Todd Sulchek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - C Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Ophthalmology, Emory University, Atlanta, GA
| |
Collapse
|
24
|
Chang Z, Zhang L, Hang JT, Liu W, Xu GK. Viscoelastic Multiscale Mechanical Indexes for Assessing Liver Fibrosis and Treatment Outcomes. NANO LETTERS 2023; 23:9618-9625. [PMID: 37793647 PMCID: PMC10603793 DOI: 10.1021/acs.nanolett.3c03341] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/28/2023] [Indexed: 10/06/2023]
Abstract
Understanding liver tissue mechanics, particularly in the context of liver pathologies like fibrosis, cirrhosis, and carcinoma, holds pivotal significance for assessing disease severity and prognosis. Although the static mechanical properties of livers have been gradually studied, the intricacies of their dynamic mechanics remain enigmatic. Here, we characterize the dynamic creep responses of healthy, fibrotic, and mesenchymal stem cells (MSCs)-treated fibrotic lives. Strikingly, we unearth a ubiquitous two-stage power-law rheology of livers across different time scales with the exponents and their distribution profiles highly correlated to liver status. Moreover, our self-similar hierarchical theory effectively captures the delicate changes in the dynamical mechanics of livers. Notably, the viscoelastic multiscale mechanical indexes (i.e., power-law exponents and elastic stiffnesses of different hierarchies) and their distribution characteristics prominently vary with liver fibrosis and MSCs therapy. This study unveils the viscoelastic characteristics of livers and underscores the potential of proposed mechanical criteria for assessing disease evolution and prognosis.
Collapse
Affiliation(s)
- Zhuo Chang
- Laboratory
for Multiscale Mechanics and Medical Science, Department of Engineering
Mechanics, State Key Laboratory for Strength and Vibration of Mechanical
Structures, School of Aerospace Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Liqiang Zhang
- Institute
for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Jiu-Tao Hang
- Laboratory
for Multiscale Mechanics and Medical Science, Department of Engineering
Mechanics, State Key Laboratory for Strength and Vibration of Mechanical
Structures, School of Aerospace Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Wenjia Liu
- Institute
for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710004, China
| | - Guang-Kui Xu
- Laboratory
for Multiscale Mechanics and Medical Science, Department of Engineering
Mechanics, State Key Laboratory for Strength and Vibration of Mechanical
Structures, School of Aerospace Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
25
|
Kurz B, Lange T, Voelker M, Hart ML, Rolauffs B. Articular Cartilage-From Basic Science Structural Imaging to Non-Invasive Clinical Quantitative Molecular Functional Information for AI Classification and Prediction. Int J Mol Sci 2023; 24:14974. [PMID: 37834422 PMCID: PMC10573252 DOI: 10.3390/ijms241914974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
This review presents the changes that the imaging of articular cartilage has undergone throughout the last decades. It highlights that the expectation is no longer to image the structure and associated functions of articular cartilage but, instead, to devise methods for generating non-invasive, function-depicting images with quantitative information that is useful for detecting the early, pre-clinical stage of diseases such as primary or post-traumatic osteoarthritis (OA/PTOA). In this context, this review summarizes (a) the structure and function of articular cartilage as a molecular imaging target, (b) quantitative MRI for non-invasive assessment of articular cartilage composition, microstructure, and function with the current state of medical diagnostic imaging, (c), non-destructive imaging methods, (c) non-destructive quantitative articular cartilage live-imaging methods, (d) artificial intelligence (AI) classification of degeneration and prediction of OA progression, and (e) our contribution to this field, which is an AI-supported, non-destructive quantitative optical biopsy for early disease detection that operates on a digital tissue architectural fingerprint. Collectively, this review shows that articular cartilage imaging has undergone profound changes in the purpose and expectations for which cartilage imaging is used; the image is becoming an AI-usable biomarker with non-invasive quantitative functional information. This may aid in the development of translational diagnostic applications and preventive or early therapeutic interventions that are yet beyond our reach.
Collapse
Affiliation(s)
- Bodo Kurz
- Department of Anatomy, Christian-Albrechts-University, Otto-Hahn-Platz 8, 24118 Kiel, Germany
| | - Thomas Lange
- Medical Physics Department of Radiology, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany;
| | - Marita Voelker
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.V.); (M.L.H.)
| | - Melanie L. Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.V.); (M.L.H.)
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.V.); (M.L.H.)
| |
Collapse
|
26
|
Saini K, Cho S, Tewari M, Jalil AR, Wang M, Kasznel AJ, Yamamoto K, Chenoweth DM, Discher DE. Pan-tissue scaling of stiffness versus fibrillar collagen reflects contractility-driven strain that inhibits fibril degradation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559759. [PMID: 37808742 PMCID: PMC10557712 DOI: 10.1101/2023.09.27.559759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Polymer network properties such as stiffness often exhibit characteristic power laws in polymer density and other parameters. However, it remains unclear whether diverse animal tissues, composed of many distinct polymers, exhibit such scaling. Here, we examined many diverse tissues from adult mouse and embryonic chick to determine if stiffness ( E tissue ) follows a power law in relation to the most abundant animal protein, Collagen-I, even with molecular perturbations. We quantified fibrillar collagen in intact tissue by second harmonic generation (SHG) imaging and from tissue extracts by mass spectrometry (MS), and collagenase-mediated decreases were also tracked. Pan-tissue power laws for tissue stiffness versus Collagen-I levels measured by SHG or MS exhibit sub-linear scaling that aligns with results from cellularized gels of Collagen-I but not acellular gels. Inhibition of cellular myosin-II based contraction fits the scaling, and combination with inhibitors of matrix metalloproteinases (MMPs) show collagenase activity is strain - not stress- suppressed in tissues, consistent with past studies of gels and fibrils. Beating embryonic hearts and tendons, which differ in both collagen levels and stiffness by >1000-fold, similarly suppressed collagenases at physiological strains of ∼5%, with fiber-orientation regulating degradation. Scaling of E tissue based on 'use-it-or-lose-it' kinetics provides insight into scaling of organ size, microgravity effects, and regeneration processes while suggesting contractility-driven therapeutics.
Collapse
|
27
|
Matsuoka K, Bakiri L, Bilban M, Toegel S, Haschemi A, Yuan H, Kasper M, Windhager R, Wagner EF. Metabolic rewiring controlled by c-Fos governs cartilage integrity in osteoarthritis. Ann Rheum Dis 2023; 82:1227-1239. [PMID: 37344157 PMCID: PMC10423482 DOI: 10.1136/ard-2023-224002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023]
Abstract
OBJECTIVES The activator protein-1 (AP-1) transcription factor component c-Fos regulates chondrocyte proliferation and differentiation, but its involvement in osteoarthritis (OA) has not been functionally assessed. METHODS c-Fos expression was evaluated by immunohistochemistry on articular cartilage sections from patients with OA and mice subjected to the destabilisation of the medial meniscus (DMM) model of OA. Cartilage-specific c-Fos knockout (c-FosΔCh) mice were generated by crossing c-fosfl/fl to Col2a1-CreERT mice. Articular cartilage was evaluated by histology, immunohistochemistry, RNA sequencing (RNA-seq), quantitative reverse transcription PCR (qRT-PCR) and in situ metabolic enzyme assays. The effect of dichloroacetic acid (DCA), an inhibitor of pyruvate dehydrogenase kinase (Pdk), was assessed in c-FosΔCh mice subjected to DMM. RESULTS FOS-positive chondrocytes were increased in human and murine OA cartilage during disease progression. Compared with c-FosWT mice, c-FosΔCh mice exhibited exacerbated DMM-induced cartilage destruction. Chondrocytes lacking c-Fos proliferate less, have shorter collagen fibres and reduced cartilage matrix. Comparative RNA-seq revealed a prominent anaerobic glycolysis gene expression signature. Consistently decreased pyruvate dehydrogenase (Pdh) and elevated lactate dehydrogenase (Ldh) enzymatic activities were measured in situ, which are likely due to higher expression of hypoxia-inducible factor-1α, Ldha, and Pdk1 in chondrocytes. In vivo treatment of c-FosΔCh mice with DCA restored Pdh/Ldh activity, chondrocyte proliferation, collagen biosynthesis and decreased cartilage damage after DMM, thereby reverting the deleterious effects of c-Fos inactivation. CONCLUSIONS c-Fos modulates cellular bioenergetics in chondrocytes by balancing pyruvate flux between anaerobic glycolysis and the tricarboxylic acid cycle in response to OA signals. We identify a novel metabolic adaptation of chondrocytes controlled by c-Fos-containing AP-1 dimers that could be therapeutically relevant.
