51
|
Huang L, Li P, Guo L, Li L, Yuan J, Zhao R, Li H, Wei X. Zinc finger protein 521 attenuates osteoarthritis via the histone deacetylases 4 in the nucleus. Bioengineered 2022; 13:14489-14502. [PMID: 36694467 PMCID: PMC9995124 DOI: 10.1080/21655979.2022.2090203] [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] [Indexed: 01/26/2023] Open
Abstract
To determine whether zinc finger protein 521 (Zfp521) has a chondroprotective effect by maintaining extracellular matrix (ECM) homeostasis to attenuate osteoarthritis (OA). In chondrocytes, Zfp521 was overexpressed or silenced to detect its effects on proliferation, apoptosis, and ECM homeostasis. Adenovirus encoding Zfp521 was injected into the knee joints of anterior cruciate ligament transection rats to test its efficacy against OA. Combined with proteomic analysis, the molecular mechanism of Zfp521 in cartilage degeneration was further explored. An intra-articular injection of adenovirus carrying a Zfp521 sequence showed a chondroprotective effect against OA. The molecular mechanism around Zfp521 was classified at the molecular, cellular, histological, and functional levels. It was reported that Zfp521 could effectively promote cartilage proliferation, inhibit apoptosis, and maintain the balance of anabolism and catabolism of ECM. Moreover, it was confirmed that Zfp521 exerted its effect better by upregulating histone deacetylases 4 (HDAC4) in the nucleus and was significantly weakened in the absence of HDAC4 in the nucleus. Overall, Zfp521 better exerts its efficacy against OA by increasing the HDAC4 content in the nucleus, making it a promising strategy for OA treatment.
Collapse
Affiliation(s)
- Lingan Huang
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi Province, China
| | - Pengcui Li
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi Province, China
| | - Li Guo
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi Province, China
| | - Lu Li
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi Province, China
| | - Jie Yuan
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi Province, China
| | - Ruipeng Zhao
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi Province, China
| | - Haoqian Li
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi Province, China
| | - Xiaochun Wei
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi Province, China
| |
Collapse
|
52
|
Willantarra I, Leung S, Choi YS, Chhana A, McGlashan SR. Chondrocyte-specific response to stiffness-mediated primary cilia formation and centriole positioning. Am J Physiol Cell Physiol 2022; 323:C236-C247. [PMID: 35649254 DOI: 10.1152/ajpcell.00135.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mechanical stress and the stiffness of the extracellular matrix are key drivers of tissue development and homeostasis. Aberrant mechanosensation is associated with a wide range of pathologies, including osteoarthritis. Matrix (or substrate) stiffness plays a major role in cell spreading, adhesion, proliferation and differentiation. However, how specific cells sense substrate stiffness still remains unclude. The primary cilium is an essential cellular organelle that senses and integrates mechanical and chemical signals from the extracellular environment. We hypothesised that the primary cilium dynamically alters its length and position to fine-tune cell mechanosignalling based on substrate stiffness alone. We used a hydrogel system of varying substrate stiffness to examine the role of stiffness on cilia frequency, length and centriole position as well as cell and nuclei area over time. Contrary to other cell types, we show that chondrocyte primary cilia shorten on softer substrates demonstrating tissue-specific mechanosensing which is aligned with the tissue stiffness the cells originate from. We further show that stiffness determines centriole positioning to either the basal or apical membrane during attachment and spreading, with centriole positioned towards the basal membrane on stiffer substrates. These phenomena are mediated by force generation actin-myosin stress fibres in a time-dependent manner. Finally we show on stiff substrates, that primary cilia are involved in tension-mediated cell spreading. We propose that substrate stiffness plays a role in cilia positioning, regulating cellular responses to external forces, and may be a key driver of mechanosignalling-associated diseases.
Collapse
Affiliation(s)
- Ivanna Willantarra
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sophia Leung
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Yu Suk Choi
- School of Human Sciences, University of Western Australia, Perth, Australia
| | - Ashika Chhana
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sue R McGlashan
- Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
53
|
Atomic Force Microscopy Nanoindentation Method on Collagen Fibrils. MATERIALS 2022; 15:ma15072477. [PMID: 35407813 PMCID: PMC8999528 DOI: 10.3390/ma15072477] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/20/2022] [Accepted: 03/24/2022] [Indexed: 12/24/2022]
Abstract
Atomic Force Microscopy nanoindentation method is a powerful technique that can be used for the nano-mechanical characterization of bio-samples. Significant scientific efforts have been performed during the last two decades to accurately determine the Young’s modulus of collagen fibrils at the nanoscale, as it has been proven that mechanical alterations of collagen are related to various pathological conditions. Different contact mechanics models have been proposed for processing the force–indentation data based on assumptions regarding the shape of the indenter and collagen fibrils and on the elastic or elastic–plastic contact assumption. However, the results reported in the literature do not always agree; for example, the Young’s modulus values for dry collagen fibrils expand from 0.9 to 11.5 GPa. The most significant parameters for the broad range of values are related to the heterogeneous structure of the fibrils, the water content within the fibrils, the data processing errors, and the uncertainties in the calibration of the probe. An extensive discussion regarding the models arising from contact mechanics and the results provided in the literature is presented, while new approaches with respect to future research are proposed.
Collapse
|
54
|
Assessing Collagen D-Band Periodicity with Atomic Force Microscopy. MATERIALS 2022; 15:ma15041608. [PMID: 35208148 PMCID: PMC8877100 DOI: 10.3390/ma15041608] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/01/2023]
Abstract
The collagen superfamily includes more than fifty collagen and/or collagen-like proteins with fibril-forming collagen type I being the most abundant protein within the extracellular matrix. Collagen type I plays a crucial role in a variety of functions, it has been associated with many pathological conditions and it is widely used due to its unique properties. One unique nano-scale characteristic of natural occurring collagen type I fibers is the so-called D-band periodicity, which has been associated with collagen natural structure and properties, while it seems to play a crucial role in the interactions between cells and collagen and in various pathological conditions. An accurate characterization of the surface and structure of collagen fibers, including D-band periodicity, on collagen-based tissues and/or (nano-)biomaterials can be achieved by Atomic Force Microscopy (AFM). AFM is a scanning probe microscope and is among the few techniques that can assess D-band periodicity. This review covers issues related to collagen and collagen D-band periodicity and the use of AFM for studying them. Through a systematic search in databases (PubMed and Scopus) relevant articles were identified. The study of these articles demonstrated that AFM can offer novel information concerning D-band periodicity. This study highlights the importance of studying collagen D-band periodicity and proves that AFM is a powerful tool for investigating a number of different properties related to collagen D-band periodicity.
Collapse
|
55
|
Cornette P, Jaabar IL, Dupres V, Werthel JD, Berenbaum F, Houard X, Landoulsi J, Nourissat G. Impact of Collagen Crosslinking on Dislocated Human Shoulder Capsules-Effect on Structural and Mechanical Properties. Int J Mol Sci 2022; 23:ijms23042297. [PMID: 35216412 PMCID: PMC8877509 DOI: 10.3390/ijms23042297] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/04/2023] Open
Abstract
Classical treatments of shoulder instability are associated with recurrence. To determine whether the modification of the capsule properties may be an alternative procedure, the effect of crosslinking treatment on the structure and mechanical properties of diseased human shoulder capsules was investigated. Joint capsules harvested from patients during shoulder surgery (n = 5) were treated or not with UV and/or riboflavin (0.1%, 1.0% and 2.5%). The structure and the mechanical properties of the capsules were determined by atomic force microscopy. The effect of treatments on cell death was investigated. Collagen fibrils were well-aligned and adjacent to each other with a D-periodicity of 66.9 ± 3.2 nm and a diameter of 71.8 ± 15.4 nm in control untreated capsules. No effect of treatments was observed on the organization of the collagen fibrils nor on their intrinsic characteristics, including D-periodicity or their mean diameter. The treatments also did not induce cell death. In contrast, UV + 2.5% riboflavin induced capsule stiffness, as revealed by the increased Young's modulus values (p < 0.0001 for each patient). Our results showed that the crosslinking procedure changed the biomechanics of diseased capsules, while keeping their structural organisation unchanged at the single fibril level. The UV/riboflavin crosslinking procedure may be a promising way to preserve the functions of collagen-based tissues and tune their elasticity for clinically relevant treatments.
Collapse
Affiliation(s)
- Pauline Cornette
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (P.C.); (I.L.J.); (F.B.); (X.H.)
- Laboratoire de Réactivité de Surface, CNRS, Sorbonne Université, F-75005 Paris, France;
| | - Ilhem Lilia Jaabar
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (P.C.); (I.L.J.); (F.B.); (X.H.)
- Laboratoire de Réactivité de Surface, CNRS, Sorbonne Université, F-75005 Paris, France;
| | - Vincent Dupres
- U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Institut Pasteur de Lille, CHU Lille, Inserm, CNRS, Université Lille, F-59000 Lille, France;
| | - Jean-David Werthel
- Department of Orthopedy and Traumatology, AP-HP Ambroise Paré Hospital, F-92100 Boulogne-Billancourt, France;
| | - Francis Berenbaum
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (P.C.); (I.L.J.); (F.B.); (X.H.)
- Department of Rheumatology, AP-HP Saint-Antoine Hospital, DMU 3iD, F-75012 Paris, France
| | - Xavier Houard
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (P.C.); (I.L.J.); (F.B.); (X.H.)
| | - Jessem Landoulsi
- Laboratoire de Réactivité de Surface, CNRS, Sorbonne Université, F-75005 Paris, France;
| | - Geoffroy Nourissat
- Centre de Recherche Saint-Antoine (CRSA), INSERM, Sorbonne Université, F-75012 Paris, France; (P.C.); (I.L.J.); (F.B.); (X.H.)
- Clinique des Maussins, 67 Rue de Romainville, F-75019 Paris, France
- Correspondence:
| |
Collapse
|
56
|
Cykowska A, Danalache M, Bonnaire FC, Feierabend M, Hofmann UK. Detecting early osteoarthritis through changes in biomechanical properties - A review of recent advances in indentation technologies in a clinical arthroscopic setup. J Biomech 2022; 132:110955. [PMID: 35042088 DOI: 10.1016/j.jbiomech.2022.110955] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/29/2021] [Accepted: 01/07/2022] [Indexed: 11/19/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disease currently affecting half of all women and one-third of all men aged over 65 and it is predicted to even increase in the next decades. In the variety of causes leading to OA, the first common denominator are changes in the extracellular matrix of the cartilage. In later stages, OA affects the whole joint spreading to higher levels of tissue architecture causing irreversible functional and structural damage. To date, the diagnosis of OA is only formulated in the late stages of the disease. This is also, where most present therapies apply. Since a precise diagnosis is a prerequisite for targeted therapy, tools to diagnose early OA, monitor its progression, and accurately stage the disease are wanted. This review article focuses on recent advances in indentation technologies to diagnose early OA through describing biomechanical cartilage characteristics. We provide an overview of microindentation instruments, indentation-type Atomic Force Microscopy, ultrasound, and water-jet ultrasound indentation, Optical Coherence Tomography-based air-jet indentation, as well as fiber Bragg grating.
Collapse
Affiliation(s)
- Anna Cykowska
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, D-72072 Tübingen, Germany.
| | - Marina Danalache
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, D-72072 Tübingen, Germany.
| | - Florian Christof Bonnaire
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, D-72072 Tübingen, Germany; Department of Orthopaedic Surgery, University Hospital of Tübingen, D-72076 Tübingen, Germany.
| | - Martina Feierabend
- Department of Computational Systems Biology, Faculty of Science of the University of Tübingen, D-72076 Tübingen, Germany.
| | - Ulf Krister Hofmann
- Laboratory of Cell Biology, Department of Orthopaedic Surgery, University Hospital of Tübingen, D-72072 Tübingen, Germany; Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074 Aachen, Germany.
| |
Collapse
|
57
|
Ye J, Jin Z, Chen S, Guo W. Uridine relieves MSCs and chondrocyte senescence in vitvo and exhibits the potential to treat osteoarthritis in vivo. Cell Cycle 2022; 21:33-48. [PMID: 34974808 PMCID: PMC8837230 DOI: 10.1080/15384101.2021.2010170] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative disease of extremely high incidence in the elderly. Therefore, anti-aging may be an important prerequisite for treating OA. The senescence of chondrocytes and mesenchymal stem cells (MSCs) is one of the important factors that causes OA. Here, the effect of uridine (which is a functional food derived from plants or animals) on senescence of chondrocytes and MSCs was evaluated in in vivo and in vitro experiments. For this, we established the senescence model of chondrocyte and MSCs in vitro, and established the OA model in vivo, and a series of experiments (such as CLSM, ELISA, Western blot, etc.) were conducted to evaluate the effect of uridine on chondrocyte and MSCs senescence. The results showed that uridine could alleviate chondrocyte and MSCs senescence in vitro by evaluating a series of aging markers. Furthermore, uridine could also relieve OA in vivo. In summary, in the present work, we found that uridine can alleviate chondrocyte and MSCs senescence in in vitro and in vivo experiments. Uridine has shown great potential in the treatment of OA in vivo, suggesting that uridine could be used to treat and prevent OA induced by aging, and has potential clinical applications in future.
