1
|
Xie X, Chao R, Mao Y, Wan T, Wang Y, Zhu Y, Xu W, Chen X, Wang Y, Ma Z, Zhang S. Osteoarthritis-like changes in rat temporomandibular joint induced by unilateral anterior large overjet treatment. Sci Rep 2025; 15:1646. [PMID: 39794380 PMCID: PMC11723919 DOI: 10.1038/s41598-024-81306-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 11/26/2024] [Indexed: 01/13/2025] Open
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a common degenerative disease that causes chronic pain and joint dysfunction. However, the current understanding of TMJOA pathogenesis is limited and necessitates further research. Animal models are crucial for investigating TMJOA due to the scarcity of clinical samples. Class II malocclusion is an occlusal type highly associated with TMJOA, but it currently lacks appropriate animal models for simulating this malocclusion in research. Therefore, this study develops a new malocclusion model using a unilateral anterior large overjet (UALO) dental device to cause Class II malocclusion characteristics and TMJOA-like pathological alterations in rats. By inducing a posteriorly positioned condyle, the UALO device effectively results in cartilage degradation, subchondral bone loss, condylar volume reduction, and mandibular retrusion. Furthermore, RNA sequencing of condylar cartilages revealed that the oxidative stress of chondrocytes was elevated under the UALO-triggered abnormal mechanical stress. Disruption of antioxidant systems and mitochondrial dysfunction are involved in cartilage degeneration. The current study provides a novel and reliable rat model suitable for TMJOA research and offers insights into the disease's potential mechanistic pathways and molecular targets, contributing to a deeper understanding of TMJOA.
Collapse
Affiliation(s)
- Xinru Xie
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Rui Chao
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yi Mao
- State Key Laboratory of Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, 610041, China
| | - Tianhao Wan
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yexin Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yan Zhu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Weifeng Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Xuzhuo Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- National Center for Stomatology, Shanghai, 200011, China
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China
| | - Yong Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China.
- National Center for Stomatology, Shanghai, 200011, China.
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
| | - Zhigui Ma
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China.
- National Center for Stomatology, Shanghai, 200011, China.
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
| | - Shanyong Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China.
- National Center for Stomatology, Shanghai, 200011, China.
- National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
- Shanghai Key Laboratory of Stomatology, Shanghai, 200011, China.
| |
Collapse
|
2
|
Wang J, Yang J, Fang Y, Lou C, Yu H, Li Y, Lv J, Chen H, Cai L, Zheng W. Vinpocetine protects against osteoarthritis by inhibiting ferroptosis and extracellular matrix degradation via activation of the Nrf2/GPX4 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156115. [PMID: 39368343 DOI: 10.1016/j.phymed.2024.156115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Osteoarthritis (OA) is a progressive joint condition marked by the slow degradation of articular cartilage. Vinpocetine (Vin), a synthetic derivative of vincamine derived from the vinca plant, exhibits anti-inflammatory and antioxidant properties. Nevertheless, the specific role and mechanism of Vin in the treatment of OA remain largely unexplored. OBJECTIVES The study is designed to uncover the impacts of Vin on tert‑butyl hydroperoxide (TBHP)-induced ferroptosis and to explore its potential role and underlying mechanisms in the treatment of OA. Concurrently, we established an OA mouse model through medial meniscal instability surgery to assess the therapeutic effects of Vin in vivo. METHODS Through network pharmacology analysis, we have identified the key targets and potential pathways of Vin. To simulate an oxidative stress-induced OA environment in vitro, we induced chondrocyte injury using TBHP. We tested how Vin affects chondrocytes under TBHP induction by DHE and DCFH-DA probes, BODIPY-C11 and FerroOrange staining, mitochondrial function assessment, Western blotting, co-immunoprecipitation, and immunofluorescence techniques. Simultaneously, we established an OA mouse model through medial meniscal instability surgery to assess the in vivo therapeutic effects of Vin. In this model, we used X-ray and micro-CT imaging, SO staining, TB staining, H&E staining, and immunohistochemistry to analyze the role of Vin in detail. RESULTS This study demonstrated that Vin effectively suppressed TBHP-induced ferroptosis and extracellular matrix (ECM) degradation and significantly lessened mitochondrial damage associated with ferroptosis. In the OA mouse model, Vin improved cartilage degeneration, subchondral remodeling, synovitis, and ECM degradation. Vin worked by activating the Nrf2/GPX4 pathway and inhibiting the Keap1-Nrf2 interaction. This study focused on the function of ferroptosis in OA and its influence on chondrocyte damage and disease progression, offering novel perspectives on potential treatments. CONCLUSION Vin activated the Nrf2/GPX4 pathway, thereby slowing OA progression, inhibiting ferroptosis, and preventing ECM degradation.
Collapse
Affiliation(s)
- Jinwu Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Jin Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Yuqin Fang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Chao Lou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Heng Yu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Yangbo Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Junlei Lv
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Hua Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China.
| | - Leyi Cai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China.
| | - Wenhao Zheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China.
| |
Collapse
|
3
|
Boretti G, Amirfallah A, Edmunds KJ, Hamzehpour H, Sigurjónsson ÓE. Advancing Cartilage Tissue Engineering: A Review of 3D Bioprinting Approaches and Bioink Properties. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39381849 DOI: 10.1089/ten.teb.2024.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Articular cartilage is crucial in human physiology, and its degeneration poses a significant public health challenge. While recent advancements in 3D bioprinting and tissue engineering show promise for cartilage regeneration, there remains a gap between research findings and clinical application. This review critically examines the mechanical and biological properties of hyaline cartilage, along with current 3D manufacturing methods and analysis techniques. Moreover, we provide a quantitative synthesis of bioink properties used in cartilage tissue engineering. After screening 181 initial works, 33 studies using extrusion bioprinting were analyzed and synthesized, presenting results that indicate the main materials, cells, and methods utilized for mechanical and biological evaluation. Altogether, this review motivates the standardization of mechanical analyses and biomaterial assessments of 3D bioprinted constructs to clarify their chondrogenic potential.
Collapse
Affiliation(s)
- Gabriele Boretti
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
| | - Arsalan Amirfallah
- The Blood Bank, Landspitali, The National University Hospital of Iceland, Reykjavík, Iceland
| | - Kyle J Edmunds
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
| | - Helena Hamzehpour
- Faculty of Pharmaceutical Sciences, University of Iceland, Reykjavik, Iceland
| | - Ólafur E Sigurjónsson
- School of Science and Engineering, Reykjavík University, Reykjavík, Iceland
- The Blood Bank, Landspitali, The National University Hospital of Iceland, Reykjavík, Iceland
| |
Collapse
|
4
|
Liman S, Gomez-Contreras PC, Hines MR, Witt E, Fisher JS, Lu KJ, McNally LD, Cotoia AT, Sakyi MY, Wagner BA, Tift MS, Goetz JE, Byrne JD, Coleman MC. Activation of Heme Metabolism Promotes Tissue Health After Intraarticular Injury or Surgical Exposure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596521. [PMID: 39416169 PMCID: PMC11482836 DOI: 10.1101/2024.05.29.596521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Posttraumatic osteoarthritis (PTOA) is a well-recognized public health burden without any disease modifying treatment. This occurs despite noted advances in surgical care in the past 50 years. Mitochondrial oxidative damage pathways initiate PTOA after severe injuries like intraarticular fracture that often require surgery and contribute to PTOA after less severe injuries that may or may not require surgery like meniscal injuries. When considering the mitochondrial and redox environment of the injured joint, we hypothesized that activation of heme metabolism, previously associated with healing in many settings, would cause prototypic mitochondrial reprogramming effects in cartilage ideally suited for use at the time of injury repair. Activation of heme metabolism can be accomplished through the gasotransmitter carbon monoxide (CO), which activates hemeoxygenase-1 (HO1) and subsequent heme metabolism. In this study, we employed unique carbon monoxide (CO)-containing foam (COF) to stimulate heme metabolism and restore chondrocyte oxygen metabolism in vitro and in vivo . Doxycycline-inducible, chondrocyte-specific HO1 overexpressing transgenic mice show similar mitochondrial reprogramming after induction compared to COF. CO is retained at least 24 h after COF injection into stifle joints and induces sustained increases in heme metabolism. Lastly, intraarticular injection of COF causes key redox outcomes without any adverse safety outcomes in rabbit stifle joints ex vivo and in vivo . We propose that activation of heme metabolism is an ideal adjuvant to trauma care that replenishes chondrocyte mitochondrial metabolism and restores redox homeostasis.