Collapse
Affiliation(s)
- Kazuhiko Matsuoka
- Genes and Disease group, Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Cellular and Molecular Tumor biology, Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Latifa Bakiri
- Genes and Disease group, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Stefan Toegel
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Hao Yuan
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Maria Kasper
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Reinhard Windhager
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Erwin F Wagner
- Genes and Disease group, Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Genes and Disease group, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Dudek M, Morris H, Rogers N, Pathiranage DR, Raj SS, Chan D, Kadler KE, Hoyland J, Meng QJ. The clock transcription factor BMAL1 is a key regulator of extracellular matrix homeostasis and cell fate in the intervertebral disc. Matrix Biol 2023; 122:1-9. [PMID: 37495193 DOI: 10.1016/j.matbio.2023.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/28/2023] [Accepted: 07/21/2023] [Indexed: 07/28/2023]
Abstract
The circadian clock in mammals temporally coordinates physiological and behavioural processes to anticipate daily rhythmic changes in their environment. Chronic disruption to circadian rhythms (e.g., through ageing or shift work) is thought to contribute to a multitude of diseases, including degeneration of the musculoskeletal system. The intervertebral disc (IVD) in the spine contains circadian clocks which control ∼6% of the transcriptome in a rhythmic manner, including key genes involved in extracellular matrix (ECM) homeostasis. However, it remains largely unknown to what extent the local IVD molecular clock is required to drive rhythmic gene transcription and IVD physiology. In this work, we identified profound age-related changes of ECM microarchitecture and an endochondral ossification-like phenotype in the annulus fibrosus (AF) region of the IVD in the Col2a1-Bmal1 knockout mice. Circadian time series RNA-Seq of the whole IVD in Bmal1 knockout revealed loss of circadian patterns in gene expression, with an unexpected emergence of 12 h ultradian rhythms, including FOXO transcription factors. Further RNA sequencing of the AF tissue identified region-specific changes in gene expression, evidencing a loss of AF phenotype markers and a dysregulation of ECM and FOXO pathways in Bmal1 knockout mice. Consistent with an up-regulation of FOXO1 mRNA and protein levels in Bmal1 knockout IVDs, inhibition of FOXO1 in AF cells suppressed their osteogenic differentiation. Collectively, these data highlight the importance of the local molecular clock mechanism in the maintenance of the cell fate and ECM homeostasis of the IVD. Further studies may identify potential new molecular targets for alleviating IVD degeneration.
Collapse
Affiliation(s)
- Michal Dudek
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Wellcome Centre for Cell Matrix Research, University of Manchester, Oxford Road, Manchester, UK; Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK
| | - Honor Morris
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Wellcome Centre for Cell Matrix Research, University of Manchester, Oxford Road, Manchester, UK; Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK
| | - Natalie Rogers
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Wellcome Centre for Cell Matrix Research, University of Manchester, Oxford Road, Manchester, UK; Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK
| | - Dharshika Rj Pathiranage
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Wellcome Centre for Cell Matrix Research, University of Manchester, Oxford Road, Manchester, UK; Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK
| | - Sujitha Saba Raj
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Wellcome Centre for Cell Matrix Research, University of Manchester, Oxford Road, Manchester, UK; Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Karl E Kadler
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Wellcome Centre for Cell Matrix Research, University of Manchester, Oxford Road, Manchester, UK
| | - Judith Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Wellcome Centre for Cell Matrix Research, University of Manchester, Oxford Road, Manchester, UK; Central Manchester Foundation Trust, Manchester Academic Health Science Centre, NIHR Manchester Biomedical Research Centre, Oxford Road, Manchester, UK.
| | - Qing-Jun Meng
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Wellcome Centre for Cell Matrix Research, University of Manchester, Oxford Road, Manchester, UK; Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, UK.
| |
Collapse
|
29
|
Kontomaris SV, Stylianou A, Chliveros G, Malamou A. A New Elementary Method for Determining the Tip Radius and Young's Modulus in AFM Spherical Indentations. MICROMACHINES 2023; 14:1716. [PMID: 37763878 PMCID: PMC10536531 DOI: 10.3390/mi14091716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 08/26/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
Atomic force microscopy (AFM) is a powerful tool for characterizing biological materials at the nanoscale utilizing the AFM nanoindentation method. When testing biological materials, spherical indenters are typically employed to reduce the possibility of damaging the sample. The accuracy of determining Young's modulus depends, among other factors, on the calibration of the indenter, i.e., the determination of the tip radius. This paper demonstrates that the tip radius can be approximately calculated using a single force-indentation curve on an unknown, soft sample without performing any additional experimental calibration process. The proposed method is based on plotting a tangent line on the force indentation curve at the maximum indentation depth. Subsequently, using equations that relate the applied force, maximum indentation depth, and the tip radius, the calculation of the tip radius becomes trivial. It is significant to note that the method requires only a single force-indentation curve and does not necessitate knowledge of the sample's Young's modulus. Consequently, the determination of both the sample's Young's modulus and the tip radius can be performed simultaneously. Thus, the experimental effort is significantly reduced. The method was tested on 80 force-indentation curves obtained on an agarose gel, and the results were accurate.
Collapse
Affiliation(s)
- Stylianos Vasileios Kontomaris
- Faculty of Engineering and Architecture, Metropolitan College, 15125 Athens, Greece;
- BioNanoTec Ltd., Nicosia 2043, Cyprus
| | - Andreas Stylianou
- School of Sciences, European University Cyprus, Nicosia 2404, Cyprus;
| | - Georgios Chliveros
- Faculty of Engineering and Architecture, Metropolitan College, 15125 Athens, Greece;
| | - Anna Malamou
- Independent Power Transmission Operator S.A. (IPTO), 10443 Athens, Greece;
| |
Collapse
|
30
|
Chang Z, Zhang J, Liu Y, Gao H, Xu GK. New Mechanical Markers for Tracking the Progression of Myocardial Infarction. NANO LETTERS 2023; 23:7350-7357. [PMID: 37580044 PMCID: PMC10450805 DOI: 10.1021/acs.nanolett.3c01712] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/09/2023] [Indexed: 08/16/2023]
Abstract
The mechanical properties of soft tissues can often be strongly correlated with the progression of various diseases, such as myocardial infarction (MI). However, the dynamic mechanical properties of cardiac tissues during MI progression remain poorly understood. Herein, we investigate the rheological responses of cardiac tissues at different stages of MI (i.e., early-stage, mid-stage, and late-stage) with atomic force microscopy-based microrheology. Surprisingly, we discover that all cardiac tissues exhibit a universal two-stage power-law rheological behavior at different time scales. The experimentally found power-law exponents can capture an inconspicuous initial rheological change, making them particularly suitable as markers for early-stage MI diagnosis. We further develop a self-similar hierarchical model to characterize the progressive mechanical changes from subcellular to tissue scales. The theoretically calculated mechanical indexes are found to markedly vary among different stages of MI. These new mechanical markers are applicable for tracking the subtle changes of cardiac tissues during MI progression.