Collapse
Affiliation(s)
- Jia Ye
- Department of Orthopedics, People’s Hospital of Wuhan University, Wuhan, China,CONTACT Jia Ye Department of Orthopedics, People’s Hospital of Wuhan University, No. 99, Zhangzhidong Road, Wuchang District, Wuhan, Hubei, China
| | - Zhihui Jin
- Department of Orthopedics, People’s Hospital of Wuhan University, Wuhan, China
| | - Sen Chen
- Department of Orthopedics, People’s Hospital of Wuhan University, Wuhan, China
| | - Weichun Guo
- Department of Orthopedics, People’s Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
58
|
The Hertzian theory in AFM nanoindentation experiments regarding biological samples: Overcoming limitations in data processing. Micron 2022; 155:103228. [DOI: 10.1016/j.micron.2022.103228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 12/09/2021] [Accepted: 01/26/2022] [Indexed: 11/21/2022]
|
59
|
Rellmann Y, Eidhof E, Hansen U, Fleischhauer L, Vogel J, Clausen-Schaumann H, Aszodi A, Dreier R. ER Stress in ERp57 Knockout Knee Joint Chondrocytes Induces Osteoarthritic Cartilage Degradation and Osteophyte Formation. Int J Mol Sci 2021; 23:ijms23010182. [PMID: 35008608 PMCID: PMC8745280 DOI: 10.3390/ijms23010182] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/14/2021] [Accepted: 12/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ageing or obesity are risk factors for protein aggregation in the endoplasmic reticulum (ER) of chondrocytes. This condition is called ER stress and leads to induction of the unfolded protein response (UPR), which, depending on the stress level, restores normal cell function or initiates apoptotic cell death. Here the role of ER stress in knee osteoarthritis (OA) was evaluated. It was first tested in vitro and in vivo whether a knockout (KO) of the protein disulfide isomerase ERp57 in chondrocytes induces sufficient ER stress for such analyses. ER stress in ERp57 KO chondrocytes was confirmed by immunofluorescence, immunohistochemistry, and transmission electron microscopy. Knee joints of wildtype (WT) and cartilage-specific ERp57 KO mice (ERp57 cKO) were analyzed by indentation-type atomic force microscopy (IT-AFM), toluidine blue, and immunofluorescence/-histochemical staining. Apoptotic cell death was investigated by a TUNEL assay. Additionally, OA was induced via forced exercise on a treadmill. ER stress in chondrocytes resulted in a reduced compressive stiffness of knee cartilage. With ER stress, 18-month-old mice developed osteoarthritic cartilage degeneration with osteophyte formation in knee joints. These degenerative changes were preceded by apoptotic death in articular chondrocytes. Young mice were not susceptible to OA, even when subjected to forced exercise. This study demonstrates that ER stress induces the development of age-related knee osteoarthritis owing to a decreased protective function of the UPR in chondrocytes with increasing age, while apoptosis increases. Therefore, inhibition of ER stress appears to be an attractive therapeutic target for OA.
Collapse
Affiliation(s)
- Yvonne Rellmann
- Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstraße 15, 48149 Muenster, Germany; (Y.R.); (E.E.)
| | - Elco Eidhof
- Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstraße 15, 48149 Muenster, Germany; (Y.R.); (E.E.)
| | - Uwe Hansen
- Institute of Musculoskeletal Medicine, University Hospital Münster, Albert-Schweitzer-Campus 1, Building D3, 48149 Muenster, Germany;
| | - Lutz Fleischhauer
- Center for Applied Tissue Engineering and Regenerative Medicine-CANTER, Munich University of Applied Sciences, 80335 Munich, Germany; (L.F.); (J.V.); (H.C.-S.)
- Center for Nanoscience-CeNS, 80335 Munich, Germany
- Department for Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, 80335 Munich, Germany;
| | - Jonas Vogel
- Center for Applied Tissue Engineering and Regenerative Medicine-CANTER, Munich University of Applied Sciences, 80335 Munich, Germany; (L.F.); (J.V.); (H.C.-S.)
- Center for Nanoscience-CeNS, 80335 Munich, Germany
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine-CANTER, Munich University of Applied Sciences, 80335 Munich, Germany; (L.F.); (J.V.); (H.C.-S.)
- Center for Nanoscience-CeNS, 80335 Munich, Germany
| | - Attila Aszodi
- Department for Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, 80335 Munich, Germany;
| | - Rita Dreier
- Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstraße 15, 48149 Muenster, Germany; (Y.R.); (E.E.)
- Correspondence: ; Tel.: +49-251-8355573
| |
Collapse
|
60
|
Main and Minor Types of Collagens in the Articular Cartilage: The Role of Collagens in Repair Tissue Evaluation in Chondral Defects. Int J Mol Sci 2021; 22:ijms222413329. [PMID: 34948124 PMCID: PMC8706311 DOI: 10.3390/ijms222413329] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 12/15/2022] Open
Abstract
Several collagen subtypes have been identified in hyaline articular cartilage. The main and most abundant collagens are type II, IX and XI collagens. The minor and less abundant collagens are type III, IV, V, VI, X, XII, XIV, XVI, XXII, and XXVII collagens. All these collagens have been found to play a key role in healthy cartilage, regardless of whether they are more or less abundant. Additionally, an exhaustive evaluation of collagen fibrils in a repaired cartilage tissue after a chondral lesion is necessary to determine the quality of the repaired tissue and even whether or not this repaired tissue is considered hyaline cartilage. Therefore, this review aims to describe in depth all the collagen types found in the normal articular cartilage structure, and based on this, establish the parameters that allow one to consider a repaired cartilage tissue as a hyaline cartilage.
Collapse
|
61
|
Nirmalraj PN, Schneider T, Felbecker A. Spatial organization of protein aggregates on red blood cells as physical biomarkers of Alzheimer's disease pathology. SCIENCE ADVANCES 2021; 7:eabj2137. [PMID: 34559561 PMCID: PMC8462905 DOI: 10.1126/sciadv.abj2137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/04/2021] [Indexed: 06/13/2023]
Abstract
Quantifying physical differences of protein aggregates implicated in Alzheimer’s disease (AD), in blood, could provide crucial information on disease stages. Here, red blood cells (RBCs) from 50 patients with neurocognitive complaints and 16 healthy individuals were profiled using an atomic force microscope (AFM). AFM measurements revealed patient age– and stage of neurocognitive disorder–dependent differences in size, shape, morphology, assembly, and prevalence of protein aggregates on RBCs, referred to as physical biomarkers. Crystals composed of fibrils were exclusively detected on RBCs for AD patients aged above 80 years. Fibril prevalence was negatively correlated with the cerebrospinal fluid (CSF) β-amyloid (Aβ) 42/40 ratio and was observed to be higher in the Aβ-positive patient category. Using a cutoff of ≥40% fibril prevalence, the CSF Aβ status was classified with 88% accuracy (sensitivity 100%, specificity 73%). The merits and challenges in integrating physical biomarkers in AD diagnosis are discussed.
Collapse
Affiliation(s)
- Peter Niraj Nirmalraj
- Transport at Nanoscale Interfaces Laboratory, Swiss Federal Laboratories for Materials Science and Technology, Dübendorf CH-8600, Switzerland
| | - Thomas Schneider
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen CH-9007, Switzerland
| | - Ansgar Felbecker
- Department of Neurology, Cantonal Hospital St. Gallen, St. Gallen CH-9007, Switzerland
| |
Collapse
|
62
|
Chen Z, Liang T, Li J, Gao C, He F, Luo Z, Yang H, Zhang H, Yu J. Early detection of joint degeneration in the subtalar and talonavicular joints secondary to ankle instability in a mouse model. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2021. [DOI: 10.1016/j.medntd.2021.100080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
63
|
Destouni A, Tsolis KC, Economou A, Papathanasiou I, Balis C, Mourmoura E, Tsezou A. Chondrocyte protein co-synthesis network analysis links ECM mechanosensing to metabolic adaptation in osteoarthritis. Expert Rev Proteomics 2021; 18:623-635. [PMID: 34348542 DOI: 10.1080/14789450.2021.1962299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Knee osteoarthritis (OA) is one of the most common structural OA disorders globally. Incomplete understanding of the fundamental biological aspects of osteoarthritis underlies the current lack of effective treatment or disease modifying drugs. RESEARCH DESIGN AND METHODS We implemented a systems approach by making use of the statistical network concepts in Weighted Gene Co-expression Analysis to reconstruct the organization of the core proteome network in chondrocytes obtained from OA patients and healthy individuals. Protein modules reflect groups of tightly co-ordinated changes in protein abundance across healthy and OA chondrocytes. RESULTS The unbiased systems analysis identified extracellular matrix (ECM) mechanosensing and glycolysis as two modules that are most highly correlated with ΟΑ. The ECM module was enriched in the OA genetic risk factors tenascin-C (TNC) and collagen 11A1 (COL11A1), as well as in cartilage oligomeric matrix protein (COMP), a biomarker associated with cartilage integrity. Mapping proteins that are unique to OA or healthy chondrocytes onto the core interactome, which connects microenvironment sensing and regulation of glycolysis, identified differences in metabolic and anti-inflammatory adaptation. CONCLUSION The interconnection between cartilage ECM remodeling and metabolism is indicative of the dynamic chondrocyte states and their significance in osteoarthritis.
Collapse
Affiliation(s)
- Aspasia Destouni
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Konstantinos C Tsolis
- KULeuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Molecular Bacteriology, Leuven, Belgium
| | - Anastassios Economou
- KULeuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Molecular Bacteriology, Leuven, Belgium
| | - Ioanna Papathanasiou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece.,Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Charalampos Balis
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Evanthia Mourmoura
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece.,Department of Biology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| |
Collapse
|
64
|
Ren P, Niu H, Cen H, Jia S, Gong H, Fan Y. Biochemical and Morphological Abnormalities of Subchondral Bone and Their Association with Cartilage Degeneration in Spontaneous Osteoarthritis. Calcif Tissue Int 2021; 109:179-189. [PMID: 33715052 DOI: 10.1007/s00223-021-00834-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 02/25/2021] [Indexed: 10/21/2022]
Abstract
This study aims to investigate how biochemical composition in subchondral bone (SB) relates to the sulfated glycosaminoglycan (sGAG) content of articular cartilage (AC) in the knee joint of guinea pigs from the early to moderate osteoarthritis (OA). Male Dunkin Hartley strain guinea pigs were grouped according to age (1, 3, 6, and 9 months, with 10 guinea pigs in each group). The biochemical properties of the AC and SB in the tibial plateau of the guinea pigs were determined through histology and Raman spectroscopy, respectively. Furthermore, the microstructures of the SB were investigated using micro-computed tomography (micro-CT) and histology. Increased thickness and bone mineral density (BMD) and decreased porosity were observed in the subchondral plate (SP) with the progression of spontaneous OA, accompanied by a decreasing trend in sGAG integrated optical density (IOD) of AC. Compared with the changes in the microstructure of subchondral bone, the content of sGAG was more correlated to the changes in the mineral/matrix ratio of subchondral bone. The mineralization of the matrix was significantly correlated to the content of sGAG compared with crystallinity/maturity and Type B carbonate substitution. PO43- ν1/Amide III was more correlated to the content of sGAG than PO43- ν1/Amide I, PO43- ν1/CH2 wag during the progression of spontaneous osteoarthritis. This study demonstrated that the mineralization of subchondral bone plays a crucial role in the pathogenesis of OA. Future studies may access to the mineralization of subchondral bone in addition to its microstructure in the study for pathogenesis and early diagnosis of osteoarthritis.
Collapse
Affiliation(s)
- Pengling Ren
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Haijun Niu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China
| | - Haipeng Cen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China
| | - Shaowei Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China
| | - He Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China.
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing, People's Republic of China.