Collapse
|
5
|
Hines MR, Gomez-Contreras PC, Liman S, Wilson AM, Lu KJ, O'Neill JA, Fisher JS, Fredericks DC, Wagner BA, Buettner GR, Van Remmen H, Coleman MC. A reciprocal relationship between mitochondria and lipid peroxidation determines the chondrocyte intracellular redox environment. Redox Biol 2024; 75:103306. [PMID: 39133964 PMCID: PMC11366903 DOI: 10.1016/j.redox.2024.103306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
In orthopedic research, many studies have applied vitamin E as a protective antioxidant or used tert-butyl hydroperoxide to induce oxidative injury to chondrocytes. These studies often support the hypothesis that joint pathology causes oxidative stress and increased lipid peroxidation that might be prevented with lipid antioxidants to improve cell survival or function and joint health; however, lipid antioxidant supplementation was ineffective against osteoarthritis in clinical trials and animal data have been equivocal. Moreover, increased circulating vitamin E is associated with increased rates of osteoarthritis. This disconnect between benchtop and clinical results led us to hypothesize that oxidative stress-driven paradigms of chondrocyte redox function do not capture the metabolic and physiologic effects of lipid antioxidants and prooxidants on articular chondrocytes. We used ex vivo and in vivo cartilage models to investigate the effect of lipid antioxidants on healthy, primary, articular chondrocytes and applied immuno-spin trapping techniques to provide a broad indicator of high levels of oxidative stress independent of specific reactive oxygen species. Key findings demonstrate lipid antioxidants were pro-mitochondrial while lipid prooxidants decreased mitochondrial measures. In the absence of injury, radical formation was increased by lipid antioxidants; however, in the presence of injury, radical formation was decreased. In unstressed conditions, this relationship between chondrocyte mitochondria and redox regulation was reproduced in vivo with overexpression of glutathione peroxidase 4. In mice aged 18 months or more, overexpression of glutathione peroxidase 4 significantly decreased the presence of pro-mitochondrial peroxisome proliferation activated receptor gamma and deranged the relationship between mitochondria and the redox environment. This complex interaction suggests strategies targeting articular cartilage may benefit from adopting more nuanced paradigms of articular chondrocyte redox metabolism.
Collapse
Affiliation(s)
| | | | | | | | - Kevin J Lu
- The University of Iowa, Iowa City, IA, USA
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Liu Y, Chen P, Hu B, Xiao Y, Su T, Luo X, Tu M, Cai G. Excessive mechanical loading promotes osteoarthritis development by upregulating Rcn2. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167251. [PMID: 38795835 DOI: 10.1016/j.bbadis.2024.167251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/28/2024]
Abstract
Exposure of articular cartilage to excessive mechanical loading is closely related to the pathogenesis of osteoarthritis (OA). However, the exact molecular mechanism by which excessive mechanical loading drives OA remains unclear. In vitro, primary chondrocytes were exposed to cyclic tensile strain at 0.5 Hz and 10 % elongation for 30 min to simulate excessive mechanical loading in OA. In vivo experiments involved mice undergoing anterior cruciate ligament transection (ACLT) to model OA, followed by interventions on Rcn2 expression through adeno-associated virus (AAV) injection and tamoxifen-induced gene deletion. 10 μL AAV2/5 containing AAV-Rcn2 or AAV-shRcn2 was administered to the mice by articular injection at 1 week post ACLT surgery, and Col2a1-creERT: Rcn2flox/flox mice were injected with tamoxifen intraperitoneally to obtain Rcn2-conditional knockout mice. Finally, we explored the mechanism of Rcn2 affecting OA. Here, we identified reticulocalbin-2 (Rcn2) as a mechanosensitive factor in chondrocytes, which was significantly elevated in chondrocytes under mechanical overloading. PIEZO type mechanosensitive ion channel component 1 (Piezo1) is a critical mechanosensitive ion channel, which mediates the effect of mechanical loading on chondrocytes, and we found that increased Rcn2 could be suppressed through knocking down Piezo1 under excessive mechanical loading. Furthermore, chondrocyte-specific deletion of Rcn2 in adult mice alleviated OA progression in the mice receiving the surgery of ACLT. On the contrary, articular injection of Rcn2-expressing adeno-associated virus (AAV) accelerated the progression of ACLT-induced OA in mice. Mechanistically, Rcn2 accelerated the progression of OA through promoting the phosphorylation and nuclear translocation of signal transducer and activator of transcription 3 (Stat3).
Collapse
Affiliation(s)
- Yalin Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Peng Chen
- Department of Orthopedic, Xiangya Hospital of Central South University, Changsha, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Manli Tu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China; Jiangxi Clinical Research Center for Endocrine and Metabolic Disease, China; Jiangxi Branch of National Clinical Research Center for metabolic Disease, China.
| | - Guangping Cai
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.
| |
Collapse
|
7
|
Liu K, Zhang B, Zhang X. Promoting Articular Cartilage Regeneration through Microenvironmental Regulation. J Immunol Res 2024; 2024:4751168. [PMID: 39104594 PMCID: PMC11300091 DOI: 10.1155/2024/4751168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
In recent years, as the aging population continues to grow, osteoarthritis (OA) has emerged as a leading cause of disability, with its incidence rising annually. Current treatments of OA include exercise and medications in the early stages and total joint replacement in the late stages. These approaches only relieve pain and reduce inflammation; however, they have significant side effects and high costs. Therefore, there is an urgent need to identify effective treatment methods that can delay the pathological progression of this condition. The changes in the articular cartilage microenvironment, which are complex and diverse, can aggravate the pathological progression into a vicious cycle, inhibiting the repair and regeneration of articular cartilage. Understanding these intricate changes in the microenvironment is crucial for devising effective treatment modalities. By searching relevant research articles and clinical trials in PubMed according to the keywords of articular cartilage, microenvironment, OA, mechanical force, hypoxia, cytokine, and cell senescence. This study first summarizes the factors affecting articular cartilage regeneration, then proposes corresponding treatment strategies, and finally points out the future research direction. We find that regulating the opening of mechanosensitive ion channels, regulating the expression of HIF-1, delivering growth factors, and clearing senescent cells can promote the formation of articular cartilage regeneration microenvironment. This study provides a new idea for the treatment of OA in the future, which can promote the regeneration of articular cartilage through the regulation of the microenvironment so as to achieve the purpose of treating OA.
Collapse
Affiliation(s)
- Kai Liu
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and MinistryGuangxi Medical University, Nanning, Guangxi 530021, China
| | - Bingjun Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xiaoling Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and MinistryGuangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
8
|
Shen Y, Yao W, Huang Y, Ye L, Liu J, Liu M, Ding J, Zhang Y. MRI analysis of and factors related to knee injuries in amateur marathon runners. PLoS One 2024; 19:e0306257. [PMID: 38980846 PMCID: PMC11232983 DOI: 10.1371/journal.pone.0306257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/13/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Marathons are the most challenging form of running, and amateur athletes may be more prone to injury due to a lack of professional knowledge and instruction in running. PURPOSE To analyze the MRI manifestations of and factors related to knee injuries in amateur marathon runners. SUBJECTS Data were collected from a hospital database of 105 qualified amateur marathon athletes (65 males,40 females), between May 2018 and December 2021. FIELD STRENGTH/SEQUENCE 1.5T MR: sagittal fs-PDWI, sagittal T1WI and sagittal 3D-DESS sequence. ASSESSMENT The MRI manifestations of knee joint injury were analyzed and evaluated by two radiologists. STATISTICAL TESTS The inter-observer agreement on MRI readings was analyzed using the kappa coefficient, and binary logistic regression analysis was employed to identify factors associated with knee injuries. RESULTS The overall prevalence of knee cartilage lesions, meniscus lesions and bone marrow edema among amateur marathon runners was 45.7%, 72.4%, and 49.5% respectively. Our analysis revealed that older age (OR = 1.135, P<0.001), higher BMI (OR = 1.236, P = 0.044), and slower pace (OR = 2.305, P = 0.017) were associated with increased risk of articular cartilage disease. Furthermore, older age (OR = 1.425, P<0.001) was identified as a risk factor for meniscal lesions, while older age (OR = 1.088, P = 0.002) was bone marrow edema. Notably, no significant correlation was observed between knee joint injuries of amateur marathon athletes and gender or the monthly running distance (P>0.05). CONCLUSIONS The occurrence of knee injuries among amateur marathon athletes was highly prevalent, with the patellofemoral joint cartilage and posterior horn of medial meniscus being frequently affected areas. Moreover, age, BMI, running years and pace were significant risk factors of knee joint injury.