Collapse
Affiliation(s)
- Zhuo Chang
- Laboratory
for Multiscale Mechanics and Medical Science, State Key Laboratory
for Strength and Vibration of Mechanical Structures, School of Aerospace
Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Jing Zhang
- Department
of Cardiovascular Medicine, The First Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, 710061, China
| | - Yilun Liu
- Laboratory
for Multiscale Mechanics and Medical Science, State Key Laboratory
for Strength and Vibration of Mechanical Structures, School of Aerospace
Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Huajian Gao
- School
of Mechanical and Aerospace Engineering, College of Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Institute
of High Performance Computing, A*STAR, Singapore 138632, Singapore
| | - Guang-Kui Xu
- Laboratory
for Multiscale Mechanics and Medical Science, State Key Laboratory
for Strength and Vibration of Mechanical Structures, School of Aerospace
Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
31
|
Chan B, Glogauer M, Wang Y, Wrana J, Chan K, Beier F, Bali S, Hinz B, Parreno J, Ashraf S, Kandel R. Adseverin, an actin-binding protein, modulates hypertrophic chondrocyte differentiation and osteoarthritis progression. SCIENCE ADVANCES 2023; 9:eadf1130. [PMID: 37540756 PMCID: PMC10403223 DOI: 10.1126/sciadv.adf1130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 07/06/2023] [Indexed: 08/06/2023]
Abstract
In osteoarthritis (OA), a disease characterized by progressive articular cartilage degradation and calcification, the articular chondrocyte phenotype changes and this correlates with actin cytoskeleton alterations suggesting that it regulates gene expression essential for proper phenotype. This study reports that OA is associated with the loss of adseverin, an actin capping and severing protein. Adseverin deletion (Adseverin-/-) in mice compromised articular chondrocyte function, by reducing F-actin and aggrecan expression and increasing apoptosis, Indian hedgehog, Runx2, MMP13, and collagen type X expression, and cell proliferation. This led to stiffer cartilage and decreased hyaline and increased calcified cartilage thickness. Together, these changes predisposed the articular cartilage to enhanced OA severity in Adseverin-/- mice who underwent surgical induction of OA. Adseverin-/- chondrocyte RNA sequencing and in vitro studies together suggests that adseverin modulates cell viability and prevents mineralization. Thus, adseverin maintains articular chondrocyte phenotype and cartilage tissue homeostasis by preventing progression to hypertrophic differentiation in vivo. Adseverin may be chondroprotective and a potential therapeutic target.
Collapse
Affiliation(s)
- Byron Chan
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Yongqiang Wang
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Jeffrey Wrana
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Kin Chan
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Supinder Bali
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
- Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON, Canada
| | - Justin Parreno
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Sajjad Ashraf
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Rita Kandel
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Zhang S, Weng Z, Wang Z, Wang B, Zeng Y, Li J, Hu C. Attenuation of alcohol-induced hepatocyte damage by ginsenoside Rg1 evaluated using atomic force microscopy. Microsc Res Tech 2023; 86:1037-1046. [PMID: 37382340 DOI: 10.1002/jemt.24381] [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: 02/28/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/30/2023]
Abstract
Alcoholic liver disease is an important cause of death worldwide. Hepatocyte apoptosis is commonly observed in alcoholic liver disease. In this study, we investigated the effect of ginsenoside Rg1 (G-Rg1), an organic component of ginseng, on the alcohol-induced morphological and biophysical properties of hepatocytes. Human hepatocytes (HL-7702) were treated in vitro with alcohol and G-Rg1. The cell morphology was observed using scanning electron microscopy. Cell height, roughness, adhesion, and elastic modulus were detected using atomic force microscopy. We found that alcohol significantly induced hepatocyte apoptosis, whereas G-Rg1 attenuated the alcohol-induced hepatocyte damage. Scanning electron microscopy revealed that alcohol-induced significant morphological changes in hepatocytes, including decreased cell contraction, roundness, and pseudopods, whereas G-Rg1 inhibited these negative changes. Atomic force microscopy revealed that alcohol increased the cell height and decreased the adhesion and elastic modulus of hepatocytes. Following treatment with G-Rg1, the cell height, adhesion, and elastic modulus of alcohol-injured hepatocytes were all similar to those of normal cells. Thus, G-Rg1 can attenuate the alcohol-induced damage to hepatocytes by modulating the morphology and biomechanics of the cells. RESEARCH HIGHLIGHTS: In this study, the morphological characteristics of hepatocytes were observed using SEM. The changes in hepatocyte three-dimensional images and biomechanical action caused by alcohol and G-Rg1 were examined at the nanoscale using AFM under near-physiological conditions. Alcohol-induced hepatocytes showed abnormal morphology and biophysical properties. G-Rg1 attenuated the alcohol-induced damage to hepatocytes by modulating the morphology and biomechanics of the cells.
Collapse
Affiliation(s)
- Shengli Zhang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Zhankun Weng
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Zuobin Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
- JR3CN & IRAC, University of Bedfordshire, Luton, UK
| | - Bowei Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Yi Zeng
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Jiani Li
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| | - Cuihua Hu
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun, China
- Ministry of Education Key Laboratory for Cross-Scale Micro and Nano Manufacturing, Changchun University of Science and Technology, Changchun, China
| |
Collapse
|
33
|
Iijima H, Wang K, D'Amico E, Tang WY, Rogers RJ, Jakicic JM, Ambrosio F. Exercise-primed extracellular vesicles improve cell-matrix adhesion and chondrocyte health. RESEARCH SQUARE 2023:rs.3.rs-2958821. [PMID: 37333349 PMCID: PMC10274961 DOI: 10.21203/rs.3.rs-2958821/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Extracellular vesicles (EVs) have been suggested to transmit the health-promoting effects of exercise throughout the body. Yet, the mechanisms by which beneficial information is transmitted from extracellular vesicles to recipient cells are poorly understood, precluding a holistic understanding of how exercise promotes cellular and tissue health. In this study, using articular cartilage as a model, we introduced a network medicine paradigm to simulate how exercise facilitates communication between circulating EVs and chondrocytes, the cells resident in articular cartilage. Using the archived small RNA-seq data of EV before and after aerobic exercise, microRNA regulatory network analysis based on network propagation inferred that circulating EVs activated by aerobic exercise perturb chondrocyte-matrix interactions and downstream cellular aging processes. Building on the mechanistic framework identified through computational analyses, follow up experimental studies interrogated the direct influence of exercise on EV-mediated chondrocyte-matrix interactions. We found that pathogenic matrix signaling in chondrocytes was abrogated in the presence of exercise-primed EVs, restoring a more youthful phenotype, as determined by chondrocyte morphological profiling and evaluation of chondrogenicity. Epigenetic reprograming of the gene encoding the longevity protein, α-Klotho, mediated these effects. These studies provide mechanistic evidence that exercise transduces rejuvenation signals to circulating EVs, endowing EVs with the capacity to ameliorate cellular health even in the presence of an unfavorable microenvironmental signals.