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China
| |
Collapse
|
65
|
Khoeini R, Nosrati H, Akbarzadeh A, Eftekhari A, Kavetskyy T, Khalilov R, Ahmadian E, Nasibova A, Datta P, Roshangar L, Deluca DC, Davaran S, Cucchiarini M, Ozbolat IT. Natural and Synthetic Bioinks for 3D Bioprinting. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000097] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Roghayeh Khoeini
- Department of Medicinal Chemistry Faculty of Pharmacy Tabriz University of Medical Sciences P.O. Box: 51664-14766 Tabriz Iran
- Drug Applied Research Center Tabriz University of Medical Sciences P.O. Box: 51656-65811 Tabriz Iran
| | - Hamed Nosrati
- Drug Applied Research Center Tabriz University of Medical Sciences P.O. Box: 51656-65811 Tabriz Iran
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
| | - Abolfazl Akbarzadeh
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Department of Medical Nanotechnology Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences P.O. Box: 516615731 Tabriz Iran
| | - Aziz Eftekhari
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Russian Institute for Advanced Study Moscow State Pedagogical University 1/1, Malaya Pirogovskaya Street Moscow 119991 Russian Federation
- Pharmacology and Toxicology Department Maragheh University of Medical Sciences 78151-55158 Maragheh Iran
- Department of Synthesis and Characterization of Polymers Polymer Institute Slovak Academy of Sciences (SAS) Dúbravská cesta 9 845 41 Bratislava Slovakia
| | - Taras Kavetskyy
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Department of Biology and Chemistry Drohobych Ivan Franko State Pedagogical University 24, I. Franko Str. 82100 Drohobych Ukraine
- Department of Surface Engineering The John Paul II Catholic University of Lublin 20-950 Lublin Poland
| | - Rovshan Khalilov
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Russian Institute for Advanced Study Moscow State Pedagogical University 1/1, Malaya Pirogovskaya Street Moscow 119991 Russian Federation
- Department of Biophysics and Biochemistry Faculty of Biology Baku State University Baku AZ 1143 Azerbaijan
- Institute of Radiation Problems National Academy of Sciences of Azerbaijan Baku AZ 1143 Azerbaijan
| | - Elham Ahmadian
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Kidney Research Center Tabriz University of Medical Sciences P.O. Box: 5166/15731 Tabriz Iran
| | - Aygun Nasibova
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Institute of Radiation Problems National Academy of Sciences of Azerbaijan Baku AZ 1143 Azerbaijan
| | - Pallab Datta
- Department of Pharmaceutics National Institute of Pharmaceutical Education and Research Kolkata West Bengal 700054 India
| | - Leila Roshangar
- Stem Cell Research Center Tabriz University of Medical Sciences P.O. Box: 5166/15731 Tabriz Iran
| | - Dante C. Deluca
- Agricultural and Biological Engineering Department Penn State University University Park 16802 PA USA
| | - Soodabeh Davaran
- Department of Medicinal Chemistry Faculty of Pharmacy Tabriz University of Medical Sciences P.O. Box: 51664-14766 Tabriz Iran
- Drug Applied Research Center Tabriz University of Medical Sciences P.O. Box: 51656-65811 Tabriz Iran
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 24, I. Franko Str. 82100 Drohobych Ukraine
- Joint Ukraine-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems 9 B.Vahabzade Str. 1143 Baku Azerbaijan
- Department of Medical Nanotechnology Faculty of Advanced Medical Sciences Tabriz University of Medical Sciences P.O. Box: 516615731 Tabriz Iran
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics Saarland University Medical Center Kirrbergerstr. Bldg 37 D-66421 Homburg/Saar Germany
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics Department Penn State University University Park 16802 PA USA
- The Huck Institutes of the Life Sciences Penn State University University Park 16802 PA USA
- Biomedical Engineering Department Penn State University University Park 16802 PA USA
- Materials Research Institute Penn State University University Park 16802 PA USA
- Department of Neurosurgery Penn State University Hershey 17033 PA USA
| |
Collapse
|
66
|
Madry H, Grässel S, Nöth U, Relja B, Bernstein A, Docheva D, Kauther MD, Katthagen JC, Bader R, van Griensven M, Wirtz DC, Raschke MJ, Huber-Lang M. The future of basic science in orthopaedics and traumatology: Cassandra or Prometheus? Eur J Med Res 2021; 26:56. [PMID: 34127057 PMCID: PMC8200553 DOI: 10.1186/s40001-021-00521-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/20/2021] [Indexed: 12/23/2022] Open
Abstract
Orthopaedic and trauma research is a gateway to better health and mobility, reflecting the ever-increasing and complex burden of musculoskeletal diseases and injuries in Germany, Europe and worldwide. Basic science in orthopaedics and traumatology addresses the complete organism down to the molecule among an entire life of musculoskeletal mobility. Reflecting the complex and intertwined underlying mechanisms, cooperative research in this field has discovered important mechanisms on the molecular, cellular and organ levels, which subsequently led to innovative diagnostic and therapeutic strategies that reduced individual suffering as well as the burden on the society. However, research efforts are considerably threatened by economical pressures on clinicians and scientists, growing obstacles for urgently needed translational animal research, and insufficient funding. Although sophisticated science is feasible and realized in ever more individual research groups, a main goal of the multidisciplinary members of the Basic Science Section of the German Society for Orthopaedics and Trauma Surgery is to generate overarching structures and networks to answer to the growing clinical needs. The future of basic science in orthopaedics and traumatology can only be managed by an even more intensified exchange between basic scientists and clinicians while fuelling enthusiasm of talented junior scientists and clinicians. Prioritized future projects will master a broad range of opportunities from artificial intelligence, gene- and nano-technologies to large-scale, multi-centre clinical studies. Like Prometheus in the ancient Greek myth, transferring the elucidating knowledge from basic science to the real (clinical) world will reduce the individual suffering from orthopaedic diseases and trauma as well as their socio-economic impact.
Collapse
Affiliation(s)
- Henning Madry
- Institute of Experimental Orthopaedics and Osteoarthritis Research, Saarland University, Homburg, Germany
| | - Susanne Grässel
- Experimental Orthopedics, Department of Orthopedic Surgery, University of Regensburg, Regensburg, Germany
| | - Ulrich Nöth
- Department of Orthopaedics and Trauma Surgery, Evangelisches Waldkrankenhaus Berlin Spandau, Berlin, Germany
| | - Borna Relja
- Experimental Radiology, University Clinic for Radiology and Nuclear Medicine, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Anke Bernstein
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Breisgau, Germany
| | - Denitsa Docheva
- Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Max Daniel Kauther
- Department of Trauma-, Hand- and Reconstructive Surgery, University Hospital Essen, Essen, Germany
| | - Jan Christoph Katthagen
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Rainer Bader
- Department of Orthopaedics, Research Lab for Biomechanics and Implant Technology, Rostock University Medical Center, Rostock, Germany
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN-Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, The Netherlands
| | - Dieter C Wirtz
- Department of Orthopaedics and Trauma Surgery, University Hopsital Bonn, Bonn, Germany
| | - Michael J Raschke
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Muenster, Muenster, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma-Immunology (ITI), University Hospital Ulm, Helmholzstr. 8/1, Ulm, Germany.
| |
Collapse
|
67
|
Krajčíková K, Semančíková E, Zakutanská K, Kondrakhova D, Mašlanková J, Stupák M, Talian I, Tomašovičová N, Kimáková T, Komanický V, Dubayová K, Breznoščáková D, Pálová E, Semančík J, Tomečková V. Tear fluid biomarkers in major depressive disorder: Potential of spectral methods in biomarker discovery. J Psychiatr Res 2021; 138:75-82. [PMID: 33836432 DOI: 10.1016/j.jpsychires.2021.03.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/09/2021] [Accepted: 03/19/2021] [Indexed: 12/22/2022]
Abstract
Spectroscopic methods represent a group of analytical methods that demonstrate high potential in providing clinically relevant diagnostic information, such as biochemical, functional or structural changes of macromolecular complexes that might occur due to pathological processes or therapeutic intervention. Although application of these methods in the field of psychiatric research is still relatively recent, the preliminary results show that they have the capacity to detect subtle neurobiological abnormalities in major depressive disorder (MDD). Methods of mass spectrometry (MALDI-TOF MS), zymography, synchronous fluorescence spectroscopy (SFS), circular dichroism (CD) spectroscopy, Fourier-transform infrared (FTIR) spectroscopy and atomic force microscopy (AFM) were used to analyze the human tear fluid of subjects with MDD. Using MALDI-TOF MS, two diagnostically significant peaks (3747 and 16 411 m/z) were identified with an AUC value of 0.89 and 0.92 in tear fluid of subjects with MDD vs controls, respectively. We also identified various forms of matrix metalloproteinase 9 in subjects with MDD using zymography and synchronous fluorescence spectra (SFS) showed a significant increase in fluorescence intensity at 280 nm. CD spectra were redshifted in tear fluid of subjects with MDD vs healthy controls. FTIR spectroscopy showed changes in the positions of peaks for amide A, I, II in tear fluid of subjects with MDD vs controls. Moreover, atomic force microscopy (AFM) showed different pattern in the crystal structures of tear fluid components in subjects with MDD. SFS, CD, FTIR spectroscopy, AFM and MALDI-TOF MS confirmed, that the human tear fluid proteome could be helpful in discriminating between the group of subjects with MDD and healthy controls. These preliminary findings suggest that spectral methods could represent a useful tool in clinical psychiatry, especially in establishing differential diagnosis, monitoring illness progression and the effect of psychiatric treatment.
Collapse
Affiliation(s)
- Kristína Krajčíková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Erika Semančíková
- 2(nd) Department of Psychiatry, L. Pasteur University Hospital, Rastislavova 43, Košice, 040 11, Slovakia; EPAMED s.r.o., Private Psychiatric Practice, Hlavná 68, Košice, 040 01, Slovakia.
| | - Katarína Zakutanská
- Institute of Experimental Physics, Department of Magnetism, Slovak Academy of Sciences, Watsonova 47, Košice, 040 01, Slovakia
| | - 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
| | - Jana Mašlanková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Marek Stupák
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Ivan Talian
- Department of Medical and Clinical Biophysics, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Natália Tomašovičová
- Institute of Experimental Physics, Department of Magnetism, Slovak Academy of Sciences, Watsonova 47, Košice, 040 01, Slovakia
| | - Tatiana Kimáková
- Department of Public Health and Hygiene, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Šrobárová 2, 041 80, Košice, 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
| | - Katarína Dubayová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Dagmar Breznoščáková
- 1(st) Department of Psychiatry, L. Pasteur University Hospital, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Eva Pálová
- EPAMED s.r.o., Private Psychiatric Practice, Hlavná 68, Košice, 040 01, Slovakia; 1(st) Department of Psychiatry, L. Pasteur University Hospital, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Juraj Semančík
- 4(th) Clinic of Internal Medicine, L. Pasteur University Hospital, Trieda SNP 1, Košice, 040 11, Slovakia
| | - Vladimíra Tomečková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, Košice, 040 11, Slovakia
| |
Collapse
|
68
|
Wang B, Shao Z, Gu M, Ni L, Shi Y, Yan Y, Wu A, Jin H, Chen J, Pan X, Xu D. Hydrogen sulfide protects against IL-1β-induced inflammation and mitochondrial dysfunction-related apoptosis in chondrocytes and ameliorates osteoarthritis. J Cell Physiol 2021; 236:4369-4386. [PMID: 33164235 DOI: 10.1002/jcp.30154] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/26/2020] [Accepted: 10/29/2020] [Indexed: 12/20/2022]
Abstract
The inflammatory environment and excessive chondrocyte apoptosis have been demonstrated to play crucial roles in the onset of osteoarthritis (OA). Hydrogen sulfide (H2 S), a gaseous signalling molecule, exerts an inhibitory effect on inflammation and apoptosis in several degenerative diseases. However, the protective effect of H2 S against OA has not been fully clarified, and its underlying mechanism should be examined further. In the current study, the role of endogenous H2 S in the pathogenesis of OA and its protective effects on interleukin (IL)-1β-induced chondrocytes were identified. Our data revealed decreased H2 S expression in both human degenerative OA cartilage tissue and IL-1β-induced chondrocytes. Pretreatment with the H2 S donor sodium hydrosulfide (NaHS) dramatically attenuated IL-1β-induced overproduction of inflammatory cytokines and improved the balance between anabolic and catabolic chondrocyte capacities, and these effects were dependent on PI3K/AKT pathway-mediated inhibition of nuclear factor kappa B (NF-κB). Moreover, mitochondrial dysfunction-related apoptosis was significantly reversed by NaHS in IL-1β-stimulated chondrocytes. Mechanistically, NaHS partially suppressed IL-1β-induced phosphorylation of the mitogen-activated protein kinase (MAPK) cascades. Furthermore, in the destabilization of the medial meniscus mouse model, OA progression was ameliorated by NaHS administration. Taken together, these results suggest that H2 S may antagonize IL-1β-induced inflammation and mitochondrial dysfunction-related apoptosis via selective suppression of the PI3K/Akt/NF-κB and MAPK signalling pathways, respectively, in chondrocytes and may be a potential therapeutic agent for the treatment of OA.
Collapse
Affiliation(s)
- Ben Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
- Department of Orthopedic, Taizhou Hospital Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhenxuan Shao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Mingbao Gu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Libin Ni
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yifeng Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Yingzhao Yan
- Department of Orthopaedic Surgery, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Aimin Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Haiming Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Jiaoxiang Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Xiaoyun Pan
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
| | - Daoliang Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, Zhejiang, China
| |
Collapse
|
69
|
Hybrid fluorescence-AFM explores articular surface degeneration in early osteoarthritis across length scales. Acta Biomater 2021; 126:315-325. [PMID: 33753314 DOI: 10.1016/j.actbio.2021.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/06/2021] [Accepted: 03/14/2021] [Indexed: 12/21/2022]
Abstract
Atomic force microscopy (AFM) has become a powerful tool for the characterization of materials at the nanoscale. Nevertheless, its application to hierarchical biological tissue like cartilage is still limited. One reason is that such samples are usually millimeters in size, while the AFM delivers much more localized information. Here a combination of AFM and fluorescence microscopy is presented where features on a millimeter sized tissue sample are selected by fluorescence microscopy on the micrometer scale and then mapped down to nanometer precision by AFM under native conditions. This served us to show that local changes in the organization of fluorescent stained cells, a marker for early osteoarthritis, correlate with a significant local reduction of the elastic modulus, local thinning of the collagen fibers, and a roughening of the articular surface. This approach is not only relevant for cartilage, but in general for the characterization of native biological tissue from the macro- to the nanoscale. STATEMENT OF SIGNIFICANCE: Different length scales have to be studied to understand the function and dysfunction of hierarchically organized biomaterials or tissues. Here we combine a highly stable AFM with fluorescence microscopy and precisely motorized movement to correlate micro- and nanoscopic properties of articular cartilage on a millimeter sized sample under native conditions. This is necessary for unraveling the relationship between microscale organization of chondrocytes, micrometer scale changes in articular cartilage properties and nanoscale organization of collagen (including D-banding). We anticipate that such studies pave the way for a guided design of hierarchical biomaterials.