Collapse
Affiliation(s)
- Yiying Shen
- Department of Radiology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wanzhen Yao
- Department of Radiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yi Huang
- Department of Radiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Lingxiao Ye
- Department of Radiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jie Liu
- Department of Radiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengxiao Liu
- MR scientific Marketing, Diagnostic Imaging, Siemens Healthineers Ltd, Shanghai, China
| | - Jianping Ding
- Department of Radiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Yanjing Zhang
- Department of Radiology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Liao S, Jia S, Yue Y, Zeng H, Lin J, Liu P. Advancements in pH-Responsive nanoparticles for osteoarthritis treatment: Opportunities and challenges. Front Bioeng Biotechnol 2024; 12:1426794. [PMID: 39036562 PMCID: PMC11260422 DOI: 10.3389/fbioe.2024.1426794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/06/2024] [Indexed: 07/23/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative disease linked to aging and obesity. The global aging population has led to an increasing number of OA patients, imposing a significant economic burden on society. Traditional drugs treatment methods often fail to achieve satisfactory outcomes. With the rapid advancement of nanomaterial delivery systems, numerous studies have focused on utilizing nanomaterials as carriers to achieve efficient OA treatment by effectively loading and delivering bioactive ingredients (e.g., drugs, nucleic acids) tailored to the unique pathological conditions, such as the weakly acidic microenvironment of synovial fluid in OA patients. This review highlights the latest advancements in the use of pH-responsive nanoparticles for OA treatment, emphasizing the principle of targeted drug delivery leveraging the acidic microenvironment of inflamed joints. It further discusses the composition, synthesis, response mechanism, target selection, application, and recent research findings of nanoparticles, while also addressing the challenges and future directions in this promising field.
Collapse
Affiliation(s)
- Shuai Liao
- Department of Bone and Joint Surgery, Peking
University Shenzhen Hospital, Shenzhen,
China
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen University School of Medicine,
Shenzhen, China
| | - Shicheng Jia
- Department of Sport Medicine, Peking
University Shenzhen Hospital, Shenzhen,
China
| | - Yaohang Yue
- Department of Bone and Joint Surgery, Peking
University Shenzhen Hospital, Shenzhen,
China
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials
Research, Shenzhen,
China
| | - Hui Zeng
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials
Research, Shenzhen,
China
| | - Jianjin Lin
- Department of Sport Medicine, Peking
University Shenzhen Hospital, Shenzhen,
China
| | - Peng Liu
- Department of Bone and Joint Surgery, Peking
University Shenzhen Hospital, Shenzhen,
China
- National and Local Joint Engineering Research Center of Orthopaedic
Biomaterials, Peking University Shenzhen
Hospital, Shenzhen,
China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials
Research, Shenzhen,
China
| |
Collapse
|
10
|
ZHU W, GUO C, DU M, MA Y, CUI Y, CHEN X, GUO C. Acupotomy alleviates knee osteoarthritis in rabbit by regulating chondrocyte mitophagy Pink1-Parkin pathway. J TRADIT CHIN MED 2024; 44:468-477. [PMID: 38767630 PMCID: PMC11077155 DOI: 10.19852/j.cnki.jtcm.20240402.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/19/2023] [Indexed: 05/22/2024]
Abstract
OBJECTIVE To investigate the effect of acupotomy, on mitophagy and the Pink1-Parkin pathway in chondrocytes from rabbits with knee osteoarthritis (KOA). METHODS A KOA model was established via the modified Videman method. Rabbits were randomly divided into a control group (CON), KOA group and KOA + acupotomy group (Acu). Rabbits in the acupotomy group were subjected to acupotomy for 4 weeks after model establishment. The behavior of the rabbits before and after intervention was recorded. Cartilage degeneration was evaluated by optical microscopy and fluorescence microscopy. The level of mitophagy was evaluated by transmission electron microscopy, immunofluorescence and enzyme-linked immunosorbent assay (ELISA). The expression of phosphatase and tensin homolog (PTEN)-induced kinase 1 (Pink1)-Parkin mitophagy pathway components was evaluated by immunofluorescence, Western blotting and real-time polymerase chain reaction. RESULTS In rabbits with KOA, joint pain, mobility disorders and cartilage degeneration were observed, the Mankin score was increased, collagen type Ⅱ (Col-Ⅱ) expression was significantly decreased, mitophagy was inhibited, mitochondrial function was impaired, and factors associated with the Pink1-Parkin pathway were inhibited. Acupotomy regulated the expression of Pink1-Parkin pathway-related proteins, the mitophagy-related protein microtubule-associated protein-1 light chain-3, the translocase of the outer membrane, and the inner mitochondrial membrane 23; increased the colocalization of mitochondria and autophagosomes; promoted the removal of damaged mitochondria; restored mitochondrial adenosine-triphosphate (ATP) production; and alleviated cartilage degeneration in rabbits with KOA. CONCLUSIONS Acupotomy played a role in alleviating KOA in rabbits by activating mitophagy in chondrocytes via the regulation of proteins that are related to the Pink1-Parkin pathway.
Collapse
Affiliation(s)
- Wenting ZHU
- 1 the Third Affiliated Hospital of Beijing University of Chinese Medicine, Beijing 100029, China
| | - Changqing GUO
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Mei DU
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yunxuan MA
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yongqi CUI
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xilin CHEN
- 2 School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Changqing GUO
- 3 Department of Medical Technology, Shijiazhuang Medical College, Hebei 050599, China
| |
Collapse
|
11
|
Liao Z, Cai X, Zheng Y, Lin J, Yang X, Lin W, Zhang Y, He X, Liu C. Sirtuin 1 in osteoarthritis: Perspectives on regulating glucose metabolism. Pharmacol Res 2024; 202:107141. [PMID: 38490314 DOI: 10.1016/j.phrs.2024.107141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/03/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024]
Abstract
Osteoarthritis (OA) is a degenerative disease characterised by articular cartilage destruction, and its complex aetiology contributes to suboptimal clinical treatment outcomes. A close association exists between glucose metabolism dysregulation and OA pathogenesis. Owing to the unique environment of low oxygen and glucose concentrations, chondrocytes rely heavily on their glycolytic capacity, exhibiting distinct spatiotemporal differences. However, under pathological stimulation, chondrocytes undergo excessive glycolytic activity while mitochondrial respiration and other branches of glucose metabolism are compromised. This metabolic change induces cartilage degeneration by reprogramming the inflammatory responses. Sirtuins, a highly conserved family of nicotinamide adenine dinucleotide (NAD+)-dependent deacetylases, regulate glucose metabolism in response to energy fluctuations in different cellular compartments,alleviating metabolic stress. SIRT1, the most extensively studied sirtuin, participates in maintaining glucose homeostasis in almost all key metabolic tissues. While actively contributing to the OA progression and displaying diverse biological effects in cartilage protection, SIRT1's role in regulating glucose metabolism in chondrocytes has not received sufficient attention. This review focuses on discussing the beneficial role of SIRT1 in OA progression from a metabolic regulation perspective based on elucidating the primary characteristics of chondrocyte glucose metabolism. We also summarise the potential mechanisms and therapeutic strategies targeting SIRT1 in chondrocytes to guide clinical practice and explore novel therapeutic directions.