Collapse
Affiliation(s)
- Hirotaka Iijima
- Institute for Advanced Research, Nagoya University, Nagoya, Japan
- Biomedical and Health Informatics Unit, Graduate School of Medicine, Nagoya University, Nagoya, Japan
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
| | - Kai Wang
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA
| | - Ella D'Amico
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
| | - Wan-Yee Tang
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | - Renee J Rogers
- Department of Internal Medicine, Division of Physical Activity and Weight Management, University of Kansas Medical Center, Kansas City, KS
| | - John M Jakicic
- Department of Internal Medicine, Division of Physical Activity and Weight Management, University of Kansas Medical Center, Kansas City, KS
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA
| |
Collapse
|
34
|
Bernabei I, Hansen U, Ehirchiou D, Brinckmann J, Chobaz V, Busso N, Nasi S. CD11b Deficiency Favors Cartilage Calcification via Increased Matrix Vesicles, Apoptosis, and Lysyl Oxidase Activity. Int J Mol Sci 2023; 24:ijms24119776. [PMID: 37298730 DOI: 10.3390/ijms24119776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Pathological cartilage calcification is a hallmark feature of osteoarthritis, a common degenerative joint disease, characterized by cartilage damage, progressively causing pain and loss of movement. The integrin subunit CD11b was shown to play a protective role against cartilage calcification in a mouse model of surgery-induced OA. Here, we investigated the possible mechanism by which CD11b deficiency could favor cartilage calcification by using naïve mice. First, we found by transmission electron microscopy (TEM) that CD11b KO cartilage from young mice presented early calcification spots compared with WT. CD11b KO cartilage from old mice showed progression of calcification areas. Mechanistically, we found more calcification-competent matrix vesicles and more apoptosis in both cartilage and chondrocytes isolated from CD11b-deficient mice. Additionally, the extracellular matrix from cartilage lacking the integrin was dysregulated with increased collagen fibrils with smaller diameters. Moreover, we revealed by TEM that CD11b KO cartilage had increased expression of lysyl oxidase (LOX), the enzyme that catalyzes matrix crosslinks. We confirmed this in murine primary CD11b KO chondrocytes, where Lox gene expression and crosslinking activity were increased. Overall, our results suggest that CD11b integrin regulates cartilage calcification through reduced MV release, apoptosis, LOX activity, and matrix crosslinking. As such, CD11b activation might be a key pathway for maintaining cartilage integrity.
Collapse
Affiliation(s)
- Ilaria Bernabei
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University Hospital of Münster, 48149 Münster, Germany
| | - Driss Ehirchiou
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Jürgen Brinckmann
- Department of Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Veronique Chobaz
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Nathalie Busso
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Sonia Nasi
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, 1011 Lausanne, Switzerland
| |
Collapse
|
35
|
Duan M, Xia S, Liu Y, Pu X, Chen Y, Zhou Y, Huang M, Pi C, Zhang D, Xie J. Stiffened fibre-like microenvironment based on patterned equidistant micropillars directs chondrocyte hypertrophy. Mater Today Bio 2023; 20:100682. [PMID: 37304578 PMCID: PMC10251154 DOI: 10.1016/j.mtbio.2023.100682] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/04/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Articular cartilage, composed of collagen type II as a major extracellular matrix and chondrocyte as a unique cell type, is a specialized connective tissue without blood vessels, lymphatic vessels and nerves. This distinctive characteristic of articular cartilage determines its very limited ability to repair when damaged. It is well known that physical microenvironmental signals regulate many cell behaviors such as cell morphology, adhesion, proliferation and cell communication even determine chondrocyte fate. Interestingly, with increasing age or progression of joint diseases such as osteoarthritis (OA), the major collagen fibrils in the extracellular matrix of articular cartilage become larger in diameter, leading to stiffening of articular tissue and reducing its resistance to external tension, which in turn aggravates joint damage or progression of joint diseases. Therefore, designing a physical microenvironment closer to the real tissue and thus obtaining data closer to the real cellular behaviour, and then revealing the biological mechanisms of chondrocytes in pathological states is of crucial importance for the treatment of OA disease. Here we fabricated micropillar substrates with the same topology but different stiffnesses to mimic the matrix stiffening that occurs in the transition from normal to diseased cartilage. It was first found that chondrocytes responded to stiffened micropillar substrates by showing a larger cell spreading area, a stronger enhancement of cytoskeleton rearrangement and more stability of focal adhesion plaques. The activation of Erk/MAPK signalling in chondrocytes was detected in response to the stiffened micropillar substrate. Interestingly, a larger nuclear spreading area of chondrocytes at the interface layer between the cells and top surfaces of micropillars was observed in response to the stiffened micropillar substrate. Finally, it was found that the stiffened micropillar substrate promoted chondrocyte hypertrophy. Taken together, these results revealed the cell responses of chondrocytes in terms of cell morphology, cytoskeleton, focal adhesion, nuclei and cell hypertrophy, and may be beneficial for understanding the cellular functional changes affected by the matrix stiffening that occurs during the transition from a normal state to a state of osteoarthritis.
Collapse
Affiliation(s)
- Mengmeng Duan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Shuang Xia
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu, 610065, China
| | - Yang Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaohua Pu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yukun Chen
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu, 610065, China
| | - Yilin Zhou
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Chengdu, 610065, China
| | - Minglei Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Caixia Pi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
36
|
Wang X, Wu Q, Zhang R, Fan Z, Li W, Mao R, Du Z, Yao X, Ma Y, Yan Y, Sun W, Wu H, Wei W, Hu Y, Hong Y, Hu H, Koh YW, Duan W, Chen X, Ouyang H. Stage-specific and location-specific cartilage calcification in osteoarthritis development. Ann Rheum Dis 2023; 82:393-402. [PMID: 36261249 DOI: 10.1136/ard-2022-222944] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/30/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVES This study investigated the stage-specific and location-specific deposition and characteristics of minerals in human osteoarthritis (OA) cartilages via multiple nano-analytical technologies. METHODS Normal and OA cartilages were serially sectioned for micro-CT, scanning electron microscopy with energy dispersive X-ray spectroscopy, micro-Raman spectroscopy, focused ion beam scanning electron microscopy, high-resolution electron energy loss spectrometry with transmission electron microscopy, nanoindentation and atomic force microscopy to analyse the structural, compositional and mechanical properties of cartilage in OA progression. RESULTS We found that OA progressed by both top-down calcification at the joint surface and bottom-up calcification at the osteochondral interface. The top-down calcification process started with spherical mineral particle formation in the joint surface during early-stage OA (OA-E), followed by fibre formation and densely packed material transformation deep into the cartilage during advanced-stage OA (OA-A). The bottom-up calcification in OA-E started when an excessive layer of calcified tissue formed above the original calcified cartilage, exhibiting a calcified sandwich structure. Over time, the original and upper layers of calcified cartilage fused, which thickened the calcified cartilage region and disrupted the cartilage structure. During OA-E, the calcified cartilage was hypermineralised, containing stiffer carbonated hydroxyapatite (HAp). During OA-A, it was hypomineralised and contained softer HAp. This discrepancy may be attributed to matrix vesicle nucleation during OA-E and carbonate cores during OA-A. CONCLUSIONS This work refines our current understanding of the mechanism underlying OA progression and provides the foothold for potential therapeutic targeting strategies once the location-specific cartilage calcification features in OA are established.