Collapse
|
70
|
Ching KY, Andriotis O, Sengers B, Stolz M. Genipin crosslinked chitosan/PEO nanofibrous scaffolds exhibiting an improved microenvironment for the regeneration of articular cartilage. J Biomater Appl 2021; 36:503-516. [PMID: 33730922 PMCID: PMC8419291 DOI: 10.1177/08853282211002015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Towards optimizing the growth of extracellular matrix to produce repair cartilage for healing articular cartilage (AC) defects in joints, scaffold-based tissue engineering approaches have recently become a focus of clinical research. Scaffold-based approaches by electrospinning aim to support the differentiation of chondrocytes by providing an ultrastructure similar to the fibrillar meshwork in native cartilage. In a first step, we demonstrate how the blending of chitosan with poly(ethylene oxide) (PEO) allows concentrated chitosan solution to become electrospinnable. The chitosan-based scaffolds share the chemical structure and characteristics of glycosaminoglycans, which are important structural components of the cartilage extracellular matrix. Electrospinning produced nanofibrils of ∼100 nm thickness that are closely mimicking the size of collagen fibrils in human AC. The polymer scaffolds were stabilized in physiological conditions and their stiffness was tuned by introducing the biocompatible natural crosslinker genipin. We produced scaffolds that were crosslinked with 1.0% genipin to obtain values of stiffness that were in between the stiffness of the superficial zone human AC of 600 ± 150 kPa and deep zone AC of 1854 ± 483 kPa, whereas the stiffness of 1.5% genipin crosslinked scaffold was similar to the stiffness of deep zone AC. The scaffolds were degradable, which was indicated by changes in the fibril structure and a decrease in the scaffold stiffness after seven months. Histological and immunohistochemical analysis after three weeks of culture with human articular chondrocytes (HACs) showed a cell viability of over 90% on the scaffolds and new extracellular matrix deposited on the scaffolds.
Collapse
Affiliation(s)
- Kuan Yong Ching
- Foundation, Study and Language Institute, University of Reading-Malaysia Campus, Iskandar Puteri, Malaysia.,Bioengineering Science, Faculty of Engineering, University of Southampton, Southampton, UK
| | - Orestis Andriotis
- Faculty of Engineering, National Centre for Advanced Tribology at Southampton, University of Southampton, Southampton, UK
| | - Bram Sengers
- Institute of Lightweight Design and Structural Biomechanics, Vienna University of Technology, Vienna, Austria
| | - Martin Stolz
- Bioengineering Science, Faculty of Engineering, University of Southampton, Southampton, UK
| |
Collapse
|
71
|
Zhan X, Teng W, Sun K, He J, Yang J, Tian J, Huang X, Zhou L, Zhou C. CD47-mediated DTIC-loaded chitosan oligosaccharide-grafted nGO for synergistic chemo-photothermal therapy against malignant melanoma. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:112014. [PMID: 33812633 DOI: 10.1016/j.msec.2021.112014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/11/2021] [Accepted: 02/27/2021] [Indexed: 12/11/2022]
Abstract
Nano-graphene oxide (nGO), an effective drug nanocarrier, is used for simultaneous photothermal therapy (PTT) and near-infrared fluorescence imaging. Dacarbazine (DTIC) is used in the treatment of melanoma with limited clinical efficacy. PTT shows promise in the treatment of skin cancer. Herein, chitosan oligosaccharide (COS)-grafted nGO was further modified with CD47 antibody, and loaded DTIC was prepared using a versatile nanoplatform (nGO-COS-CD47/DTIC) for the treatment of melanoma as a synergistic targeted chemo-photothermal therapy. The in vitro results demonstrated that nGO-COS-CD47/DTIC nanocarriers have excellent biocompatibility, photothermal conversion efficiency, high targeting efficiency, fast drug release under NIR irradiation, and tumor cell killing efficiency. Notably, nGO-COS-CD47/DTIC plus NIR irradiation significantly promoted early cell apoptosis through the mitochondrial apoptosis pathway and exhibited a significant joint function of antitumor efficacy. The demonstrated nGO-COS-CD47/DTIC can provide a highly efficient malignant melanoma therapy using this multifunctional intelligent nanoplatform.
Collapse
Affiliation(s)
- Xiaozhen Zhan
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, PR China.
| | - Wanqing Teng
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi, Shandong 276005, PR China
| | - Kai Sun
- College of Chemistry and Material Science, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Jiexiang He
- College of Chemistry and Material Science, Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Jie Yang
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, PR China
| | - Jinhuan Tian
- College of Chemistry and Material Science, Jinan University, Guangzhou, Guangdong 510632, PR China.
| | - Xun Huang
- Collaborative Innovation Center of Tumor Marker Detection Technology, Equipment and Diagnosis-Therapy Integration in Universities of Shandong, Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi, Shandong 276005, PR China.
| | - Lin Zhou
- The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, PR China.
| | - Changren Zhou
- College of Chemistry and Material Science, Jinan University, Guangzhou, Guangdong 510632, PR China.
| |
Collapse
|
72
|
Tschaikowsky M, Selig M, Brander S, Balzer BN, Hugel T, Rolauffs B. Proof-of-concept for the detection of early osteoarthritis pathology by clinically applicable endomicroscopy and quantitative AI-supported optical biopsy. Osteoarthritis Cartilage 2021; 29:269-279. [PMID: 33220445 DOI: 10.1016/j.joca.2020.10.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/30/2020] [Accepted: 10/14/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Clinical trials for osteoarthritis (OA), the leading cause of global disability, are unable to pinpoint the early, potentially reversible disease with clinical technology. Hence, disease-modifying drug candidates cannot be tested early in the disease. To overcome this obstacle, we asked whether early OA-pathology detection is possible with current clinical technology. METHODS We determined the relationship between two sensitive early OA markers, atomic force microscopy (AFM)-measured human articular cartilage (AC) surface stiffness, and location-matched superficial zone chondrocyte spatial organizations (SCSOs), asking whether a significant loss of surface stiffness can be detected in early OA SCSO stages. We then tested whether current clinical technology can visualize and accurately diagnose the SCSOs using an approved probe-based confocal laser-endomicroscope and a random forest (RF) model. RESULTS We demonstrated a correlation between AC surface stiffness and the SCSO (rrm = -0.91; 95%CI: -0.97, -0.73), and an extensive loss of surface stiffness specifically in those ACs with early OA-typical SCSO (95%CIs: string SCSO: 269-173 kPa, double string SCSO: 77-46 kPa). This established the SCSO as a visualizable, functionally relevant surrogate marker of early OA AC surface pathology. Moreover, SCSO-based stiffness discrimination worked well in each patient's AC. We then demonstrated feasibility of visualizing the SCSO by clinical laser-endomicroscopy and, importantly, accurate SCSO diagnosis using RF. CONCLUSION We present the proof-of-concept of early OA-pathology detection with available clinical technology, introducing a future-oriented, AI-supported, non-destructive quantitative optical biopsy for early disease detection. Operationalizing SCSO recognition, this approach allows testing for correlations between local tissue architectures with other experimental and clinical read-outs, but needs clinical validation and a larger sample size for defining diagnostic thresholds.
Collapse
Affiliation(s)
- M Tschaikowsky
- Institute of Physical Chemistry, Albert-Ludwigs-University Freiburg, Albertstr. 21, 79104, Freiburg, Germany; G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Germany
| | - M Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Germany
| | - S Brander
- Institute of Physical Chemistry, Albert-Ludwigs-University Freiburg, Albertstr. 21, 79104, Freiburg, Germany
| | - B N Balzer
- Institute of Physical Chemistry, Albert-Ludwigs-University Freiburg, Albertstr. 21, 79104, Freiburg, Germany; Cluster of Excellence LivMatS @ FIT - Freiburg Center for Interactive Materials and Bioinspired Technologies, Freiburg, Germany
| | - T Hugel
- Institute of Physical Chemistry, Albert-Ludwigs-University Freiburg, Albertstr. 21, 79104, Freiburg, Germany; Cluster of Excellence LivMatS @ FIT - Freiburg Center for Interactive Materials and Bioinspired Technologies, Freiburg, Germany.
| | - B Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center - Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Germany.
| |
Collapse
|
73
|
Raspanti M, Protasoni M, Zecca PA, Reguzzoni M. Slippery when wet: The free surface of the articular cartilage. Microsc Res Tech 2020; 84:1257-1264. [PMID: 33378558 DOI: 10.1002/jemt.23684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/24/2020] [Accepted: 12/15/2020] [Indexed: 12/26/2022]
Abstract
The free surface of the articular cartilage must withstand compressive and shearing forces, maintain a low friction coefficient and allow oxygen and metabolites to reach the underlying matrix. In many ways it is critical to the physiology of the whole tissue and its disruption always involves deep pathological alterations and loss of the joint integrity. Being very difficult to image with section-based conventional techniques, it was often described by previous research in conflicting terms or entirely overlooked. High-magnification face-on observations with high resolution scanning electron microscopy and with scanning probe microscopy revealed a very thin, delicate superficial layer rich in glycoconjugates, which may explain the very low friction coefficient of the tissue but which was very easily altered and/or dissolved in the preparation. Beneath this superficial sheet lies a thicker coat of thin, highly uniform, slightly wavy collagen fibrils lying parallel to the surface and mutually interconnected by a huge number of interfibrillar glycosaminoglycan bridges. These bridges and the collagen fibrils form an extended reticular structure able to redistribute tensile and compressive stress across a larger area of the surface and hence a greater volume of tissue.
Collapse
Affiliation(s)
- Mario Raspanti
- Laboratory of Human Morphology, Department of Medicine & Surgery, Insubria University, Varese, Italy
| | - Marina Protasoni
- Laboratory of Human Morphology, Department of Medicine & Surgery, Insubria University, Varese, Italy
| | - Piero Antonio Zecca
- Laboratory of Human Morphology, Department of Medicine & Surgery, Insubria University, Varese, Italy
| | - Marcella Reguzzoni
- Laboratory of Human Morphology, Department of Medicine & Surgery, Insubria University, Varese, Italy
| |
Collapse
|
74
|
Chery DR, Han B, Zhou Y, Wang C, Adams SM, Chandrasekaran P, Kwok B, Heo SJ, Enomoto-Iwamoto M, Lu XL, Kong D, Iozzo RV, Birk DE, Mauck RL, Han L. Decorin regulates cartilage pericellular matrix micromechanobiology. Matrix Biol 2020; 96:1-17. [PMID: 33246102 DOI: 10.1016/j.matbio.2020.11.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
Abstract
In cartilage tissue engineering, one key challenge is for regenerative tissue to recapitulate the biomechanical functions of native cartilage while maintaining normal mechanosensitive activities of chondrocytes. Thus, it is imperative to discern the micromechanobiological functions of the pericellular matrix, the ~ 2-4 µm-thick domain that is in immediate contact with chondrocytes. In this study, we discovered that decorin, a small leucine-rich proteoglycan, is a key determinant of cartilage pericellular matrix micromechanics and chondrocyte mechanotransduction in vivo. The pericellular matrix of decorin-null murine cartilage developed reduced content of aggrecan, the major chondroitin sulfate proteoglycan of cartilage and a mild increase in collagen II fibril diameter vis-à-vis wild-type controls. As a result, decorin-null pericellular matrix showed a significant reduction in micromodulus, which became progressively more pronounced with maturation. In alignment with the defects of pericellular matrix, decorin-null chondrocytes exhibited decreased intracellular calcium activities, [Ca2+]i, in both physiologic and osmotically evoked fluidic environments in situ, illustrating impaired chondrocyte mechanotransduction. Next, we compared [Ca2+]i activities of wild-type and decorin-null chondrocytes following enzymatic removal of chondroitin sulfate glycosaminoglycans. The results showed that decorin mediates chondrocyte mechanotransduction primarily through regulating the integrity of aggrecan network, and thus, aggrecan-endowed negative charge microenvironment in the pericellular matrix. Collectively, our results provide robust genetic and biomechanical evidence that decorin is an essential constituent of the native cartilage matrix, and suggest that modulating decorin activities could improve cartilage regeneration.
Collapse
Affiliation(s)
- Daphney R Chery
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Biao Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Ying Zhou
- Department of Statistical Sciences, University of Toronto, Toronto, ON M5S 3G3, Canada
| | - Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Bryan Kwok
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201, United States
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - Dehan Kong
- Department of Statistical Sciences, University of Toronto, Toronto, ON M5S 3G3, Canada
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
75
|
Xiao S, Chen L. The emerging landscape of nanotheranostic-based diagnosis and therapy for osteoarthritis. J Control Release 2020; 328:817-833. [PMID: 33176171 DOI: 10.1016/j.jconrel.2020.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a common degenerative disease involving numerous joint tissues and cells, with a growing rate in prevalence that ultimately results in a negative social impact. Early diagnosis, OA progression monitoring and effective treatment are of significant importance in halting OA process. However, traditional imaging techniques lack sensitivity and specificity, which lead to a delay in timely clinical intervention. Additionally, current treatments only slow the progression of OA but have not meet the largely medical need for disease-modifying therapy. In order to overcome the above-mentioned problems and improve clinical efficacy, nanotheranostics has been proposed on OA remedy, which has confirmed success in animal models. In this review, different imaging targets-based nanoprobe for early and timely OA diagnosis is first discussed. Second, therapeutic strategies delivered by nanosystem are summarized as much as possible. Their advantages and the potential for clinical translation are detailed discussed. Third, nanomedicine simultaneously combined with the imaging for OA treatment is introduced. Nanotheranostics dynamically tracked the OA treatment outcomes to timely and individually adjust therapy. Finally, future prospects and challenges of nanotechnology-based OA diagnosis, imaging and treatment are concluded and predicted. It is believed that nanoprobe and nanomedicine will become prospective in OA therapeutic revolution.