Collapse
Affiliation(s)
- Zhihao Liao
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, 366, Jiangnan Avenue South, Guangzhou 510280, China
| | - Xuepei Cai
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yifan Zheng
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, 366, Jiangnan Avenue South, Guangzhou 510280, China
| | - Jiayu Lin
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, 366, Jiangnan Avenue South, Guangzhou 510280, China
| | - Xia Yang
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, 366, Jiangnan Avenue South, Guangzhou 510280, China
| | - Weiyin Lin
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, 366, Jiangnan Avenue South, Guangzhou 510280, China
| | - Ying Zhang
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, 366, Jiangnan Avenue South, Guangzhou 510280, China
| | - Xin He
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, 366, Jiangnan Avenue South, Guangzhou 510280, China
| | - Chufeng Liu
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, 366, Jiangnan Avenue South, Guangzhou 510280, China.
| |
Collapse
|
12
|
Lin S, Chen W, Alqahtani MS, Elkamchouchi DH, Ge Y, Lu Y, Zhang G, Wang M. Exploring the therapeutic potential of layered double hydroxides and transition metal dichalcogenides through the convergence of rheumatology and nanotechnology using generative adversarial network. ENVIRONMENTAL RESEARCH 2024; 241:117262. [PMID: 37839531 DOI: 10.1016/j.envres.2023.117262] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/10/2023] [Accepted: 09/27/2023] [Indexed: 10/17/2023]
Abstract
Two-dimensional Layered double hydroxides (LDHs) are highly used in the biomedical domain due to their biocompatibility, biodegradability, controlled drug loading and release capabilities, and improved cellular permeability. The interaction of LDHs with biological systems could facilitate targeted drug delivery and make them an attractive option for various biomedical applications. Rheumatoid Arthritis (RA) requires targeted drug delivery for optimum therapeutic outcomes. In this study, stacked double hydroxide nanocomposites with dextran sulphate modification (LDH-DS) were developed while exhibiting both targeting and pH-sensitivity for rheumatological conditions. This research examines the loading, release kinetics, and efficiency of the therapeutics of interest in the LDH-based drug delivery system. The mean size of LDH-DS particles (300.1 ± 8.12 nm) is -12.11 ± 0.4 mV. The encapsulation efficiency was 48.52%, and the loading efficacy was 16.81%. In vitro release tests indicate that the drug's discharge is modified more rapidly in PBS at pH 5.4 compared to pH 5.6, which later reached 7.3, showing the case sensitivity to pH. A generative adversarial network (GAN) is used to analyze the drug delivery system in rheumatology. The GAN model achieved high accuracy and classification rates of 99.3% and 99.0%, respectively, and a validity of 99.5%. The second and third administrations resulted in a significant change with p-values of 0.001 and 0.05, respectively. This investigation unequivocally demonstrated that LDH functions as a biocompatible drug delivery matrix, significantly improving delivery effectiveness.
Collapse
Affiliation(s)
- Suxian Lin
- Department of Rheumatology, Wenzhou People's Hospital, Wenzhou, 325000, China
| | - Weiwei Chen
- Department of Rheumatology, Wenzhou People's Hospital, Wenzhou, 325000, China
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, U.K
| | - Dalia H Elkamchouchi
- Department of Information Technology, College of Computer and Information Sciences, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Yisu Ge
- College of Computer Science and Artificial Intelligence, Wenzhou University, Wenzhou 325100, China
| | - Yanjie Lu
- Department of Digital Media Technology, Hangzhou Dianzi University, Hangzhou 310018, China
| | - Guodao Zhang
- Department of Digital Media Technology, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Mudan Wang
- Department of Nephrology, Wenzhou People's Hospital, Wenzhou, 325000, China.
| |
Collapse
|
13
|
Dilley J, Noori-Dokht H, Seetharam A, Bello M, Nanavaty A, Natoli RM, McKinley T, Bault Z, Wagner D, Sankar U. A Reproducible Cartilage Impact Model to Generate Post-Traumatic Osteoarthritis in the Rabbit. J Vis Exp 2023:10.3791/64450. [PMID: 38078617 PMCID: PMC11227251 DOI: 10.3791/64450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Post-traumatic osteoarthritis (PTOA) is responsible for 12% of all osteoarthritis cases in the United States. PTOA can be initiated by a single traumatic event, such as a high-impact load acting on articular cartilage, or by joint instability, as occurs with anterior cruciate ligament rupture. There are no effective therapeutics to prevent PTOA currently. Developing a reliable animal model of PTOA is necessary to better understand the mechanisms by which cartilage damage proceeds and to investigate novel treatment strategies to alleviate or prevent the progression of PTOA. This protocol describes an open, drop tower-based rabbit femoral condyle impact model to induce cartilage damage. This model delivered peak loads of 579.1 ± 71.1 N, and peak stresses of 81.9 ± 10.1 MPa with a time-to-peak load of 2.4 ± 0.5 ms. Articular cartilage from impacted medial femoral condyles (MFCs) had higher rates of apoptotic cells (p = 0.0058) and possessed higher Osteoarthritis Research Society International (OARSI) scores of 3.38 ± 1.43 compared to the non-impacted contralateral MFCs (0.56 ± 0.42), and other cartilage surfaces of the impacted knee (p < 0.0001). No differences in OARSI scores were detected among the non-impacted articular surfaces (p > 0.05).
Collapse
Affiliation(s)
- Julian Dilley
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine; Department of Orthopaedic Surgery, Indiana University School of Medicine; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine
| | - Hessam Noori-Dokht
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine; School of Mechanical Engineering, Purdue University; Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis
| | - Abhijit Seetharam
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine
| | - Margaret Bello
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine
| | - Aaron Nanavaty
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine
| | - Roman M Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine
| | - Todd McKinley
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine; Department of Orthopaedic Surgery, Indiana University School of Medicine; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine
| | - Zachary Bault
- Large Animal Resource Center, Indiana University School of Medicine
| | - Diane Wagner
- Department of Orthopaedic Surgery, Indiana University School of Medicine; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine; Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis;
| | - Uma Sankar
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine;
| |
Collapse
|
14
|
Riggs KC, Sankar U. Inflammatory mechanisms in post-traumatic osteoarthritis: a role for CaMKK2. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00031. [PMID: 37849987 PMCID: PMC10578519 DOI: 10.1097/in9.0000000000000031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 08/23/2023] [Indexed: 10/19/2023]
Abstract
Post-traumatic osteoarthritis (PTOA) is a multifactorial disease of the cartilage, synovium, and subchondral bone resulting from direct joint trauma and altered joint mechanics after traumatic injury. There are no current disease-modifying therapies for PTOA, and early surgical interventions focused on stabilizing the joint do not halt disease progression. Chronic pain and functional disability negatively affect the quality of life and take an economic toll on affected patients. While multiple mechanisms are at play in disease progression, joint inflammation is a key contributor. Impact-induced mitochondrial dysfunction and cell death or altered joint mechanics after trauma culminate in inflammatory cytokine release from synoviocytes and chondrocytes, cartilage catabolism, suppression of cartilage anabolism, synovitis, and subchondral bone disease, highlighting the complexity of the disease. Current understanding of the cellular and molecular mechanisms underlying the disease pathology has allowed for the investigation of a variety of therapeutic strategies that target unique apoptotic and/or inflammatory processes in the joint. This review provides a concise overview of the inflammatory and apoptotic mechanisms underlying PTOA pathogenesis and identifies potential therapeutic targets to mitigate disease progression. We highlight Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2), a serine/threonine protein kinase that was recently identified to play a role in murine and human osteoarthritis pathogenesis by coordinating chondrocyte inflammatory responses and apoptosis. Given its additional effects in regulating macrophage inflammatory signaling and bone remodeling, CaMKK2 emerges as a promising disease-modifying therapeutic target against PTOA.