Collapse
Affiliation(s)
- Xiaozhao Wang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Qin Wu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ru Zhang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Zhang Fan
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wenyue Li
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Renwei Mao
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining, Zhejiang, China
| | - Zihao Du
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining, Zhejiang, China
| | - Xudong Yao
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yuanzhu Ma
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Yiyang Yan
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Wei Sun
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Hongwei Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Wei Wei
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Yejun Hu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Yi Hong
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Huan Hu
- ZJU-UIUC Institute, International Campus, Zhejiang University, Haining, Zhejiang, China
| | - Yi Wen Koh
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wangping Duan
- Department of Orthopedics, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiao Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China .,Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, and Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, CORMed, Hangzhou, Zhejiang, China
| |
Collapse
|
37
|
Paesa M, Alejo T, Garcia-Alvarez F, Arruebo M, Mendoza G. New insights in osteoarthritis diagnosis and treatment: Nano-strategies for an improved disease management. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1844. [PMID: 35965293 DOI: 10.1002/wnan.1844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 06/02/2022] [Accepted: 07/12/2022] [Indexed: 11/07/2022]
Abstract
Osteoarthritis (OA) is a common chronic joint pathology that has become a predominant cause of disability worldwide. Even though the origin and evolution of OA rely on different factors that are not yet elucidated nor understood, the development of novel strategies to treat OA has emerged in the last years. Cartilage degradation is the main hallmark of the pathology though alterations in bone and synovial inflammation, among other comorbidities, are also involved during OA progression. From a molecular point of view, a vast amount of signaling pathways are implicated in the progression of the disease, opening up a wide plethora of targets to attenuate or even halt OA. The main purpose of this review is to shed light on the recent strategies published based on nanotechnology for the early diagnosis of the disease as well as the most promising nano-enabling therapeutic approaches validated in preclinical models. To address the clinical issue, the key pathways involved in OA initiation and progression are described as the main potential targets for OA prevention and early treatment. Furthermore, an overview of current therapeutic strategies is depicted. Finally, to solve the drawbacks of current treatments, nanobiomedicine has shown demonstrated benefits when using drug delivery systems compared with the administration of the equivalent doses of the free drugs and the potential of disease-modifying OA drugs when using nanosystems. We anticipate that the development of smart and specific bioresponsive and biocompatible nanosystems will provide a solid and promising basis for effective OA early diagnosis and treatment. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.
Collapse
Affiliation(s)
- Monica Paesa
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Zaragoza, Spain
| | - Teresa Alejo
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Zaragoza, Spain
- Health Research Institute Aragon (IIS Aragon), Zaragoza, Spain
| | - Felicito Garcia-Alvarez
- Health Research Institute Aragon (IIS Aragon), Zaragoza, Spain
- Hospital Clínico Universitario Lozano Blesa, Department of Orthopedic Surgery & Traumatology, University of Zaragoza, Zaragoza, Spain
| | - Manuel Arruebo
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Aragón Materials Science Institute, ICMA, Zaragoza, Spain
- Health Research Institute Aragon (IIS Aragon), Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
| | - Gracia Mendoza
- Health Research Institute Aragon (IIS Aragon), Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, Madrid, Spain
| |
Collapse
|
38
|
Arnold KM, Sicard D, Tschumperlin DJ, Westendorf JJ. Atomic Force Microscopy Micro-Indentation Methods for Determining the Elastic Modulus of Murine Articular Cartilage. SENSORS (BASEL, SWITZERLAND) 2023; 23:1835. [PMID: 36850434 PMCID: PMC9967621 DOI: 10.3390/s23041835] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
The mechanical properties of biological tissues influence their function and can predict degenerative conditions before gross histological or physiological changes are detectable. This is especially true for structural tissues such as articular cartilage, which has a primarily mechanical function that declines after injury and in the early stages of osteoarthritis. While atomic force microscopy (AFM) has been used to test the elastic modulus of articular cartilage before, there is no agreement or consistency in methodologies reported. For murine articular cartilage, methods differ in two major ways: experimental parameter selection and sample preparation. Experimental parameters that affect AFM results include indentation force and cantilever stiffness; these are dependent on the tip, sample, and instrument used. The aim of this project was to optimize these experimental parameters to measure murine articular cartilage elastic modulus by AFM micro-indentation. We first investigated the effects of experimental parameters on a control material, polydimethylsiloxane gel (PDMS), which has an elastic modulus on the same order of magnitude as articular cartilage. Experimental parameters were narrowed on this control material, and then finalized on wildtype C57BL/6J murine articular cartilage samples that were prepared with a novel technique that allows for cryosectioning of epiphyseal segments of articular cartilage and long bones without decalcification. This technique facilitates precise localization of AFM measurements on the murine articular cartilage matrix and eliminates the need to separate cartilage from underlying bone tissues, which can be challenging in murine bones because of their small size. Together, the new sample preparation method and optimized experimental parameters provide a reliable standard operating procedure to measure microscale variations in the elastic modulus of murine articular cartilage.
Collapse
Affiliation(s)
- Katherine M. Arnold
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Delphine Sicard
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel J. Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
39
|
Gao J, Ren P, Gong H. Morphological and mechanical alterations in articular cartilage and subchondral bone during spontaneous hip osteoarthritis in guinea pigs. Front Bioeng Biotechnol 2023; 11:1080241. [PMID: 36756384 PMCID: PMC9900117 DOI: 10.3389/fbioe.2023.1080241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Objectives: This study aimed to investigate the morphological and mechanical changes in articular cartilage and subchondral bone during spontaneous hip osteoarthritis in guinea pigs. Materials and methods: Hip joints of guinea pigs were investigated at 1, 3, 6, and 9 months of age (hereafter denoted as 1 M, 3 M, 6 M, and 9 M, respectively; n = 7 in each group). Morphological and mechanical alterations during spontaneous hip osteoarthritis in guinea pigs were investigated. The alterations included the micromechanical properties of articular cartilage (stiffness and creep deformation), microstructure of the subchondral bone (bone mineral density, bone volume fraction, trabecular thickness, trabecular number, and trabecular separation), micromorphology of the articular cartilage, and surface nanostructure (grain size and roughness) of the articular cartilage and subchondral bone. Results: Micromechanical properties of articular cartilage in 1 M showed the lowest stiffness and highest creep deformation with no significant differences in stiffness or creep deformation amongst 3 M, 6 M, and 9 M. Articular cartilage thickness decreased with age. The earliest degeneration of articular cartilage occurred at 6 months of age, characterised by surface unevenness and evident chondrocytes reduction in micromorphology, as well as increased grain size and decreased roughness in nanostructure. No degeneration at micro- or nanostructure of subchondral bone was observed before 9 months. Conclusion: Morphological degeneration of cartilage occurred before degeneration of mechanical properties. Meanwhile, degeneration of cartilage occurred before degeneration of subchondral bone during hip osteoarthritis. The current study provided novel insights into the structural and micromechanical interaction of hip osteoarthritis, which can serve as a theoretical basis for understanding the formation and progression of osteoarthritis.
Collapse
Affiliation(s)
- Jiazi Gao
- Department of Engineering Mechanics, Nanling Campus, Jilin University, Changchun, China
| | - Pengling Ren
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - He Gong
- Department of Engineering Mechanics, Nanling Campus, Jilin University, Changchun, China,*Correspondence: He Gong,
| |
Collapse
|
40
|
Kontomaris SV, Stylianou A, Georgakopoulos A, Malamou A. 3D AFM Nanomechanical Characterization of Biological Materials. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:395. [PMID: 36770357 PMCID: PMC9920073 DOI: 10.3390/nano13030395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
Atomic Force Microscopy (AFM) is a powerful tool enabling the mechanical characterization of biological materials at the nanoscale. Since biological materials are highly heterogeneous, their mechanical characterization is still considered to be a challenging procedure. In this paper, a new approach that leads to a 3-dimensional (3D) nanomechanical characterization is presented based on the average Young's modulus and the AFM indentation method. The proposed method can contribute to the clarification of the variability of the mechanical properties of biological samples in the 3-dimensional space (variability at the x-y plane and depth-dependent behavior). The method was applied to agarose gels, fibroblasts, and breast cancer cells. Moreover, new mathematical methods towards a quantitative mechanical characterization are also proposed. The presented approach is a step forward to a more accurate and complete characterization of biological materials and could contribute to an accurate user-independent diagnosis of various diseases such as cancer in the future.