Collapse
Affiliation(s)
- Shuyi Xiao
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Liang Chen
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China.
| |
Collapse
|
76
|
Dai S, Liang T, Fujii T, He S, Zhang F, Jiang H, Liu B, Shi X, Luo Z, Yang H. Increased elastic modulus of the synovial membrane in a rat ACLT model of osteoarthritis revealed by atomic force microscopy. Braz J Med Biol Res 2020; 53:e10058. [PMID: 33053109 PMCID: PMC7552902 DOI: 10.1590/1414-431x202010058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/22/2020] [Indexed: 11/21/2022] Open
Abstract
This study aimed to explore changes in nanoscale elastic modulus of the synovium using atomic force microscopy (AFM) in addition to investigate changes in synovial histomorphology and secretory function in osteoarthritis (OA) in a rat anterior cruciate ligament transection (ACLT) model. Sprague-Dawley rats were randomly assigned to sham control and ACLT OA groups. All right knee joints were harvested at 4, 8, or 12 weeks (W) after surgery for histological assessment of cartilage damage and synovitis in both the anterior and posterior capsules. AFM imaging and nanoscale biomechanical testing were conducted to measure the elastic modulus of the synovial collagen fibrils. Immunohistochemistry was used to visualize the expression of interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and matrix metalloproteinase-3 (MMP-3) in the synovium. The OA groups exhibited progressive development of disease in the cartilage and synovium. Histopathological scores of the synovium in the OA groups increased gradually. Significant differences were observed between all OA groups except for the posterior 4W group. The synovial fibril arrangement in all OA groups was significantly disordered. The synovial fibrils in all ACLT OA groups at each time point were stiffer than those in the sham controls. OA rats displayed a significantly higher expression of IL-1β and MMP3 in the anterior capsule. In summary, synovial stiffening was closely associated with joint degeneration and might be a factor contributing to synovitis and increased production of proinflammatory mediators. Our data provided insights into the role of synovitis, particularly stiffening of the synovium, in OA pathogenesis.
Collapse
Affiliation(s)
- Shouqian Dai
- Orthopedic Institute, Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.,Department of Emergency Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Ting Liang
- Orthopedic Institute, Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tadashi Fujii
- Department of Orthopaedic Surgery, Kashiba Asahigaoka Hospital, Kashiba, Nara, Japan
| | - Shuangjun He
- Orthopedic Institute, Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fan Zhang
- Orthopedic Institute, Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Huaye Jiang
- Orthopedic Institute, Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bo Liu
- Orthopedic Institute, Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiu Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zongping Luo
- Orthopedic Institute, Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Huilin Yang
- Orthopedic Institute, Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
77
|
Hartmann B, Marchi G, Alberton P, Farkas Z, Aszodi A, Roths J, Clausen-Schaumann H. Early Detection of Cartilage Degeneration: A Comparison of Histology, Fiber Bragg Grating-Based Micro-Indentation, and Atomic Force Microscopy-Based Nano-Indentation. Int J Mol Sci 2020; 21:ijms21197384. [PMID: 33036285 PMCID: PMC7582717 DOI: 10.3390/ijms21197384] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/29/2020] [Accepted: 10/04/2020] [Indexed: 12/27/2022] Open
Abstract
We have determined the sensitivity and detection limit of a new fiber Bragg grating (FBG)-based optoelectronic micro-indenter for biomechanical testing of cartilage and compared the results to indentation-type atomic force microscopy (IT-AFM) and histological staining. As test samples, we used bovine articular cartilage, which was enzymatically degraded ex vivo for five minutes using different concentrations of collagenase (5, 50, 100 and 500 µg/mL) to mimic moderate extracellular matrix deterioration seen in early-stage osteoarthritis (OA). Picrosirius Red staining and polarization microscopy demonstrated gradual, concentration-dependent disorganization of the collagen fibrillar network in the superficial zone of the explants. Osteoarthritis Research Society International (OARSI) grading of histopathological changes did not discriminate between undigested and enzymatically degraded explants. IT-AFM was the most sensitive method for detecting minute changes in cartilage biomechanics induced by the lowest collagenase concentration, however, it did not distinguish different levels of cartilage degeneration for collagenase concentrations higher than 5 µg/mL. The FBG micro-indenter provided a better and more precise assessment of the level of cartilage degeneration than the OARSI histological grading system but it was less sensitive at detecting mechanical changes than IT-AFM. The FBG-sensor allowed us to observe differences in cartilage biomechanics for collagenase concentrations of 100 and 500 µg/mL. Our results confirm that the FBG sensor is capable of detecting small changes in articular cartilage stiffness, which may be associated with initial cartilage degeneration caused by early OA.
Collapse
Affiliation(s)
- Bastian Hartmann
- Center for Applied Tissue Engineering and Regenerative Medicine–CANTER, Munich University of Applied Sciences, 80335 Munich, Germany; (B.H.); (A.A.)
- Laboratory of Experimental Surgery and Regenerative Medicine (ExperiMed), Clinic for General, Trauma and Reconstructive Surgery, University of Munich, 82152 Planegg, Germany; (P.A.); (Z.F.)
- Center for Nanoscience (CeNS), University of Munich, 80331 Munich, Germany
| | - Gabriele Marchi
- Photonics Laboratory, Department of Applied Sciences and Mechatronics, Munich University of Applied Sciences, 80335 Munich, Germany; (G.M.); (J.R.)
| | - Paolo Alberton
- Laboratory of Experimental Surgery and Regenerative Medicine (ExperiMed), Clinic for General, Trauma and Reconstructive Surgery, University of Munich, 82152 Planegg, Germany; (P.A.); (Z.F.)
| | - Zsuzsanna Farkas
- Laboratory of Experimental Surgery and Regenerative Medicine (ExperiMed), Clinic for General, Trauma and Reconstructive Surgery, University of Munich, 82152 Planegg, Germany; (P.A.); (Z.F.)
| | - Attila Aszodi
- Center for Applied Tissue Engineering and Regenerative Medicine–CANTER, Munich University of Applied Sciences, 80335 Munich, Germany; (B.H.); (A.A.)
- Laboratory of Experimental Surgery and Regenerative Medicine (ExperiMed), Clinic for General, Trauma and Reconstructive Surgery, University of Munich, 82152 Planegg, Germany; (P.A.); (Z.F.)
| | - Johannes Roths
- Photonics Laboratory, Department of Applied Sciences and Mechatronics, Munich University of Applied Sciences, 80335 Munich, Germany; (G.M.); (J.R.)
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine–CANTER, Munich University of Applied Sciences, 80335 Munich, Germany; (B.H.); (A.A.)
- Center for Nanoscience (CeNS), University of Munich, 80331 Munich, Germany
- Correspondence: ; Tel.: +49-89-1265-1682
| |
Collapse
|
78
|
Spatial mapping of the collagen distribution in human and mouse tissues by force volume atomic force microscopy. Sci Rep 2020; 10:15664. [PMID: 32973235 PMCID: PMC7518416 DOI: 10.1038/s41598-020-72564-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/20/2020] [Indexed: 01/06/2023] Open
Abstract
Changes in the elastic properties of living tissues during normal development and in pathological processes are often due to modifications of the collagen component of the extracellular matrix at various length scales. Force volume AFM can precisely capture the mechanical properties of biological samples with force sensitivity and spatial resolution. The integration of AFM data with data of the molecular composition contributes to understanding the interplay between tissue biochemistry, organization and function. The detection of micrometer-size, heterogeneous domains at different elastic moduli in tissue sections by AFM has remained elusive so far, due to the lack of correlations with histological, optical and biochemical assessments. In this work, force volume AFM is used to identify collagen-enriched domains, naturally present in human and mouse tissues, by their elastic modulus. Collagen identification is obtained in a robust way and affordable timescales, through an optimal design of the sample preparation method and AFM parameters for faster scan with micrometer resolution. The choice of a separate reference sample stained for collagen allows correlating elastic modulus with collagen amount and position with high statistical significance. The proposed preparation method ensures safe handling of the tissue sections guarantees the preservation of their micromechanical characteristics over time and makes it much easier to perform correlation experiments with different biomarkers independently.
Collapse
|
79
|
Selig M, Lauer JC, Hart ML, Rolauffs B. Mechanotransduction and Stiffness-Sensing: Mechanisms and Opportunities to Control Multiple Molecular Aspects of Cell Phenotype as a Design Cornerstone of Cell-Instructive Biomaterials for Articular Cartilage Repair. Int J Mol Sci 2020; 21:E5399. [PMID: 32751354 PMCID: PMC7432012 DOI: 10.3390/ijms21155399] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/23/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since material stiffness controls many cell functions, we reviewed the currently available knowledge on stiffness sensing and elucidated what is known in the context of clinical and experimental articular cartilage (AC) repair. Remarkably, no stiffness information on the various biomaterials for clinical AC repair was accessible. Using mRNA expression profiles and morphology as surrogate markers of stiffness-related effects, we deduced that the various clinically available biomaterials control chondrocyte (CH) phenotype well, but not to equal extents, and only in non-degenerative settings. Ample evidence demonstrates that multiple molecular aspects of CH and mesenchymal stromal cell (MSC) phenotype are susceptible to material stiffness, because proliferation, migration, lineage determination, shape, cytoskeletal properties, expression profiles, cell surface receptor composition, integrin subunit expression, and nuclear shape and composition of CHs and/or MSCs are stiffness-regulated. Moreover, material stiffness modulates MSC immuno-modulatory and angiogenic properties, transforming growth factor beta 1 (TGF-β1)-induced lineage determination, and CH re-differentiation/de-differentiation, collagen type II fragment production, and TGF-β1- and interleukin 1 beta (IL-1β)-induced changes in cell stiffness and traction force. We then integrated the available molecular signaling data into a stiffness-regulated CH phenotype model. Overall, we recommend using material stiffness for controlling cell phenotype, as this would be a promising design cornerstone for novel future-oriented, cell-instructive biomaterials for clinical high-quality AC repair tissue.
Collapse
Affiliation(s)
- Mischa Selig
- 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.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Jasmin C. Lauer
- 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.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - 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.S.); (J.C.L.); (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.S.); (J.C.L.); (M.L.H.)
| |
Collapse
|
80
|
Wang S, Su S, Yu C, Gopinath SCB, Yang Z. Immunodetection of urinary C-terminal telopeptide fragment of type II collagen: An osteoarthritis biomarker analysis. Biotechnol Appl Biochem 2020; 68:726-731. [PMID: 32621620 DOI: 10.1002/bab.1981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The urinary C-terminal telopeptide fragment of type II collagen (uCTX-II) has been reported as the efficient blood-based biomarker for osteoarthritis, which affects knees, hands, spine, and hips. This study reports a sensing strategy with antibody-conjugated gold nanoparticles (GNP) on an interdigitated electrode (IDE) to determine uCTX-II. The GNP-antibody complex was chemically immobilized on the IDE surface through the amine linker. uCTX-II was determined by monitoring the alteration in current upon interacting the GNP-complexed antibody. This strategy was improved the detection by attracting higher uCTX-II molecules, and the detection limit falls in the range of 10-100 pM with an acceptable regression value [y = 0.6254x - 0.4073, R² = 0.9787]. The sensitivity of the detection was recognized at 10 pM. Additionally, upon increasing the uCTX-II concentration, the current changes were increased in a linear fashion. Control detection with nonimmune antibody and control protein do not increase the current level, confirming the specific detection of uCTX-II. This method of detection helps in diagnosing osteoarthritis and its follow-up treatment.
Collapse
Affiliation(s)
- Shuwei Wang
- Department of Orthopedics, Shouguang Hospital of TCM, Weifang, Shandong Province, 262700, China
| | - Shanlin Su
- Department of Orthopedics, Shouguang Hospital of TCM, Weifang, Shandong Province, 262700, China
| | - Chunyun Yu
- Department of Orthopedics, Shouguang Hospital of TCM, Weifang, Shandong Province, 262700, China
| | - Subash C B Gopinath
- School of Bioprocess Engineering, Universiti Malaysia Perlis, Arau, Perlis, 02600, Malaysia.,Institute of Nano Electronic Engineering, Universiti Malaysia Perlis, Kangar, Perlis, 01000, Malaysia
| | - Zhiquan Yang
- Department of Orthopedics, Shouguang Hospital of TCM, Weifang, Shandong Province, 262700, China
| |
Collapse
|
81
|
Semitela Â, Girão AF, Fernandes C, Ramalho G, Bdikin I, Completo A, Marques PA. Electrospinning of bioactive polycaprolactone-gelatin nanofibres with increased pore size for cartilage tissue engineering applications. J Biomater Appl 2020; 35:471-484. [PMID: 32635814 DOI: 10.1177/0885328220940194] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Polycaprolactone (PCL) electrospun scaffolds have been widely investigated for cartilage repair application. However, their hydrophobicity and small pore size has been known to prevent cell attachment, proliferation and migration. Here, PCL was blended with gelatin (GEL) combining the favorable biological properties of GEL with the good mechanical performance of the former. Also, polyethylene glycol (PEG) particles were introduced during the electrospinning of the polymers blend by simultaneous electrospraying. These particles were subsequently removed resulting in fibrous scaffolds with enlarged pore size. PCL, GEL and PEG scaffolds formulations were developed and extensively structural and biologically characterized. GEL incorporation on the PCL scaffolds led to a considerably improved cell attachment and proliferation. A substantial pore size and interconnectivity increase was obtained, allowing cell infiltration through the porogenic scaffolds. All together these results suggest that this combined approach may provide a potentially clinically viable strategy for cartilage regeneration.