Collapse
Affiliation(s)
- Keegan C. Riggs
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Uma Sankar
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
15
|
Paul S, Schrobback K, Tran PA, Meinert C, Davern JW, Weekes A, Nedunchezhiyan U, Klein TJ. GelMA-glycol chitosan hydrogels for cartilage regeneration: The role of uniaxial mechanical stimulation in enhancing mechanical, adhesive, and biochemical properties. APL Bioeng 2023; 7:036114. [PMID: 37692373 PMCID: PMC10492648 DOI: 10.1063/5.0160472] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Untreated osteochondral defects are a leading cause of osteoarthritis, a condition that places a heavy burden on both patients and orthopedic surgeons. Although tissue engineering has shown promise for creating mechanically similar cartilage-like constructs, their integration with cartilage remains elusive. Therefore, a formulation of biodegradable, biocompatible biomaterial with sufficient mechanical and adhesive properties for cartilage repair is required. To accomplish this, we prepared biocompatible, photo-curable, mechanically robust, and highly adhesive GelMA-glycol chitosan (GelMA-GC) hydrogels. GelMA-GC hydrogels had a modulus of 283 kPa and provided a biocompatible environment (>70% viability of embedded chondrocytes) in long-term culture within a bovine cartilage ring. The adhesive strength of bovine chondrocyte-laden GelMA-GC hydrogel to bovine cartilage increased from 38 to 52 kPa over four weeks of culture. Moreover, intermittent uniaxial mechanical stimulation enhanced the adhesive strength to ∼60 kPa, indicating that the cartilage-hydrogel integration could remain secure and functional under dynamic loading conditions. Furthermore, gene expression data and immunofluorescence staining revealed the capacity of chondrocytes in GelMA-GC hydrogel to synthesize chondrogenic markers (COL2A1 and ACAN), suggesting the potential for tissue regeneration. The promising in vitro results of this work motivate further exploration of the potential of photo-curable GelMA-GC bioadhesive hydrogels for cartilage repair and regeneration.
Collapse
Affiliation(s)
| | - Karsten Schrobback
- School of Biomedical Sciences, Centre for Genomics and Personalised Health, Translational Research Institute, Queensland University of Technology (QUT), 37 Kent Street, Woolloongabba, QLD 4102, Australia
| | | | | | | | | | | | | |
Collapse
|
16
|
Shultz SP, Buck AN, Fink PW, Kung SM, Ward MJ, Antal Z, Backus SI, Kraszewski AP, Hillstrom HJ. Body mass affects kinetic symmetry and inflammatory markers in adolescent knees during gait. Clin Biomech (Bristol, Avon) 2023; 102:105887. [PMID: 36657189 PMCID: PMC9975061 DOI: 10.1016/j.clinbiomech.2023.105887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/20/2022] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
BACKGROUND Early-onset osteoarthritis has been attributed to pro-inflammatory factors and biomechanical changes in obesity. However, research has yet to explore whether knee joint moments are asymmetrical in children with obesity and could precede the onset of knee osteoarthritis. The present study compares knee moment asymmetry between adolescents with and without obesity and examines the relationship between asymmetries and inflammatory biomarkers. METHODS Twenty-eight adolescents (13-16 years) were classified as with (n = 12) or without (n = 16) obesity. Lower extremity kinetics were measured using three-dimensional motion analysis. Bilateral knee joint moments were analyzed in the sagittal, frontal, and transverse planes across stance phase. Kinetic asymmetry was calculated between the right and left sides and represented by the R2 value. Enzyme-linked immunosorbent assays analyzed serum 25-hydroxy vitamin D, interferon gamma, tumor nercrosis factor alpha, interleukin-6, and C-reactive protein levels. Parametric and non-parametric tests determined significant group differences in asymmetries and biomarkers, respectively. Spearman's correlations identified relationships between biomarkers and asymmetries with statistically significant group differences. FINDINGS Adolescents with obesity had greater sagittal (loading, midstance) and frontal (midstance, pre-swing) plane kinetic knee asymmetry and higher concentrations of interleukin-6 and C-reactive protein. A moderately negative correlation existed between C-reactive protein and sagittal (loading, midstance) plane asymmetry, and also between interleukin-6 and frontal (pre-swing) plane asymmetry. INTERPRETATION Inflammatory response increases with greater knee joint asymmetry, suggesting knee joint damage and altered joint loading co-exist in adolescents with obesity. Increased risk to joint health may exist in sub-phases where knee joints are improperly loaded.
Collapse
Affiliation(s)
- Sarah P Shultz
- Kinesiology Department, Seattle University, Seattle, WA, USA.
| | - Ashley N Buck
- Kinesiology Department, Seattle University, Seattle, WA, USA.
| | - Philip W Fink
- STAPS, Université Sorbonne Paris Nord, Bobigny, France; Institut de Biomécanique Humaine Georges Charpak, Arts et Métiers ParisTech, Paris, France.
| | - Stacey M Kung
- Sports Medicine Department, Sports Surgery Clinic, Dublin, Ireland.
| | - Mary J Ward
- Weill Cornell Medicine, Department of Pediatrics, New York, NY, USA.
| | - Zoltan Antal
- Weill Cornell Medicine, Department of Pediatrics, New York, NY, USA.
| | - Sherry I Backus
- Leon Root, MD Motion Analysis Laboratory, Rehabilitation Department, Hospital for Special Surgery (HSS), New York, NY, USA.
| | - Andrew P Kraszewski
- Leon Root, MD Motion Analysis Laboratory, Rehabilitation Department, Hospital for Special Surgery (HSS), New York, NY, USA.
| | - Howard J Hillstrom
- Leon Root, MD Motion Analysis Laboratory, Rehabilitation Department, Hospital for Special Surgery (HSS), New York, NY, USA.
| |
Collapse
|
17
|
Dilley JE, Bello MA, Roman N, McKinley T, Sankar U. Post-traumatic osteoarthritis: A review of pathogenic mechanisms and novel targets for mitigation. Bone Rep 2023. [DOI: 10.1016/j.bonr.2023.101658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
18
|
Zhao Z, Ito A, Nakahata A, Ji X, Tai C, Saito M, Nishitani K, Aoyama T, Kuroki H. One session of 20 N cyclic compression induces chronic knee osteoarthritis in rats: A long-term study. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100325. [PMID: 36561496 PMCID: PMC9763514 DOI: 10.1016/j.ocarto.2022.100325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/15/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Objective Mechanical stimulation is a risk factor for knee osteoarthritis. Non-surgical compression has been used to study the effects of mechanical stimulation in vivo. However, the long-term effects of low-force compression on knee joint had not been studied. Therefore, we sought to identify the long-term effects of low-force cyclic compression on the rat knee joint. Design In this study, we applied one session cyclic compression with a peak load of 20 N for 60 cycles to the rat knee joint in an approximately 140-degree flexion position (Wistar, male, 12 weeks old), followed by 1 year of observation (including data from 1 week, 2 weeks, 4 weeks, 8 weeks, 6 months, and 1 year after compression), and then performed a sub-regional analysis with hematoxylin-eosin, Safranin O and Fast Green, and MMP13 immunohistochemical staining. Results We observed osteoarthritis-like cartilage damage, synovial inflammation, and high expression of MMP13 within 1 year after compression. However, these changes progressed slowly, with obvious matrix cracks that did not appear until 1 year after compression. In the regional analysis, we found that low-force compression caused a much slower development of injury at the compression contact site, and no significant structural cartilage damage was observed after 1 year of compression. In contrast, the non-contact site during compression at tibial cartilage in the same joint was the first to show significant structural damage. Conclusion This study demonstrates that one session of 20 N cyclic compression induces a chronic osteoarthritis-like phenotype in the rat knee in the long term.
Collapse
Affiliation(s)
- Zixi Zhao
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan,Corresponding author.