Collapse
Affiliation(s)
- Stylianos Vasileios Kontomaris
- BioNanoTec Ltd., 2043 Nicosia, Cyprus
- Faculty of Engineering and Architecture, Metropolitan College, 15125 Athens, Greece
| | - Andreas Stylianou
- School of Sciences, European University Cyprus, 2404 Nicosia, Cyprus
| | - Anastasios Georgakopoulos
- School of Electrical and Computer Engineering, National Technical University of Athens, 15780 Athens, Greece
| | - Anna Malamou
- School of Electrical and Computer Engineering, National Technical University of Athens, 15780 Athens, Greece
| |
Collapse
|
41
|
Jalandhra GK, Molley TG, Hung TT, Roohani I, Kilian KA. In situ formation of osteochondral interfaces through "bone-ink" printing in tailored microgel suspensions. Acta Biomater 2023; 156:75-87. [PMID: 36055612 DOI: 10.1016/j.actbio.2022.08.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/21/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2023]
Abstract
Osteochondral tissue has a complex hierarchical structure spanning subchondral bone to articular cartilage. Biomaterials approaches to mimic and repair these interfaces have had limited success, largely due to challenges in fabricating composite hard-soft interfaces with living cells. Biofabrication approaches have emerged as attractive methods to form osteochondral analogues through additive assembly of hard and soft components. We have developed a unique printing platform that is able to integrate soft and hard materials concurrently through freeform printing of mineralized constructs within tunable microgel suspensions containing living cells. A library of microgels based on gelatin were prepared, where the stiffness of the microgels and a liquid "filler" phase can be tuned for bioprinting while simultaneously directing differentiation. Tuning microgel stiffness and filler content differentially directs chondrogenesis and osteogenesis within the same construct, demonstrating how this technique can be used to fabricate osteochondral interfaces in a single step. Printing of a rapidly setting calcium phosphate cement, so called "bone-ink" within a cell laden suspension bath further guides differentiation, where the cells adjacent to the nucleated hydroxyapatite phase undergo osteogenesis with cells in the surrounding medium undergoing chondrogenesis. In this way, bone analogues with hierarchical structure can be formed within cell-laden gradient soft matrices to yield multiphasic osteochondral constructs. This technique provides a versatile one-pot biofabrication approach without harsh post-processing which will aid efforts in bone disease modelling and tissue engineering. STATEMENT OF SIGNIFICANCE: This paper demonstrates the first example of a biofabrication approach to rapidly form osteochondral constructs in a single step under physiological conditions. Key to this advance is a tunable suspension of extracellular matrix microgels that are packed together with stem cells, providing a unique and modular scaffolding for guiding the simultaneous formation of bone and cartilage tissue. The physical properties of the suspension allow direct writing of a ceramic "bone-ink", resulting in an ordered structure of microscale hydrogels, living cells, and bone mimics in a single step. This platform reveals a simple approach to making complex skeletal tissue for disease modelling, with the possibility of repairing and replacing bone-cartilage interfaces in the clinic using a patient's own cells.
Collapse
Affiliation(s)
- Gagan K Jalandhra
- School of Materials Science and Engineering, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052
| | - Thomas G Molley
- School of Materials Science and Engineering, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052
| | - Tzong-Tyng Hung
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney NSW 2052
| | - Iman Roohani
- School of Chemistry, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052
| | - Kristopher A Kilian
- School of Materials Science and Engineering, University of New South Wales, Sydney NSW 2052; School of Chemistry, University of New South Wales, Sydney NSW 2052; Australian Centre for NanoMedicine, University of New South Wales, Sydney NSW 2052.
| |
Collapse
|
42
|
Kontomaris SV, Stylianou A, Chliveros G, Malamou A. Determining Spatial Variability of Elastic Properties for Biological Samples Using AFM. MICROMACHINES 2023; 14:mi14010182. [PMID: 36677243 PMCID: PMC9862197 DOI: 10.3390/mi14010182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/26/2022] [Accepted: 01/09/2023] [Indexed: 05/29/2023]
Abstract
Measuring the mechanical properties (i.e., elasticity in terms of Young's modulus) of biological samples using Atomic Force Microscopy (AFM) indentation at the nanoscale has opened new horizons in studying and detecting various pathological conditions at early stages, including cancer and osteoarthritis. It is expected that AFM techniques will play a key role in the future in disease diagnosis and modeling using rigorous mathematical criteria (i.e., automated user-independent diagnosis). In this review, AFM techniques and mathematical models for determining the spatial variability of elastic properties of biological materials at the nanoscale are presented and discussed. Significant issues concerning the rationality of the elastic half-space assumption, the possibility of monitoring the depth-dependent mechanical properties, and the construction of 3D Young's modulus maps are also presented.
Collapse
Affiliation(s)
- Stylianos Vasileios Kontomaris
- BioNanoTec Ltd., Nicosia 2043, Cyprus
- Faculty of Engineering and Architecture, Metropolitan College, 15125 Athens, Greece
| | - Andreas Stylianou
- School of Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Georgios Chliveros
- Faculty of Engineering and Architecture, Metropolitan College, 15125 Athens, Greece
| | - Anna Malamou
- School of Electrical and Computer Engineering, National Technical University of Athens, 15780 Athens, Greece
| |
Collapse
|
43
|
Iijima H, Gilmer G, Wang K, Bean AC, He Y, Lin H, Tang WY, Lamont D, Tai C, Ito A, Jones JJ, Evans C, Ambrosio F. Age-related matrix stiffening epigenetically regulates α-Klotho expression and compromises chondrocyte integrity. Nat Commun 2023; 14:18. [PMID: 36627269 PMCID: PMC9832042 DOI: 10.1038/s41467-022-35359-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/29/2022] [Indexed: 01/12/2023] Open
Abstract
Extracellular matrix stiffening is a quintessential feature of cartilage aging, a leading cause of knee osteoarthritis. Yet, the downstream molecular and cellular consequences of age-related biophysical alterations are poorly understood. Here, we show that epigenetic regulation of α-Klotho represents a novel mechanosensitive mechanism by which the aged extracellular matrix influences chondrocyte physiology. Using mass spectrometry proteomics followed by a series of genetic and pharmacological manipulations, we discovered that increased matrix stiffness drove Klotho promoter methylation, downregulated Klotho gene expression, and accelerated chondrocyte senescence in vitro. In contrast, exposing aged chondrocytes to a soft matrix restored a more youthful phenotype in vitro and enhanced cartilage integrity in vivo. Our findings demonstrate that age-related alterations in extracellular matrix biophysical properties initiate pathogenic mechanotransductive signaling that promotes Klotho promoter methylation and compromises cellular health. These findings are likely to have broad implications even beyond cartilage for the field of aging research.
Collapse
Affiliation(s)
- Hirotaka Iijima
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA.
- Japan Society for the Promotion of Science, Tokyo, Japan.
- Institute for Advanced Research, Nagoya University, Nagoya, Japan.
| | - Gabrielle Gilmer
- Medical Scientist Training Program, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Cellular and Molecular Pathology Graduate Program, University of Pittsburgh, Pittsburgh, PA, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Kai Wang
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA
| | - Allison C Bean
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuchen He
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hang Lin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wan-Yee Tang
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Daniel Lamont
- Petersen Institute of Nanoscience and Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chia Tai
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Jeffrey J Jones
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, USA
| | - Christopher Evans
- Department of Physical Medicine & Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA, USA.