Collapse
Affiliation(s)
- Ângela Semitela
- TEMA, Department of Mechanical Engineering, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - André F Girão
- TEMA, Department of Mechanical Engineering, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Carla Fernandes
- TEMA, Department of Mechanical Engineering, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Gonçalo Ramalho
- TEMA, Department of Mechanical Engineering, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Igor Bdikin
- TEMA, Department of Mechanical Engineering, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - António Completo
- TEMA, Department of Mechanical Engineering, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Paula Aap Marques
- TEMA, Department of Mechanical Engineering, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| |
Collapse
|
82
|
Lee D, Hong KT, Lim TS, Lee E, Lee YH, Park JS, Kim W, Oh JH, Choi JA, Song Y. Alterations in articular cartilage T2 star relaxation time following mechanical disorders: in vivo canine supraspinatus tendon resection models. BMC Musculoskelet Disord 2020; 21:424. [PMID: 32615950 PMCID: PMC7331159 DOI: 10.1186/s12891-020-03447-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/23/2020] [Indexed: 11/10/2022] Open
Abstract
Background The role of altered joint mechanics on cartilage degeneration in in vivo models has not been studied successfully due to a lack of pre-injury information. We aimed 1) to develop an accurate in vivo canine model to measure the changes in joint loading and T2 star (T2*) relaxation time before and after unilateral supraspinatus tendon resections, and 2) to find the relationship between regional variations in articular cartilage loading patterns and T2* relaxation time distributions. Methods Rigid markers were implanted in the scapula and humerus of tested dogs. The movement of the shoulder bones were measured by a motion tracking system during normal gaits. In vivo cartilage contact strain was measured by aligning 3D shoulder models with the motion tracking data. Articular cartilage T2* relaxation times were measured by quantitative MRI scans. Articular cartilage contact strain and T2* relaxation time were compared in the shoulders before and 3 months after the supraspinatus tendon resections. Results Excellent accuracy and reproducibility were found in our in vivo contact strain measurements with less than 1% errors. Changes in articular cartilage contact strain exhibited similar patterns with the changes in the T2* relaxation time after resection surgeries. Regional changes in the articular cartilage T2* relaxation time exhibited positive correlations with regional contact strain variations 3 months after the supraspinatus resection surgeries. Conclusion This is the first study to measure in vivo articular cartilage contact strains with high accuracy and reproducibility. Positive correlations between contact strain and T2* relaxation time suggest that the articular cartilage extracellular matrix may responds to mechanical changes in local areas.
Collapse
Affiliation(s)
- Dokwan Lee
- Department of Mechanical Engineering, Korea University Engineering Campus, Innovation Hall, Room 306, Anam-dong, Seongbuk-gu, Seoul, 02841, South Korea
| | - Ki-Taek Hong
- Department of Mechanical Engineering, Korea University Engineering Campus, Innovation Hall, Room 306, Anam-dong, Seongbuk-gu, Seoul, 02841, South Korea
| | - Tae Seong Lim
- Department of Radiology, Gachon University Gil Medical Center, Incheon, South Korea
| | - Eugene Lee
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Ye Hyun Lee
- Department of Orthopedic Surgery, National Police Hospital, Seoul, South Korea
| | - Ji Soon Park
- Department of Orthopedic Surgery, Sheikh Khalifa Specialty Hospital, Ras Al Khaimah, United Arab Emirates
| | - Woo Kim
- Seoul Kiwoonchan Orthopedics Clinic, Seoul, South Korea
| | - Joo Han Oh
- Department of Orthopedic Surgery, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jung-Ah Choi
- Department of Radiology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, South Korea
| | - Yongnam Song
- Department of Mechanical Engineering, Korea University Engineering Campus, Innovation Hall, Room 306, Anam-dong, Seongbuk-gu, Seoul, 02841, South Korea.
| |
Collapse
|
83
|
Collagen IX deficiency leads to premature vascularization and ossification of murine femoral heads through an imbalance of pro- and antiangiogenic factors. Osteoarthritis Cartilage 2020; 28:988-999. [PMID: 32283184 DOI: 10.1016/j.joca.2020.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/20/2020] [Accepted: 03/30/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The vascular invasion of cartilage is an essential process in the endochondral ossification of long bones. In contrast, vascularization of articular cartilage constitutes a pathological mechanism in the development of osteoarthritis. Polymorphisms of Col9a1 have been described as risk factors for hip osteoarthritis (OA) and the loss of collagen IX is known to lead to premature OA of the hip joint in mice but the underlying mechanism is so far unknown. DESIGN To understand the contribution of collagen IX to OA development in the hip joint, we analyzed the early development of murine Col9a1-/- femoral heads between newborn stage and 16 weeks of age. RESULTS We found significantly accelerated ossification of the femoral heads in the absence of collagen IX as well as premature vascular and osteoclast invasion, even though hypertrophic differentiation was delayed. The loss of collagen IX led to anatomically altered femoral heads lacking the epiphyseal tubercle. Interestingly, this region was found to contain highest levels of the antiangiogenic protein thrombospondin-1 (TSP-1). Hence, TSP-1 levels were strongly reduced in the Col9a1-/- femoral heads. In addition, antiangiogenic matrilin-1 was found to be decreased, while proangiogenic active MMP-9 levels were increased in the collagen IX deficient mice compared to wildtype controls. CONCLUSION We conclude that collagen IX protects against premature vascularization and cartilage to bone transition in femoral heads by increasing the levels of antiangiogenic TSP-1 and matrilin-1 and decreasing the levels of proangiogenic active MMP-9.
Collapse
|
84
|
Magerle R, Dehnert M, Voigt D, Bernstein A. Nanomechanical 3D Depth Profiling of Collagen Fibrils in Native Tendon. Anal Chem 2020; 92:8741-8749. [PMID: 32484331 DOI: 10.1021/acs.analchem.9b05582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Connective tissue displays a large compositional and structural complexity that involves multiple length scales. In particular, on the molecular and the nanometer level, the elementary processes that determine the biomechanics of collagen fibrils in connective tissues are still poorly understood. Here, we use atomic force microscopy (AFM) to determine the three-dimensional (3D) depth profiles of the local nanomechanical properties of collagen fibrils and their embedding interfibrillar matrix in native (unfixed), hydrated Achilles tendon of sheep and chickens. AFM imaging in air with controlled humidity preserves the tissue's water content, allowing the assembly of collagen fibrils to be imaged in high resolution beneath an approximately 5-10 nm thick layer of the fluid components of the interfibrillar matrix. We collect pointwise force-distance (FD) data and amplitude-phase-distance (APD) data, from which we construct 3D depth profiles of the local tip-sample interaction forces. The 3D images reveal the nanomechanical morphology of unfixed, hydrated collagen fibrils in native tendon with a 0.1 nm depth resolution and a 10 nm lateral resolution. We observe a diversity in the nanomechanical properties among individual collagen fibrils in their adhesive and in their repulsive, viscoelastic mechanical response as well as among the contact points between adjacent collagen fibrils. This sheds new light on the role of interfibrillar bonds and the mechanical properties of the interfibrillar matrix in the biomechanics of tendon.
Collapse
Affiliation(s)
- Robert Magerle
- Fakultät für Naturwissenschaften, Technische Universität Chemnitz, 09107 Chemnitz, Germany
| | - Martin Dehnert
- Fakultät für Naturwissenschaften, Technische Universität Chemnitz, 09107 Chemnitz, Germany
| | - Diana Voigt
- Fakultät für Naturwissenschaften, Technische Universität Chemnitz, 09107 Chemnitz, Germany
| | - Anke Bernstein
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg and Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
| |
Collapse
|
85
|
Dusfour G, Maumus M, Cañadas P, Ambard D, Jorgensen C, Noël D, Le Floc'h S. Mesenchymal stem cells-derived cartilage micropellets: A relevant in vitro model for biomechanical and mechanobiological studies of cartilage growth. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 112:110808. [PMID: 32409025 DOI: 10.1016/j.msec.2020.110808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 11/20/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
The prevalence of diseases that affect the articular cartilage is increasing due to population ageing, but the current treatments are only palliative. One innovative approach to repair cartilage defects is tissue engineering and the use of mesenchymal stem/stromal cells (MSCs). Although the combination of MSCs with biocompatible scaffolds has been extensively investigated, no product is commercially available yet. This could be explained by the lack of mechanical stimulation during in vitro culture and the absence of proper and stable cartilage matrix formation, leading to poor integration after implantation. The objective of the present study was to investigate the biomechanical behaviour of MSC differentiation in micropellets, a well-defined 3D in vitro model of cartilage differentiation and growth, in view of tissue engineering applications. MSC micropellet chondrogenic differentiation was induced by exposure to TGFβ3. At different time points during differentiation (35 days of culture), their global mechanical properties were assessed using a very sensitive compression device coupled to an identification procedure based on a finite element parametric model. Micropellets displayed both a non-linear strain-induced stiffening behaviour and a dissipative behaviour that increased from day 14 to day 29, with a maximum instantaneous Young's modulus of 179.9 ± 18.8 kPa. Moreover, chondrocyte gene expression levels were strongly correlated with the observed mechanical properties. This study indicates that cartilage micropellets display the biochemical and biomechanical characteristics required for investigating and recapitulating the different stages of cartilage development.
Collapse
Affiliation(s)
- G Dusfour
- LMGC, Univ. Montpellier, CNRS, Montpellier, France
| | - M Maumus
- IRMB, Univ. Montpellier, INSERM, CHU Montpellier, Montpellier, France; Hopital Lapeyronie, Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Montpellier, France
| | - P Cañadas
- LMGC, Univ. Montpellier, CNRS, Montpellier, France
| | - D Ambard
- LMGC, Univ. Montpellier, CNRS, Montpellier, France
| | - C Jorgensen
- IRMB, Univ. Montpellier, INSERM, CHU Montpellier, Montpellier, France; Hopital Lapeyronie, Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Montpellier, France
| | - D Noël
- IRMB, Univ. Montpellier, INSERM, CHU Montpellier, Montpellier, France; Hopital Lapeyronie, Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Montpellier, France
| | - S Le Floc'h
- LMGC, Univ. Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
86
|
Ihnatouski M, Pauk J, Karev D, Karev B. AFM-Based Method for Measurement of Normal and Osteoarthritic Human Articular Cartilage Surface Roughness. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E2302. [PMID: 32429426 PMCID: PMC7288191 DOI: 10.3390/ma13102302] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/09/2020] [Accepted: 05/13/2020] [Indexed: 12/23/2022]
Abstract
In osteoarthrosis, pathological features of articular cartilage are associated with degeneration and nanomechanical changes. The aim of this paper is to show that indentation-atomic force microscopy can monitor wear-related biomechanical changes in the hip joint of patients with osteoarthritis. Fifty patients (N = 50), aged 40 to 65, were included in the study. The mechanical properties and the submicron surface morphology of hyaline cartilage were investigated using atomic force microscopy. Measurements of the roughness parameters of cartilage surfaces were performed, including the arithmetic average of absolute values (Ra), the maximum peak height (Rp), and the mean spacing between local peaks (S). The arithmetic mean of the absolute values of the height of healthy cartilage was 86 nm, while wear began at Ra = 73 nm. The maximum changes of values of the roughness parameters differed from the healthy ones by 71%, 80%, and 51% for Ra, Rp, and S, respectively. Young's modulus for healthy cartilage surfaces ranged from 1.7 to 0.5 MPa. For the three stages of cartilage wear, Young's modulus increased, and then it approached the maximum value and decreased. AFM seems to be a powerful tool for surface analysis of biological samples as it enables indentation measurements in addition to imaging.
Collapse
Affiliation(s)
- Mikhail Ihnatouski
- Scientific and Research Department, Yanka Kupala State University of Grodno, Grodno, Ozheshko str., 22, 230023 Grodno, Belarus;
| | - Jolanta Pauk
- Biomedical Engineering Institute, Bialystok University of Technology, Wiejska 45A, 15-351 Bialystok, Poland
| | - Dmitrij Karev
- Department of Traumatology, Orthopedics and Field Surgery, Grodno State Medical University, Gorkogo str. 80, 230009 Grodno, Belarus;
| | - Boris Karev
- Department of Orthopedic and Traumatology, Grodno City Emergency Hospital, Sovietskih Pogranichnikov str., 115, 230027 Grodno, Belarus;
| |
Collapse
|
87
|
Bachmann B, Spitz S, Schädl B, Teuschl AH, Redl H, Nürnberger S, Ertl P. Stiffness Matters: Fine-Tuned Hydrogel Elasticity Alters Chondrogenic Redifferentiation. Front Bioeng Biotechnol 2020; 8:373. [PMID: 32426347 PMCID: PMC7204401 DOI: 10.3389/fbioe.2020.00373] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/03/2020] [Indexed: 12/20/2022] Open
Abstract
Biomechanical cues such as shear stress, stretching, compression, and matrix elasticity are vital in the establishment of next generation physiological in vitro tissue models. Matrix elasticity, for instance, is known to guide stem cell differentiation, influence healing processes and modulate extracellular matrix (ECM) deposition needed for tissue development and maintenance. To better understand the biomechanical effect of matrix elasticity on the formation of articular cartilage analogs in vitro, this study aims at assessing the redifferentiation capacity of primary human chondrocytes in three different hydrogel matrices of predefined matrix elasticities. The hydrogel elasticities were chosen to represent a broad spectrum of tissue stiffness ranging from very soft tissues with a Young’s modulus of 1 kPa up to elasticities of 30 kPa, representative of the perichondral-space. In addition, the interplay of matrix elasticity and transforming growth factor beta-3 (TGF-β3) on the redifferentiation of primary human articular chondrocytes was studied by analyzing both qualitative (viability, morphology, histology) and quantitative (RT-qPCR, sGAG, DNA) parameters, crucial to the chondrotypic phenotype. Results show that fibrin hydrogels of 30 kPa Young’s modulus best guide chondrocyte redifferentiation resulting in a native-like morphology as well as induces the synthesis of physiologic ECM constituents such as glycosaminoglycans (sGAG) and collagen type II. This comprehensive study sheds light onto the mechanobiological impact of matrix elasticity on formation and maintenance of articular cartilage and thus represents a major step toward meeting the need for advanced in vitro tissue models to study both re- and degeneration of articular cartilage.