| | - Akihiro Nakahata
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Xiang Ji
- Department of Motor Function Development, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chia Tai
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motoo Saito
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kohei Nishitani
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Kuroki
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
19
|
Hines MR, Goetz JE, Gomez-Contreras PC, Rodman SN, Liman S, Femino EL, Kluz PN, Wagner BA, Buettner GR, Kelley EE, Coleman MC. Extracellular biomolecular free radical formation during injury. Free Radic Biol Med 2022; 188:175-184. [PMID: 35724853 PMCID: PMC9725094 DOI: 10.1016/j.freeradbiomed.2022.06.223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/15/2022] [Accepted: 06/11/2022] [Indexed: 11/30/2022]
Abstract
Determine if oxidative damage increases in articular cartilage as a result of injury and matrix failure and whether modulation of the local redox environment influences this damage. Osteoarthritis is an age associated disease with no current disease modifying approaches available. Mechanisms of cartilage damage in vitro suggest tissue free radical production could be critical to early degeneration, but these mechanisms have not been described in intact tissue. To assess free radical production as a result of traumatic injury, we measured biomolecular free radical generation via immuno-spin trapping (IST) of protein/proteoglycan/lipid free radicals after a 2 J/cm2 impact to swine articular cartilage explants. This technique allows visualization of free radical formation upon a wide variety of molecules using formalin-fixed, paraffin-embedded approaches. Scoring of extracellular staining by trained, blinded scorers demonstrated significant increases with impact injury, particularly at sites of cartilage cracking. Increases remain in the absence of live chondrocytes but are diminished; thus, they appear to be a cell-dependent and -independent feature of injury. We then modulated the extracellular environment with a pulse of heparin to demonstrate the responsiveness of the IST signal to changes in cartilage biology. Addition of heparin caused a distinct change in the distribution of protein/lipid free radicals at sites of failure alongside a variety of pertinent redox changes related to osteoarthritis. This study directly confirms the production of biomolecular free radicals from articular trauma, providing a rigorous characterization of their formation by injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Paige N Kluz
- University of Wisconsin-Madison, Madison, WI, USA
| | | | | | | | | |
Collapse
|
20
|
Walsh SK, Soni R, Arendt LM, Skala MC, Henak CR. Maturation- and degeneration-dependent articular cartilage metabolism via optical redox ratio imaging. J Orthop Res 2022; 40:1735-1743. [PMID: 34792214 DOI: 10.1002/jor.25214] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/24/2021] [Accepted: 11/09/2021] [Indexed: 02/04/2023]
Abstract
From the two metabolic processes in healthy cartilage, glycolysis has been associated with proliferation and oxidative phosphorylation (oxphos) with matrix synthesis. Recently, metabolic dysregulation was significantly correlated with cartilage degradation and osteoarthritis progression. While these findings suggest maturation predisposes cartilage to metabolic instability with consequences for tissue maintenance, these links have not been shown. Therefore, this study sought to address three hypotheses (a) chondrocytes exhibit differential metabolic activity between immaturity (0-4 months), adolescence (5-18 months), and maturity (>18 months); (b) perturbation of metabolic activity has consequences on expression of genes pertinent to cartilage tissue maintenance; and (c) severity of cartilage damage is positively correlated with glycolysis and oxphos activity as well as optical redox ratio in postadolescent cartilage. Porcine femoral cartilage samples from pigs (3 days to 6 years) underwent optical redox ratio imaging, which measures autofluorescence of NAD(P)H and FAD. Gene expression analysis and histological scoring was conducted for comparison against imaging metrics. NAD(P)H and FAD autofluorescence both demonstrated increasing intensity with age, while optical redox ratio was lowest in adolescent samples compared to immature or mature samples. Inhibition of glycolysis suppressed expression of Col2, Col1, ADAMTS4, and ADAMTS5, while oxphos inhibition had no effect. FAD fluorescence and optical redox ratio were positively correlated with histological degeneration. This study demonstrates maturation- and degeneration-dependent metabolic activity in cartilage and explores the consequences of this differential activity on gene expression. This study aids our basic understanding of cartilage biology and highlights opportunity for potential diagnostic applications.
Collapse
Affiliation(s)
- Shannon K Walsh
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Rikin Soni
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lisa M Arendt
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Melissa C Skala
- Morgridge Institute for Research, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Corinne R Henak
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
21
|
The Role of Mitochondrial Metabolism, AMPK-SIRT Mediated Pathway, LncRNA and MicroRNA in Osteoarthritis. Biomedicines 2022; 10:biomedicines10071477. [PMID: 35884782 PMCID: PMC9312479 DOI: 10.3390/biomedicines10071477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/20/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease characterized by degeneration of articular cartilage and causes severe joint pain, physical disability, and impaired quality of life. Recently, it was found that mitochondria not only act as a powerhouse of cells that provide energy for cellular metabolism, but are also involved in crucial pathways responsible for maintaining chondrocyte physiology. Therefore, a growing amount of evidence emphasizes that impairment of mitochondrial function is associated with OA pathogenesis; however, the exact mechanism is not well known. Moreover, the AMP-activated protein kinase (AMPK)–Sirtuin (SIRT) signaling pathway, long non-coding RNA (lncRNA), and microRNA (miRNA) are important for regulating the physiological and pathological processes of chondrocytes, indicating that these may be targets for OA treatment. In this review, we first focus on the importance of mitochondria metabolic dysregulation related to OA. Then, we show recent evidence on the AMPK-SIRT mediated pathway associated with OA pathogenesis and potential treatment options. Finally, we discuss current research into the effects of lncRNA and miRNA on OA progression or inhibition.
Collapse
|
22
|
Yang L, Brouillette MJ, Coleman MC, Kluz PN, Goetz JE. Automated quantification of live articular chondrocyte fluorescent staining using a custom image analysis framework. J Orthop Res 2022; 40:1203-1212. [PMID: 34191348 DOI: 10.1002/jor.25137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 05/03/2021] [Accepted: 06/09/2021] [Indexed: 02/04/2023]
Abstract
The goal of this study was to develop, validate, and implement an image analysis framework to automatically analyze chondrocytes in 3D image stacks of cartilage acquired using a fluorescent confocal microscope. Source specimens consist of viable osteochondral tissue co-stained with multiple live-cell dyes. Our framework utilizes a seeded watershed-based algorithm to automatically segment individual chondrocytes in each 2D slice of the confocal image stack. The resulting cell segmentations are colocalized in 3D to eliminate duplicate segmentation of the same cell resulting from the visibility of fluorescence signal in multiple imaging planes, and the 3D cell distribution is used to automatically define the cartilage tissue volume. The algorithm then provides chondrocyte density data, and the associated segmentation can be used as a mask to extract and quantify per cell intensity of a secondary, functional dye co-staining the chondrocytes. The accuracy of the automated chondrocyte segmentation was validated against manual segmentations (average IOU = 0.79). When applied to a cartilage surrogate, this analysis framework estimated chondrocyte density within 10% of the true density and demonstrated a good agreement between framework's counts and manual counts (R2 = 0.99). In a real application, the framework was able to detect the increased dye signal of monochlorobimane (MCB) in chondrocytes treated with N-acetylcysteine (NAC) after mechanical injury, quantifying intracellular biochemical changes in living cells. This new framework allows for fast and accurate quantification of intracellular activities of chondrocytes, and it can be adapted for broader application in many imaging and treatment modalities, including therapeutic OA research.
Collapse
Affiliation(s)
- Linjun Yang
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, Iowa, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
| | - Marc J Brouillette
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, Iowa, USA
| | - Mitchell C Coleman
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, Iowa, USA.,Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Paige N Kluz
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, Iowa, USA
| | - Jessica E Goetz
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, Iowa, USA.,Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
23
|
Batushansky A, Zhu S, Komaravolu RK, South S, Mehta-D'souza P, Griffin TM. Fundamentals of OA. An initiative of Osteoarthritis and Cartilage. Obesity and metabolic factors in OA. Osteoarthritis Cartilage 2022; 30:501-515. [PMID: 34537381 PMCID: PMC8926936 DOI: 10.1016/j.joca.2021.06.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/14/2021] [Accepted: 06/07/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Obesity was once considered a risk factor for knee osteoarthritis (OA) primarily for biomechanical reasons. Here we provide an additional perspective by discussing how obesity also increases OA risk by altering metabolism and inflammation. DESIGN This narrative review is presented in four sections: 1) metabolic syndrome and OA, 2) metabolic biomarkers of OA, 3) evidence for dysregulated chondrocyte metabolism in OA, and 4) metabolic inflammation: joint tissue mediators and mechanisms. RESULTS Metabolic syndrome and its components are strongly associated with OA. However, evidence for a causal relationship is context dependent, varying by joint, gender, diagnostic criteria, and demographics, with additional environmental and genetic interactions yet to be fully defined. Importantly, some aspects of the etiology of obesity-induced OA appear to be distinct between men and women, especially regarding the role of adipose tissue. Metabolomic analyses of serum and synovial fluid have identified potential diagnostic biomarkers of knee OA and prognostic biomarkers of disease progression. Connecting these biomarkers to cellular pathophysiology will require future in vivo studies of joint tissue metabolism. Such studies will help reveal when a metabolic process or a metabolite itself is a causal factor in disease progression. Current evidence points towards impaired chondrocyte metabolic homeostasis and metabolic-immune dysregulation as likely factors connecting obesity to the increased risk of OA. CONCLUSIONS A deeper understanding of how obesity alters metabolic and inflammatory pathways in synovial joint tissues is expected to provide new therapeutic targets and an improved definition of "metabolic" and "obesity" OA phenotypes.