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
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: 1.0] [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
|
45
|
Masson AO, Besler B, Edwards WB, Krawetz RJ. High spatial resolution analysis using automated indentation mapping differentiates biomechanical properties of normal vs. degenerated articular cartilage in mice. eLife 2022; 11:74664. [PMID: 36444976 DOI: 10.7554/elife.74664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/28/2022] [Indexed: 11/30/2022] Open
Abstract
Characterizing the biomechanical properties of articular cartilage is crucial to understanding processes of tissue homeostasis vs. degeneration. In mouse models, however, limitations are imposed by their small joint size and thin cartilage surfaces. Here we present a three-dimensional (3D) automated surface mapping system and methodology that allows for mechanical characterization of mouse cartilage with high spatial resolution. We performed repeated indentation mappings, followed by cartilage thickness measurement via needle probing, at 31 predefined positions distributed over the medial and lateral femoral condyles of healthy mice. High-resolution 3D x-ray microscopy (XRM) imaging was used to validate tissue thickness measurements. The automated indentation mapping was reproducible, and needle probing yielded cartilage thicknesses comparable to XRM imaging. When comparing healthy vs. degenerated cartilage, topographical variations in biomechanics were identified, with altered thickness and stiffness (instantaneous modulus) across condyles and within anteroposterior sub-regions. This quantitative technique comprehensively characterized cartilage function in mice femoral condyle cartilage. Hence, it has the potential to improve our understanding of tissue structure-function interplay in mouse models of repair and disease.
Collapse
Affiliation(s)
- Anand O Masson
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada
| | - Bryce Besler
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada
| | - W Brent Edwards
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada.,Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | - Roman J Krawetz
- Biomedical Engineering Graduate Program, University of Calgary, Calgary, Canada.,McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Canada.,Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
46
|
Moo EK, Ebrahimi M, Sibole SC, Tanska P, Korhonen RK. The intrinsic quality of proteoglycans, but not collagen fibres, degrades in osteoarthritic cartilage. Acta Biomater 2022; 153:178-189. [PMID: 36113721 DOI: 10.1016/j.actbio.2022.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/01/2022]
Abstract
The function of articular cartilage as a load-bearing connective tissue is derived primarily from a balanced interaction between the swelling proteoglycan (PG) matrix and tension-resistant collagen fibrous network. Such balance is compromised during joint disease such as osteoarthritis (OA) due to degradation to PGs and/or collagens. While the PG degradation is generally thought to be related to a loss of protein abundance, the collagenous degradation is more complex as it can be caused independently by a decrease of collagen content, disorganisation of fibrous structure and softening of individual collagen fibrils. A comprehensive understanding of the initial trajectories of degradation of PGs and collagen network can improve our chance of finding potential therapeutic solutions for OA. Here, we developed geometrically, structurally, and compositionally realistic and sample-specific Finite Element (FE) models under the framework of multiphasic mixture theory, from which the elastic moduli of collagen fibres and the PG load-bearing quality in healthy and diseased cartilages were estimated by numerical optimisation of the multi-step indentation stress relaxation force-time curves. We found the intrinsic quality of collagen fibres, measured by their elastic moduli, to stay constant for healthy and diseased cartilages. Combining with previous findings which show unaltered collagen content during early stages of OA, our results suggest the disorganisation of collagen fibrous network as the first form of collagenous degradation in osteoarthritic cartilage. We also found that PG degradation involves not only a loss of protein abundance, but also the quality of the remaining PGs in generating sufficient osmotic pressure for load bearing. This study sheds light on the mechanism of OA pathogenesis and highlights the restoration of collageneous organisation in cartilage as key medical intervention for OA. STATEMENT OF SIGNIFICANCE: Collagen network in articular cartilage consists of individual fibres that are organised into depth-dependent structure specialised for joint load-bearing and lubrication. During osteoarthritis, the collagen network undergoes mechanical degradation, but it is unclear if a loss of content, disorganisation of fibrous structure, or softening of individual fibres causes this degeneration. Using mechanical indentation, Finite Element modelling, and numerical optimisation methods, we determined that individual fibres did not soften in early disease stage. Together with previous findings showing unaltered collagen content, our results pinpoint the disorganisation of collagen structure as the main culprit for early collagenous degradation in osteoarthritic cartilage. Thus, early restoration in cartilage of collagen organisation, instead of individual fibre quality, may be key to slow osteoarthritis development.
Collapse
Affiliation(s)
- Eng Kuan Moo
- Department of Applied Physics, University of Eastern Finland, POB 1627, Kuopio 70211, Finland; Human Performance Laboratory, University of Calgary, 2500, University Drive NW, Calgary, Alberta 2N1N4, Canada.
| | | | - Scott C Sibole
- Human Performance Laboratory, University of Calgary, 2500, University Drive NW, Calgary, Alberta 2N1N4, Canada
| | - Petri Tanska
- Department of Applied Physics, University of Eastern Finland, POB 1627, Kuopio 70211, Finland
| | - Rami K Korhonen
- Department of Applied Physics, University of Eastern Finland, POB 1627, Kuopio 70211, Finland.
| |
Collapse
|
47
|
Forrester LA, Fang F, Jacobsen T, Hu Y, Kurtaliaj I, Roye BD, Guo XE, Chahine NO, Thomopoulos S. Transient neonatal shoulder paralysis causes early osteoarthritis in a mouse model. J Orthop Res 2022; 40:1981-1992. [PMID: 34812543 PMCID: PMC9124737 DOI: 10.1002/jor.25225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/08/2021] [Accepted: 11/20/2021] [Indexed: 02/04/2023]
Abstract
Neonatal brachial plexus palsy (NBPP) occurs in approximately 1.5 of every 1,000 live births. The majority of children with NBPP recover function of the shoulder. However, the long-term risk of osteoarthritis (OA) in this population is unknown. The purpose of this study was to investigate the development of OA in a mouse model of transient neonatal shoulder paralysis. Neonatal mice were injected twice per week for 4 weeks with saline in the right supraspinatus muscle (Saline, control) and botulinum toxin A (BtxA, transient paralysis) in the left supraspinatus muscle, and then allowed to recover for 20 or 36 weeks. Control mice received no injections, and all mice were sacrificed at 24 or 40 weeks. BtxA mice exhibited abnormalities in gait compared to controls through 10 weeks of age, but these differences did not persist into adulthood. BtxA shoulders had decreased bone volume (-9%) and abnormal trabecular microstructure compared to controls. Histomorphometry analysis demonstrated that BtxA shoulders had higher murine shoulder arthritis scale scores (+30%), and therefore more shoulder OA compared to controls. Articular cartilage of BtxA shoulders demonstrated stiffening of the tissue. Compared with controls, articular cartilage from BtxA shoulders had 2-fold and 10-fold decreases in Dkk1 and BMP2 expression, respectively, and 3-fold and 14-fold increases in Col10A1 and BGLAP expression, respectively, consistent with established models of OA. In summary, a brief period of paralysis of the neonatal mouse shoulder was sufficient to generate early signs of OA in adult cartilage and bone.