Collapse
Affiliation(s)
- Barbara Bachmann
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Competence Center MechanoBiology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sarah Spitz
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Barbara Schädl
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Andreas H Teuschl
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
| | - Heinz Redl
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sylvia Nürnberger
- AUVA Research Centre, Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Division of Trauma-Surgery, Department of Orthopedics and Trauma-Surgery, Medical University of Vienna, Vienna, Austria
| | - Peter Ertl
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
88
|
Walsh SK, Schneider SE, Amundson LA, Neu CP, Henak CR. Maturity-dependent cartilage cell plasticity and sensitivity to external perturbation. J Mech Behav Biomed Mater 2020; 106:103732. [PMID: 32321631 DOI: 10.1016/j.jmbbm.2020.103732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/04/2020] [Accepted: 03/13/2020] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Articular cartilage undergoes biological and morphological changes throughout maturation. The prevalence of osteoarthritis in the aged population suggests that maturation predisposes cartilage to degradation and/or impaired regeneration, but this process is not fully understood. Therefore, the objective of this study was to characterize the cellular and genetic profile of cartilage, as well as biological plasticity in response to mechanical and culture time stimuli, as a function of animal maturity. METHODS/DESIGN Porcine articular cartilage explants were harvested from stifle joints of immature (2-4 weeks), adolescent (5-6 months), and mature (1-5 years) animals. Half of all samples were subjected to a single compressive mechanical load. Loaded samples were paired with unloaded controls for downstream analyses. Expression of cartilage progenitor cell markers CD105, CD44, and CD29 were determined via flow cytometry. Expression of matrix synthesis genes Col1, Col2, Col10, ACAN, and SOX9 were determined via qPCR. Tissue morphology and matrix content were examined histologically. Post-loading assays were performed immediately and following 7 days in culture. RESULTS CD105 and CD29 expression decreased with maturity, while CD44 expression was upregulated in cartilage from mature animals. Expression of matrix synthesis genes were generally upregulated in cartilage from mature animals, and adolescent animals showed the lowest expression of several matrix synthesizing genes. Culture time and mechanical loading analyses revealed greater plasticity to mechanical loading and culture time in cartilage from younger animals. Histology confirmed distinct structural and biochemical profiles across maturity. CONCLUSION This study demonstrates differential, nonlinear expression of chondroprogenitor markers and matrix synthesis genes as a function of cartilage maturity, as well as loss of biological plasticity in aged tissue. These findings have likely implications for age-related loss of regeneration and osteoarthritis progression.
Collapse
Affiliation(s)
- Shannon K Walsh
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI, USA.
| | - Stephanie E Schneider
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA.
| | - Laura A Amundson
- Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| | - Corey P Neu
- Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO, USA.
| | - Corinne R Henak
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
89
|
Muschter D, Fleischhauer L, Taheri S, Schilling AF, Clausen-Schaumann H, Grässel S. Sensory neuropeptides are required for bone and cartilage homeostasis in a murine destabilization-induced osteoarthritis model. Bone 2020; 133:115181. [PMID: 31926346 DOI: 10.1016/j.bone.2019.115181] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 11/13/2019] [Accepted: 11/28/2019] [Indexed: 12/16/2022]
Abstract
Numerous studies identified a role for the sensory neuropeptides substance P (SP) and alpha calcitonin gene-related peptide (αCGRP) in osteoarthritis (OA) pain behavior. Surprisingly, little attention has been paid on how their trophic effects on cartilage and bone cells might affect structural changes of bone and cartilage in OA pathology. Here, we sought to elucidate sensory neuropeptides influence on structural alterations of bone and cartilage during murine OA pathophysiology. OA was induced by destabilization of the medial meniscus (DMM) in the right knee joint of 12 weeks old male C57Bl/6J wildtype (WT) mice and mice either deficient for SP (tachykinin 1 (Tac1)-/-) or αCGRP. By OARSI histopathological grading we observed significant cartilage matrix degradation after DMM surgery in αCGRP-deficient mice after 4 weeks whereas Tac1-/- scores were not different to sham mice before 12 weeks after surgery. Indentation-type atomic force microscopy (IT-AFM) identified a strong superficial zone (SZ) cartilage phenotype in Tac1-/- Sham mice. Opposed to WT and αCGRP-/- mice, SZ cartilage of Tac1-/- mice softened 2 weeks after OA induction. In Tac1-/- DMM mice, bone volume to total volume ratio (BV/TV) increased significantly compared to the Tac1-/- Sham group, 2 weeks after surgery. WT mice had reduced BV/TV compared to αCGRP-/- and Tac1-/- mice after 12 weeks. Increased calcified cartilage thickness and medial condyle diameter were detected in the medial tibia of all groups 8 weeks after OA induction by nanoCT analysis. Meniscal ossification occurred in all OA groups, but was significantly stronger in the absence of neuropeptides. Increased serum concentration of the respective non-deleted neuropeptide was observed in both neuropeptide-deficient mice strains. Both neuropeptides protect from age-related bone structural changes under physiological conditions and SP additionally demonstrates an anabolic effect on bone structure preservation in a pathophysiological situation. Both neuropeptide deficient mice display an intrinsic structural cartilage matrix phenotype that might alter progression of cartilage degeneration in OA.
Collapse
Affiliation(s)
- Dominique Muschter
- Dept. of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), Bio Park 1, University of Regensburg, Germany.
| | - Lutz Fleischhauer
- Department of Applied Sciences and Mechatronics, University of Applied Sciences Munich, Germany; Laboratory of Experimental Surgery and Regenerative Medicine, Clinic for General, Trauma and Reconstructive Surgery, Ludwig-Maximilians-University, Munich, Germany; Center for NanoScience, Ludwig-Maximilians-University, Munich, Germany.
| | - Shahed Taheri
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medicine Göttingen.
| | - Arndt F Schilling
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medicine Göttingen.
| | - Hauke Clausen-Schaumann
- Department of Applied Sciences and Mechatronics, University of Applied Sciences Munich, Germany.
| | - Susanne Grässel
- Dept. of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), Bio Park 1, University of Regensburg, Germany.
| |
Collapse
|
90
|
Embrittlement of collagen in early-stage human osteoarthritis. J Mech Behav Biomed Mater 2020; 104:103663. [PMID: 32174421 DOI: 10.1016/j.jmbbm.2020.103663] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 01/08/2020] [Accepted: 01/27/2020] [Indexed: 11/20/2022]
Abstract
Articular cartilage is a remarkable material with mechanical performance that surpasses engineering standards. Collagen, the most abundant protein in cartilage, plays an important role in this performance, and also in disease. Building on observations of network-level collagen changes at the earliest stages of osteoarthritis, this study explores the physical role of the collagen fibril in the disease process. Specifically, we focus on the material properties of collagen fibrils in the cartilage surface. Ten human tibial plateaus were characterised by atomic force microscopy (AFM) and Raman spectroscopy, with histological scoring used to define disease state. Measures of tropocollagen remained stable with disease progression, yet a marked mechanical change was observed. A slight stiffening coupled with a substantial decrease in loss tangent suggests a physical embrittlement caused by increased inter-molecular interactions.
Collapse
|
91
|
Kiio TM, Park S. Nano-scientific Application of Atomic Force Microscopy in Pathology: from Molecules to Tissues. Int J Med Sci 2020; 17:844-858. [PMID: 32308537 PMCID: PMC7163363 DOI: 10.7150/ijms.41805] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/26/2020] [Indexed: 12/28/2022] Open
Abstract
The advantages of atomic force microscopy (AFM) in biological research are its high imaging resolution, sensitivity, and ability to operate in physiological conditions. Over the past decades, rigorous studies have been performed to determine the potential applications of AFM techniques in disease diagnosis and prognosis. Many pathological conditions are accompanied by alterations in the morphology, adhesion properties, mechanical compliances, and molecular composition of cells and tissues. The accurate determination of such alterations can be utilized as a diagnostic and prognostic marker. Alteration in cell morphology represents changes in cell structure and membrane proteins induced by pathologic progression of diseases. Mechanical compliances are also modulated by the active rearrangements of cytoskeleton or extracellular matrix triggered by disease pathogenesis. In addition, adhesion is a critical step in the progression of many diseases including infectious and neurodegenerative diseases. Recent advances in AFM techniques have demonstrated their ability to obtain molecular composition as well as topographic information. The quantitative characterization of molecular alteration in biological specimens in terms of disease progression provides a new avenue to understand the underlying mechanisms of disease onset and progression. In this review, we have highlighted the application of diverse AFM techniques in pathological investigations.
Collapse
Affiliation(s)
| | - Soyeun Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Daegu 42601, Republic of Korea
| |
Collapse
|
92
|
Hernandez PA, Wells J, Usheva E, Nakonezny PA, Barati Z, Gonzalez R, Kassem L, Henson FMD. Early-Onset Osteoarthritis originates at the chondrocyte level in Hip Dysplasia. Sci Rep 2020; 10:627. [PMID: 31953438 PMCID: PMC6969105 DOI: 10.1038/s41598-020-57431-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/31/2019] [Indexed: 01/05/2023] Open
Abstract
Subjects with developmental dysplasia of the hip (DDH) often show early-onset osteoarthritis (OA); however, the molecular mechanisms underlying this pathology are not known. We investigated whether cellular changes in chondrocytes from OA cartilage can be detected in chondrocytes from DDH cartilage before histological manifestations of degeneration. We characterized undamaged and damaged articular cartilage from 22 participants having hip replacement surgery with and without DDH (9 DDH-OA, 12 OA-only, one femoral fracture). Tissue immunostaining revealed changes in damaged OA-only cartilage that was also found in undamaged DDH-OA cartilage. Chondrocytes in situ from both groups show: (i) thicker fibers of vimentin intermediate filaments, (ii) clusters of integrin α5β1, (iii) positive MMP13 staining and (iv) a higher percentage of cells expressing the serine protease HtrA1. Further characterization of the extracellular matrix showed strong aggrecan and collagen II immunostaining in undamaged DDH cartilage, with no evidence of augmented cell death by activation of caspase 3. These findings suggest that early events in DDH cartilage originate at the chondrocyte level and that DDH cartilage may provide a novel opportunity to study these early changes for the development of therapeutic targets for OA.
Collapse
Affiliation(s)
- Paula A Hernandez
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Joel Wells
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Emiliya Usheva
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Paul A Nakonezny
- Department of Population and Data Sciences, Division of Biostatistics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Zahra Barati
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Roberto Gonzalez
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Layla Kassem
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Frances M D Henson
- Division of Trauma and Orthopaedic Surgery, University of Cambridge, Cambridge, CB2 2QQ, UK
| |
Collapse
|
93
|
Shoaib T, Yuh C, Wimmer MA, Schmid TM, Espinosa-Marzal RM. Nanoscale insight into the degradation mechanisms of the cartilage articulating surface preceding OA. Biomater Sci 2020; 8:3944-3955. [DOI: 10.1039/d0bm00496k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Osteoarthritis (OA) is a degenerative disease and leading cause of disability globally. We report the a fundamental study of the mechanisms underlying deterioration of hydrated cartilage in the presence of elevated calcium content preceding OA.
Collapse
Affiliation(s)
- Tooba Shoaib
- Materials Science and Engineering
- University of Illinois at Urbana-Champaign
- Urbana
- USA
| | - Catherine Yuh
- Department of Orthopedics
- Rush University Medical Center
- Chicago
- USA
| | - Markus A. Wimmer
- Department of Orthopedics
- Rush University Medical Center
- Chicago
- USA
| | - Thomas M. Schmid
- Department of Orthopedics
- Rush University Medical Center
- Chicago
- USA
| | - Rosa M. Espinosa-Marzal
- Materials Science and Engineering
- University of Illinois at Urbana-Champaign
- Urbana
- USA
- Civil and Environmental Engineering Department
| |
Collapse
|
94
|
Zhang X, Cai D, Zhou F, Yu J, Wu X, Yu D, Zou Y, Hong Y, Yuan C, Chen Y, Pan Z, Bunpetch V, Sun H, An C, Yi-Chin T, Ouyang H, Zhang S. Targeting downstream subcellular YAP activity as a function of matrix stiffness with Verteporfin-encapsulated chitosan microsphere attenuates osteoarthritis. Biomaterials 2019; 232:119724. [PMID: 31918221 DOI: 10.1016/j.biomaterials.2019.119724] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/20/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022]
Abstract
Changes in the stiffness of chondrocyte extracellular matrix (ECM) are involved in the pathological progression of osteoarthritis (OA). However, the downstream responses of cartilage ECM stiffness are still unclear. YAP (Yes-associated protein) has been extensively studied as a mechanotransducer, we thus hypothesized that by targeting the downstream molecule activity of ECM stiffness could maintain chondrocyte phenotype and prevent cartilage degeneration in OA. Here, we showed that human cartilage matrix stiffened during pathological progression of OA, and the chondrocyte YAP activity was associated with ECM stiffness. We then mimicked the physiological and pathological stiffness of human cartilage by using PDMS-based substrates, and found that YAP was activated in chondrocytes seeded on stiff substrate, gradually losing their phenotype. In addition, it was observed that YAP was also significantly activated in mice OA development, and conditional knockout (cKO) of YAP in mice preserved collagen II expression and protected cartilage from degeneration in the OA model. Furthermore, intra-articular injection of YAP-selective inhibitor, Verteporfin, significantly maintained cartilage homeostasis in mice OA model. This study indicates that the application of mechanotransducer-targeted drugs could be a potential therapeutic approach for cartilage repair in OA.