Collapse
Affiliation(s)
- A Batushansky
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - S Zhu
- Department of Biomedical Sciences, Ohio Musculoskeletal and Neurological Institute (OMNI), Ohio University, Athens, OH, 45701, USA.
| | - R K Komaravolu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - S South
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - P Mehta-D'souza
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - T M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA; Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
24
|
Zhang J, Hao X, Chi R, Liu J, Shang X, Deng X, Qi J, Xu T. Whole Transcriptome Mapping Identifies an Immune- and Metabolism-Related Non-coding RNA Landscape Remodeled by Mechanical Stress in IL-1β-Induced Rat OA-like Chondrocytes. Front Genet 2022; 13:821508. [PMID: 35309149 PMCID: PMC8927047 DOI: 10.3389/fgene.2022.821508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/17/2022] [Indexed: 12/21/2022] Open
Abstract
Background: Osteoarthritis (OA) is a common degenerative joint disease. The aims of this study are to explore the effects of mechanical stress on whole transcriptome landscape and to identify a non-coding transcriptome signature of mechanical stress. Methods: Next-generation RNA sequencing (RNA-seq) was performed on IL-1β-induced OA-like chondrocytes stimulated by mechanical stress. Integrated bioinformatics analysis was performed and further verified by experimental validations. Results: A total of 5,022 differentially expressed mRNAs (DEMs), 88 differentially expressed miRNAs (DEMIs), 1,259 differentially expressed lncRNAs (DELs), and 393 differentially expressed circRNAs (DECs) were identified as the transcriptome response to mechanical stress. The functional annotation of the DEMs revealed the effects of mechanical stress on chondrocyte biology, ranging from cell fate, metabolism, and motility to endocrine, immune response, and signaling transduction. Among the DELs, ∼92.6% were identified as the novel lncRNAs. According to the co-expressing DEMs potentially regulated by the responsive DELs, we found that these DELs were involved in the modification of immune and metabolism. Moreover, immune- and metabolism-relevant DELs exhibited a notable involvement in the competing endogenous RNA (ceRNA) regulation networks. Silencing lncRNA TCONS_00029778 attenuated cellular senescence induced by mechanical stress. Moreover, the expression of Cd80 was elevated by mechanical stress, which was rescued by silencing TCONS_00029778. Conclusion: The transcriptome landscape of IL-1β-induced OA-like chondrocytes was remarkably remodeled by mechanical stress. This study identified an immune- and metabolism-related ncRNA transcriptome signature responsive to mechanical stress and provides an insight of ncRNAs into chondrocyte biology and OA.
Collapse
Affiliation(s)
- Jiaming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruimin Chi
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingru Shang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofeng Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Qi
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jun Qi, ; Tao Xu,
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jun Qi, ; Tao Xu,
| |
Collapse
|
25
|
Hodgkinson T, Amado IN, O'Brien FJ, Kennedy OD. The role of mechanobiology in bone and cartilage model systems in characterizing initiation and progression of osteoarthritis. APL Bioeng 2022. [DOI: 10.1063/5.0068277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Tom Hodgkinson
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Isabel N. Amado
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fergal J. O'Brien
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| | - Oran D. Kennedy
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials Bio-Engineering Research Centre (AMBER), Dublin, Ireland
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
26
|
Zhang M, Hu W, Cai C, Wu Y, Li J, Dong S. Advanced application of stimuli-responsive drug delivery system for inflammatory arthritis treatment. Mater Today Bio 2022; 14:100223. [PMID: 35243298 PMCID: PMC8881671 DOI: 10.1016/j.mtbio.2022.100223] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
|
27
|
Gonzalez-Fernandez P, Rodríguez-Nogales C, Jordan O, Allémann E. Combination of mesenchymal stem cells and bioactive molecules in hydrogels for osteoarthritis treatment. Eur J Pharm Biopharm 2022; 172:41-52. [PMID: 35114357 DOI: 10.1016/j.ejpb.2022.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/13/2021] [Accepted: 01/17/2022] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) is a chronic and inflammatory disease with no effective regenerative treatments to date. The therapeutic potential of mesenchymal stem cells (MSCs) remains to be fully explored. Intra-articular injection of these cells promotes cartilage protection and regeneration by paracrine signaling and differentiation into chondrocytes. However, joints display a harsh avascular environment for these cells upon injection. This phenomenon prompted researchers to develop suitable injectable materials or systems for MSCs to enhance their function and survival. Among them, hydrogels can absorb a large amount of water and maintain their 3D structure but also allow incorporation of bioactive agents or small molecules in their matrix that maximize the action of MSCs. These materials possess advantageous cartilage-like features such as collagen or hyaluronic acid moieties that interact with MSC receptors, thereby promoting cell adhesion. This review provides an up-to-date overview of the progress and opportunities of MSCs entrapped into hydrogels, combined with bioactive/small molecules to improve the therapeutic effects in OA treatment.
Collapse
Affiliation(s)
- P Gonzalez-Fernandez
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - C Rodríguez-Nogales
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - O Jordan
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland
| | - E Allémann
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, Rue Michel-Servet 1, 1211 Geneva 4, Switzerland.
| |
Collapse
|
28
|
Liu Y, Shah KM, Luo J. Strategies for Articular Cartilage Repair and Regeneration. Front Bioeng Biotechnol 2022; 9:770655. [PMID: 34976967 PMCID: PMC8719005 DOI: 10.3389/fbioe.2021.770655] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/01/2021] [Indexed: 12/19/2022] Open
Abstract
Articular cartilage is an avascular tissue, with limited ability to repair and self-renew. Defects in articular cartilage can induce debilitating degenerative joint diseases such as osteoarthritis. Currently, clinical treatments have limited ability to repair, for they often result in the formation of mechanically inferior cartilage. In this review, we discuss the factors that affect cartilage homeostasis and function, and describe the emerging regenerative approaches that are informing the future treatment options.
Collapse
Affiliation(s)
- Yanxi Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Karan M Shah
- Department of Oncology and Metabolism, The Medical School, The University of Sheffield, Sheffield, United Kingdom
| | - Jian Luo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.,Shanghai Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Centre), Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Wang S, Li W, Zhang P, Wang Z, Ma X, Liu C, Vasilev K, Zhang L, Zhou X, Liu L, Hayball J, Dong S, Li Y, Gao Y, Cheng L, Zhao Y. Mechanical overloading induces GPX4-regulated chondrocyte ferroptosis in osteoarthritis via Piezo1 channel facilitated calcium influx. J Adv Res 2022; 41:63-75. [PMID: 36328754 PMCID: PMC9637484 DOI: 10.1016/j.jare.2022.01.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 12/19/2021] [Accepted: 01/07/2022] [Indexed: 11/17/2022] Open
Abstract
Our study proved that mechanical overloading induces ferroptosis of chondrocyte, which might be a potential therapeutic target for mechanical damage of chondrocyte and OA. Our study demonstrated Piezo1 facilitated calcium influx leads to reduction of GSH, decrease of Gpx4 and activation of oxidative stress in chondrocyte under high strain mechanical stimulation. Mechanical signals were converted into ferroptosis-associated signals through Piezo1 channel induced calcium influx, which might shed light on therapeutic interventions for treatment of OA and other diseases associated with ferroptosis.