Collapse
Affiliation(s)
- Lynn Ann Forrester
- Department of Orthopedic Surgery, Columbia University, New York, New York, USA
| | - Fei Fang
- Department of Orthopedic Surgery, Columbia University, New York, New York, USA
| | - Timothy Jacobsen
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Yizhong Hu
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Iden Kurtaliaj
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Benjamin D. Roye
- Department of Orthopedic Surgery, Columbia University, New York, New York, USA
| | - X. Edward Guo
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Nadeen O. Chahine
- Department of Orthopedic Surgery, Columbia University, New York, New York, USA
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, New York, USA
- Department of Biomedical Engineering, Columbia University, New York, New York, USA
| |
Collapse
|
48
|
Tschaikowsky M, Brander S, Barth V, Thomann R, Rolauffs B, Balzer BN, Hugel T. The articular cartilage surface is impaired by a loss of thick collagen fibers and formation of type I collagen in early osteoarthritis. Acta Biomater 2022; 146:274-283. [PMID: 35487427 DOI: 10.1016/j.actbio.2022.04.036] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/03/2022] [Accepted: 04/21/2022] [Indexed: 12/14/2022]
Abstract
Osteoarthritis (OA) is a joint disease affecting millions of patients worldwide. During OA onset and progression, the articular cartilage is destroyed, but the underlying complex mechanisms remain unclear. Here, we uncover changes in the thickness of collagen fibers and their composition at the onset of OA. For articular cartilage explants from knee joints of OA patients, we find that type I collagen-rich fibrocartilage-like tissue was formed in macroscopically intact cartilage, distant from OA lesions. Importantly, the number of thick fibers (>100 nm) has decreased early in the disease, followed by complete absence of thick fibers in advanced OA. We have obtained these results by a combination of high-resolution atomic force microscopy imaging under near-native conditions, immunofluorescence, scanning electron microscopy and a fluorescence-based classification of the superficial chondrocyte spatial organization. Taken together, our data suggests that the loss of tissue functionality in early OA cartilage is caused by a reduction of thick type II collagen fibers, likely due to the formation of type I collagen-rich fibrocartilage, followed by the development of focal defects in later OA stages. We anticipate that such an integrative characterization will be very beneficial for an in-depth understanding of other native biological tissues and the development of sustainable biomaterials. STATEMENT OF SIGNIFICANCE: In early osteoarthritis (OA) the cartilage appears macroscopically intact. However, this study demonstrates that the collagen network already changes in early OA by collagen fiber thinning and the formation of fibrocartilage-like tissue. Both nanoscopic deficiencies already occur in macroscopically intact regions of the human knee joint and are likely connected to processes that result in a weakened extracellular matrix. This study enhances the understanding of earliest progressive cartilage degeneration in the absence of external damage. The results suggest a determination of the mean collagen fiber thickness as a new target for the detection of early OA and a regulation of type I collagen synthesis as a new path for OA treatment.
Collapse
|
49
|
Perera D, Wang Q, Schniepp HC. Multi-Point Nanoindentation Method to Determine Mechanical Anisotropy in Nanofibrillar Thin Films. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202065. [PMID: 35780468 DOI: 10.1002/smll.202202065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/26/2022] [Indexed: 06/15/2023]
Abstract
Biomaterials with outstanding mechanical properties, including spider silk, wood, and cartilage, often feature an oriented nanofibrillar structure. The orientation of nanofibrils gives rise to a significant mechanical anisotropy, which is extremely challenging to characterize, especially for microscopically small or inhomogeneous samples. Here, a technique utilizing atomic force microscope indentation at multiple points combined with finite element analysis to sample the mechanical anisotropy of a thin film in a microscopically small area is reported. The system studied here is the tape-like silk of the Chilean recluse spider, which entirely consists of strictly oriented nanofibrils giving rise to a large mechanical anisotropy. The most detailed directional nanoscale structure-property characterization of spider silk to date is presented, revealing the tensile and transverse elastic moduli as 9 and 1 GPa, respectively, and the binding strength between silk nanofibrils as 159 ± 13 MPa. Furthermore, based on this binding strength, the nanofibrils' surface energy is derived as 37 mJ m-2 , and concludes that van der Waals forces play a decisive role in interfibrillar binding. Due to its versatility, this technique has many potential applications, including early disease diagnostics, as underlying pathological conditions can alter the local mechanical properties of tissues.
Collapse
Affiliation(s)
- Dinidu Perera
- Department of Applied Science, William & Mary, P.O. Box 8795, Williamsburg, VA, 23187-8795, USA
| | - Qijue Wang
- Department of Applied Science, William & Mary, P.O. Box 8795, Williamsburg, VA, 23187-8795, USA
| | - Hannes C Schniepp
- Department of Applied Science, William & Mary, P.O. Box 8795, Williamsburg, VA, 23187-8795, USA
| |
Collapse
|
50
|
Jaabar IL, Cornette P, Miche A, Wanherdrick K, Dupres V, Ehkirch FP, Cambon Binder A, Berenbaum F, Houard X, Landoulsi J. Deciphering pathological remodelling of the human cartilage extracellular matrix in osteoarthritis at the supramolecular level. NANOSCALE 2022; 14:8691-8708. [PMID: 35673929 DOI: 10.1039/d2nr00474g] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The extracellular matrix (ECM) of articular cartilage is a three-dimensional network mainly constituted of entangled collagen fibrils and interfibrillar aggrecan aggregates. During the development of osteoarthritis (OA), the most common musculoskeletal disorder, the ECM is subjected to a combination of chemical and structural changes that play a pivotal role in the initiation and the progress of the disease. While the molecular mechanisms involved in the pathological remodelling of the ECM are considered as decisive, they remain, however, not completely elucidated. Herein, we report a relevant way for unravelling the role and nature of OA progress on human cartilage tissues, in terms of chemical composition and morphological and mechanical properties at the level of supramolecular assemblies constituting the cartilage ECM. For this purpose, we used X-ray photoelectron spectroscopy (XPS), and developed an innovative methodological approach that provides the molecular composition of the ECM. Moreover, we used atomic force microscopy (AFM) to probe the tissues at the level of individual collagen fibrils, both imaging and force spectroscopy modes being explored to this end. Taken together, these nanoscale characterization studies reveal the existence of two stages in the OA progress. At the early stage, a marked increase in the aggrecan and collagen content is observed, reflecting the homeostatic chondrocyte activity that tends to repair the cartilage ECM. At the late stage, we observe a failed attempt to stabilize and/or restore the tissue, yielding significant degradation of the supramolecular assemblies. This suggests an imbalance in the chondrocyte activity that turns in favor of catabolic events. Chemical changes are also accompanied by ECM structural changes and stiffening. Interestingly, we showed the possibility to mimic the imbalanced activities of chondrocytes by applying enzymatic digestions of healthy cartilage, through the combined action of hyaluronidase and collagenase. This yields damage strictly analogous to that observed at high OA severity. These findings bring mechanistic insights leading to a better understanding of the mechanism by which OA is initiated and progresses in the cartilage ECM. They offer guidelines for the development of curative treatments, such as targeting the homeostatic balance of chondrocyte metabolism through the control of enzymatic reactions involved in catabolic processes.
Collapse
Affiliation(s)
- Ilhem Lilia Jaabar
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Pauline Cornette
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Antoine Miche
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
| | - Kristell Wanherdrick
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Vincent Dupres
- Université Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000, Lille, France
| | - François-Paul Ehkirch
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
- Clinique Maussins-Nollet, F-75019 Paris, France
| | - Adeline Cambon Binder
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
- Orthopedics and Hand Surgery Department, Saint-Antoine Hospital, 184 Rue du Faubourg Saint-Antoine, Paris, 75012, France
| | - Francis Berenbaum
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
- Rheumatology Department, AP-HP Saint-Antoine Hospital, 184, rue du Faubourg Saint-Antoine, 75012, Paris, France
| | - Xavier Houard
- Sorbonne Université, INSERM (UMR_S938), Centre de Recherche Saint-Antoine, CRSA, F-75012 Paris, France.
| | - Jessem Landoulsi
- Sorbonne Université, CNRS, Laboratoire de Réactivité de Surface, LRS, F-75005 Paris, France.
| |
Collapse
|