Collapse
Affiliation(s)
- Xianzhu Zhang
- School of Basic Medical Sciences and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Dandan Cai
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Feifei Zhou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Yu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyu Wu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongsheng Yu
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiwei Zou
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Hong
- School of Basic Medical Sciences and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunhui Yuan
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Yishan Chen
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongyou Pan
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Varitsara Bunpetch
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Sun
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengrui An
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Toh Yi-Chin
- Department of Biomedical Engineering, National University of Singapore 4, Engineering Drive 3, E4-04-10, 117583, Singapore
| | - Hongwei Ouyang
- School of Basic Medical Sciences and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University-University of Edinburgh Institute & School of Basic Medicine, Zhejiang University School of Medicine, Hangzhou, China; Department of Sports Medicine, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| | - Shufang Zhang
- School of Basic Medical Sciences and Department of Orthopedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China; China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, China.
| |
Collapse
|
95
|
Mieloch AA, Richter M, Trzeciak T, Giersig M, Rybka JD. Osteoarthritis Severely Decreases the Elasticity and Hardness of Knee Joint Cartilage: A Nanoindentation Study. J Clin Med 2019; 8:jcm8111865. [PMID: 31684201 PMCID: PMC6912408 DOI: 10.3390/jcm8111865] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/30/2019] [Accepted: 11/01/2019] [Indexed: 01/19/2023] Open
Abstract
The nanoindentation method was applied to determine the elastic modulus and hardness of knee articular cartilage. Cartilage samples from both high weight bearing (HWB) and low weight bearing (LWB) femoral condyles were collected from patients diagnosed with osteoarthritis (OA). The mean elastic modulus of HWB cartilage was 4.46 ± 4.44 MPa in comparison to that of the LWB region (9.81 ± 8.88 MPa, p < 0.001). Similarly, the hardness was significantly lower in HWB tissue (0.317 ± 0.397 MPa) than in LWB cartilage (0.455 ± 0.434 MPa, p < 0.001). When adjusted to patients’ ages, the mean elastic modulus and hardness were both significantly lower in the age group over 70 years (p < 0.001). A statistically significant difference in mechanical parameters was also found in grade 3 and 4 OA. This study provides an insight into the nanomechanical properties of the knee articular cartilage and provides a starting point for personalized cartilage grafts that are compatible with the mechanical properties of the native tissue.
Collapse
Affiliation(s)
- Adam Aron Mieloch
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Uniwersytetu Poznańskiego 10 Street, 61-614 Poznan, Poland.
- Faculty of Chemistry, Adam Mickiewicz University in Poznan, Uniwersytetu Poznańskiego 8 Street, 61-614 Poznan, Poland.
| | - Magdalena Richter
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Uniwersytetu Poznańskiego 10 Street, 61-614 Poznan, Poland.
| | - Tomasz Trzeciak
- Department of Orthopedics and Traumatology, Poznan University of Medical Sciences, 28 czerwca 1956r. Street No. 135/147, 61-545 Poznan, Poland.
| | - Michael Giersig
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Uniwersytetu Poznańskiego 10 Street, 61-614 Poznan, Poland.
- Department of Physics, Institute of Experimental Physics, Freie Universität, Arnimallee 14, 14195 Berlin, Germany.
| | - Jakub Dalibor Rybka
- Center for Advanced Technology, Adam Mickiewicz University in Poznan, Uniwersytetu Poznańskiego 10 Street, 61-614 Poznan, Poland.
| |
Collapse
|
96
|
Han B, Li Q, Wang C, Patel P, Adams SM, Doyran B, Nia HT, Oftadeh R, Zhou S, Li CY, Liu XS, Lu XL, Enomoto-Iwamoto M, Qin L, Mauck RL, Iozzo RV, Birk DE, Han L. Decorin Regulates the Aggrecan Network Integrity and Biomechanical Functions of Cartilage Extracellular Matrix. ACS NANO 2019; 13:11320-11333. [PMID: 31550133 PMCID: PMC6892632 DOI: 10.1021/acsnano.9b04477] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Joint biomechanical functions rely on the integrity of cartilage extracellular matrix. Understanding the molecular activities that govern cartilage matrix assembly is critical for developing effective cartilage regeneration strategies. This study elucidated the role of decorin, a small leucine-rich proteoglycan, in the structure and biomechanical functions of cartilage. In decorin-null cartilage, we discovered a substantial reduction of aggrecan content, the major proteoglycan of cartilage matrix, and mild changes in collagen fibril nanostructure. This loss of aggrecan resulted in significantly impaired biomechanical properties of cartilage, including decreased modulus, elevated hydraulic permeability, and reduced energy dissipation capabilities. At the cellular level, we found that decorin functions to increase the retention of aggrecan in the neo-matrix of chondrocytes, rather than to directly influence the biosynthesis of aggrecan. At the molecular level, we demonstrated that decorin significantly increases the adhesion between aggrecan and aggrecan molecules and between aggrecan molecules and collagen II fibrils. We hypothesize that decorin plays a crucial structural role in mediating the matrix integrity and biomechanical functions of cartilage by providing physical linkages to increase the adhesion and assembly of aggrecan molecules at the nanoscale.
Collapse
Affiliation(s)
- Biao Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Qing Li
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Pavan Patel
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Sheila M. Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Basak Doyran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Hadi T. Nia
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts 02215, United States
| | - Ramin Oftadeh
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Siyuan Zhou
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Christopher Y. Li
- Department of Materials Science and Engineering, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - X. Sherry Liu
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - X. Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Motomi Enomoto-Iwamoto
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, Maryland 21201, United States
| | - Ling Qin
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Robert L. Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Renato V. Iozzo
- Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, United States
| | - David E. Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, Florida 33612, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
97
|
Marchiori G, Berni M, Boi M, Filardo G. Cartilage mechanical tests: Evolution of current standards for cartilage repair and tissue engineering. A literature review. Clin Biomech (Bristol, Avon) 2019; 68:58-72. [PMID: 31158591 DOI: 10.1016/j.clinbiomech.2019.05.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Repair procedures and tissue engineering are solutions available in the clinical practice for the treatment of damaged articular cartilage. Regulatory bodies defined the requirements that any products, intended to regenerate cartilage, should have to be applied. In order to verify these requirements, the Food and Drug Administration (FDA, USA) and the International Standard Organization (ISO) indicated some Standard tests, which allow evaluating, in a reproducible way, the performances of scaffolds/treatments for cartilage tissue regeneration. METHODS A review of the literature about cartilage mechanical characterization found 394 studies, from 1970 to date. They were classified by material (simulated/animal/human cartilage) and method (theoretical/applied; static/dynamic; standard/non-standard study), and analyzed by nation and year of publication. FINDINGS While Standard methods for cartilage mechanical characterization still refer to studies developed in the eighties, expertise and interest on cartilage mechanics research are evolving continuously and internationally, with studies both in vitro - on human and animal tissues - and in silico, dealing with tissue function and modelling, using static and dynamic loading conditions. INTERPRETATION there is a consensus on the importance of mechanical characterization that should be considered to evaluate cartilage treatments. Still, relative Standards need to be updated to describe advanced constructs and procedures for cartilage regeneration in a more exhaustive way. The use of the more complex, fibre-reinforced biphasic model, instead of the standard simple biphasic model, to describe cartilage response to loading, and the standardisation of dynamic tests can represent a first step in this direction.
Collapse
Affiliation(s)
- Gregorio Marchiori
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Biomechanics and Technology Innovation, Via di Barbiano 1/10, 40136 Bologna, Italy.
| | - Matteo Berni
- IRCCS Istituto Ortopedico Rizzoli, Laboratory of Biomechanics and Technology Innovation, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Marco Boi
- IRCCS Istituto Ortopedico Rizzoli, NanoBiotechnology Laboratory (NaBi), Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Giuseppe Filardo
- IRCCS Istituto Ortopedico Rizzoli, NanoBiotechnology Laboratory (NaBi), Via di Barbiano 1/10, 40136 Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Applied and Translational Research Center, Via di Barbiano 1/10, 40136 Bologna, Italy
| |
Collapse
|
98
|
Marchi G, Foehr P, Baier V, Kinne RW, Burgkart R, Roths J. Discrimination between healthy and degenerated bovine articular cartilage with a fiber Bragg grating based microindenter. J Mech Behav Biomed Mater 2019; 99:11-17. [PMID: 31323532 DOI: 10.1016/j.jmbbm.2019.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE In this study we aim to show that an optical fiber Bragg grating-based microindentation system, which has the potential to be deployed arthroscopically, can differentiate between healthy and degenerated articular cartilage, which represents an important challenge in minimally-invasive surgery. DESIGN Twenty bovine osteochondral cylinders, extracted from the patellar groove of ten 24 months old animals were subjected to stepwise in vitro stress-relaxation indentation measurements. The indentation procedure comprised 15 indentation steps of 20 μm each, reaching a total depth of 300 μm. Ten samples remained untreated and served as a control group for healthy cartilage. A second group of ten samples was treated for 12 h with an aqueous trypsin solution (concentration 2.5%) to deplete the proteoglycans. For both groups and all indentation depths deeper than 100 μm, the step response functions of a two elements Maxwell-Wiechert model fitted well to the measured relaxation curves. RESULTS The standard deviations of the identified stiffness parameters within each group were much smaller than the difference of the average stiffness values between both groups. Based on the measured stiffness values, the system was capable to discriminate between healthy and degenerated cartilage with a high level of significance (p < 0.001). The experimental results are also discussed in terms of the biomechanical changes of cartilage under the action of trypsin. CONCLUSION The fiber Bragg grating microindentation system showed the capability to differentiate intact and proteoglycan depleted cartilage with high significance.
Collapse
Affiliation(s)
- G Marchi
- Munich University of Applied Sciences, Photonics Laboratory, Lothstr. 34, Munich, 80335, Germany.
| | - P Foehr
- Department of Orthopaedics and Sports Orthopaedics, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, Munich, 81675, Germany.
| | - V Baier
- Munich University of Applied Sciences, Photonics Laboratory, Lothstr. 34, Munich, 80335, Germany.
| | - R W Kinne
- Experimental Rheumatology Unit, Department of Orthopaedics, Jena University Hospital, Waldkrankenhaus "Rudolf Elle", Klosterlausnitzer Str. 81, Eisenberg, 07607, Germany.
| | - R Burgkart
- Department of Orthopaedics and Sports Orthopaedics, Klinikum Rechts der Isar, Technical University of Munich, Ismaninger Str. 22, Munich, 81675, Germany.
| | - J Roths
- Munich University of Applied Sciences, Photonics Laboratory, Lothstr. 34, Munich, 80335, Germany.
| |
Collapse
|
99
|
Kontomaris SV, Stylianou A, Nikita KS, Malamou A, Stylianopoulos T. A simplified approach for the determination of fitting constants in Oliver-Pharr method regarding biological samples. Phys Biol 2019; 16:056003. [PMID: 31137017 DOI: 10.1088/1478-3975/ab252e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The atomic force microscopy (AFM) nanoindentation regarding biological samples is a challenging procedure. Biological samples at the nanoscale can be considered as purely elastic materials under the condition that the indentation depth is very small and the indenter is smooth. However, the indenters that are commonly used are pyramidal and in several cases the indentation depths are big comparing to the dimensions of the tip apex. Hence, pyramidal indenters usually cause a permanent damage to the sample. In this case, the best model that can be applied for the data processing is the Oliver-Pharr model which takes into account the elastic-plastic behavior of the sample. The Oliver-Pharr model is based on the fitting of the unloading load-indentation data to a power law equation. In this paper a simplified procedure which ensures the accurate fitting of the unloading load-indentation data to the Oliver-Pharr model is presented and validated on experimental data obtained from a human glioma cell line. It should be noted that the proposed method can be also applied for the data fitting in the case of purely elastic response.
Collapse
Affiliation(s)
- S V Kontomaris
- Mobile Radio Communications Laboratory, School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece. Athens Metropolitan College, Athens, Greece. Author to whom any correspondence should be addressed
| | | | | | | | | |
Collapse
|
100
|
Viji Babu PK, Radmacher M. Mechanics of Brain Tissues Studied by Atomic Force Microscopy: A Perspective. Front Neurosci 2019; 13:600. [PMID: 31258462 PMCID: PMC6587663 DOI: 10.3389/fnins.2019.00600] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/27/2019] [Indexed: 01/17/2023] Open
Abstract
Tissue morphology and mechanics are crucial to the regulation of organ function. Investigating the exceptionally complex tissue of the brain at the sub-micron scale is challenging due to the complex structure and softness of this tissue, despite the large interest of biologists, medical engineers, biophysicists, and others in this topic. Atomic force microscopy (AFM) both as an imaging and as a mechanical tool provides an excellent opportunity to study soft biological samples such as live brain tissues. Here we review the principles of AFM, the performance of AFM in tissue imaging and mechanical mapping of cells and tissues, and finally opening the prospects and challenges of probing the biophysical properties of brain tissue using AFM.
Collapse
|