Introductions Excessive mechanical stress is closely associated with cell death in various conditions. Exposure of chondrocytes to excessive mechanical loading leads to a catabolic response as well as exaggerated cell death. Ferroptosis is a recently identified form of cell death during cell aging and degeneration. However, it's potential association with mechanical stress remains to be illustrated. Objectives To identify whether excessive mechanical stress can cause ferroptosis. To explore the role of mechanical overloading in chondrocyte ferroptosis. Methods Chondrocytes were collected from loading and unloading zones of cartilage in patients with osteoarthritis (OA), and the ferroptosis phenotype was analyzed through transmission electron microscope and microarray. Moreover, the relationship between ferroptosis and OA was analyzed by GPX4-conditional knockout (Col2a1-CreERT: GPX4flox/flox) mice OA model and chondrocytes cultured with high strain mechanical stress. Furthermore, the role of Piezo1 ion channel in chondrocyte ferroptosis and OA development was explored by using its inhibitor (GsMTx4) and agonist (Yoda1). Additionally, chondrocyte was cultured in calcium-free medium with mechanical stress, and ferroptosis phenotype was tested. Results Human cartilage and mouse chondrocyte experiments revealed that mechanical overloading can induce GPX4-associated ferroptosis. Conditional knockout of GPX4 in cartilage aggravated experimental OA process, while additional treatment with ferroptosis suppressor protein (FSP-1) and coenzyme Q10 (CoQ10) abated OA development in GPX4-CKO mice. In mouse OA model and chondrocyte experiments, inhibition of Piezo1 channel activity increased GPX4 expression, attenuated ferroptosis phenotype and reduced the severity of osteoarthritis. Additionally, high strain mechanical stress induced ferroptosis damage in chondrocyte was largely abolished by blocking calcium influx through calcium-free medium. Conclusions Our findings show that mechanical overloading induces ferroptosis through Piezo1 activation and subsequent calcium influx in chondrocytes, which might provide a potential target for OA treatment.
Collapse
|
30
|
Zhang K, Wang L, Liu Z, Geng B, Teng Y, Liu X, Yi Q, Yu D, Chen X, Zhao D, Xia Y. Mechanosensory and mechanotransductive processes mediated by ion channels in articular chondrocytes: Potential therapeutic targets for osteoarthritis. Channels (Austin) 2021; 15:339-359. [PMID: 33775217 PMCID: PMC8018402 DOI: 10.1080/19336950.2021.1903184] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Articular cartilage consists of an extracellular matrix including many proteins as well as embedded chondrocytes. Articular cartilage formation and function are influenced by mechanical forces. Hind limb unloading or simulated microgravity causes articular cartilage loss, suggesting the importance of the healthy mechanical environment in articular cartilage homeostasis and implying a significant role of appropriate mechanical stimulation in articular cartilage degeneration. Mechanosensitive ion channels participate in regulating the metabolism of articular chondrocytes, including matrix protein production and extracellular matrix synthesis. Mechanical stimuli, including fluid shear stress, stretch, compression and cell swelling and decreased mechanical conditions (such as simulated microgravity) can alter the membrane potential and regulate the metabolism of articular chondrocytes via transmembrane ion channel-induced ionic fluxes. This process includes Ca2+ influx and the resulting mobilization of Ca2+ that is due to massive released Ca2+ from stores, intracellular cation efflux and extracellular cation influx. This review brings together published information on mechanosensitive ion channels, such as stretch-activated channels (SACs), voltage-gated Ca2+ channels (VGCCs), large conductance Ca2+-activated K+ channels (BKCa channels), Ca2+-activated K+ channels (SKCa channels), voltage-activated H+ channels (VAHCs), acid sensing ion channels (ASICs), transient receptor potential (TRP) family channels, and piezo1/2 channels. Data based on epithelial sodium channels (ENaCs), purinergic receptors and N-methyl-d-aspartate (NMDA) receptors are also included. These channels mediate mechanoelectrical physiological processes essential for converting physical force signals into biological signals. The primary channel-mediated effects and signaling pathways regulated by these mechanosensitive ion channels can influence the progression of osteoarthritis during the mechanosensory and mechanoadaptive process of articular chondrocytes.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Lifu Wang
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Zhongcheng Liu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Bin Geng
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Yuanjun Teng
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Xuening Liu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Qiong Yi
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Dechen Yu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Xiangyi Chen
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Dacheng Zhao
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Yayi Xia
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| |
Collapse
|
31
|
Zhang J, Hao X, Chi R, Qi J, Xu T. Moderate mechanical stress suppresses the IL-1β-induced chondrocyte apoptosis by regulating mitochondrial dynamics. J Cell Physiol 2021; 236:7504-7515. [PMID: 33821497 DOI: 10.1002/jcp.30386] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 12/17/2022]
Abstract
Mitochondrial dysfunction contributes to osteoarthritis (OA) onset and progress. Mitochondrial dynamics, coupled with mitophagy, is critical for the maintenance of mitochondrial fitness, involving many cellular processes, such as proliferation and apoptosis. Excessive mechanical stress induces chondrocyte apoptosis; however, the effects of mechanical stress on mitochondrial dynamics remain elusive. In this study, we performed fluorescence staining, flow cytometry, transmission electron microscope, Western blot analysis, and RNA-sequencing to assess the effects of different strength of mechanical stimulation on mitochondrial functions of chondrocyte treated with interleukin-1β (IL-1β). We found that moderate mechanical stress reduced the IL-1β-induced apoptosis by maintaining mitochondrial function and scavenging the reactive oxygen species, while excessive mechanical stress induced strong mitochondrial dysfunction and apoptosis. Moreover, RNAsequencing revealed that mitophagy and mitochondrial dynamics were involved in the regulation of mechanical stress on chondrocyte biology. In addition to the elevated mitophagy, moderate mechanical stress also promoted mitochondrial dynamics by enhancing the expression of MFN1/2 and OPA1 and the translocation of dynamin-related protein 1 from the cytoplasm to the mitochondria. However, an uncoupling of mitochondrial dynamics, characterized by strongly elevated fission, resulted in the unfavorable apoptosis of excessive mechanical stress-stimulated chondrocytes. This study revealed the effects of mechanical stress upon mitochondrial dynamics in chondrocyte.
Collapse
Affiliation(s)
- Jiaming Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruimin Chi
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Qi
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
32
|
Early JO, Fagan LE, Curtis AM, Kennedy OD. Mitochondria in Injury, Inflammation and Disease of Articular Skeletal Joints. Front Immunol 2021; 12:695257. [PMID: 34539627 PMCID: PMC8448207 DOI: 10.3389/fimmu.2021.695257] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammation is an important biological response to tissue damage caused by injury, with a crucial role in initiating and controlling the healing process. However, dysregulation of the process can also be a major contributor to tissue damage. Related to this, although mitochondria are typically thought of in terms of energy production, it has recently become clear that these important organelles also orchestrate the inflammatory response via multiple mechanisms. Dysregulated inflammation is a well-recognised problem in skeletal joint diseases, such as rheumatoid arthritis. Interestingly osteoarthritis (OA), despite traditionally being known as a ‘non-inflammatory arthritis’, now appears to involve an element of chronic inflammation. OA is considered an umbrella term for a family of diseases stemming from a range of aetiologies (age, obesity etc.), but all with a common presentation. One particular OA sub-set called Post-Traumatic OA (PTOA) results from acute mechanical injury to the joint. Whether the initial mechanical tissue damage, or the subsequent inflammatory response drives disease, is currently unclear. In the former case; mechanobiological properties of cells/tissues in the joint are a crucial consideration. Many such cell-types have been shown to be exquisitely sensitive to their mechanical environment, which can alter their mitochondrial and cellular function. For example, in bone and cartilage cells fluid-flow induced shear stresses can modulate cytoskeletal dynamics and gene expression profiles. More recently, immune cells were shown to be highly sensitive to hydrostatic pressure. In each of these cases mitochondria were central to these responses. In terms of acute inflammation, mitochondria may have a pivotal role in linking joint tissue injury with chronic disease. These processes could involve the immune cells recruited to the joint, native/resident joint cells that have been damaged, or both. Taken together, these observations suggest that mitochondria are likely to play an important role in linking acute joint tissue injury, inflammation, and long-term chronic joint degeneration - and that the process involves mechanobiological factors. In this review, we will explore the links between mechanobiology, mitochondrial function, inflammation/tissue-damage in joint injury and disease. We will also explore some emerging mitochondrial therapeutics and their potential for application in PTOA.
Collapse
Affiliation(s)
- James Orman Early
- Department of Anatomy and Regenerative Medicine and Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Lauren E Fagan
- Department of Anatomy and Regenerative Medicine and Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,School of Pharmacy and Biomolecular Sciences and Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Annie M Curtis
- School of Pharmacy and Biomolecular Sciences and Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Oran D Kennedy
- Department of Anatomy and Regenerative Medicine and Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Mechanical and Manufacturing Engineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
33
|
He Y, Yocum L, Alexander PG, Jurczak MJ, Lin H. Urolithin A Protects Chondrocytes From Mechanical Overloading-Induced Injuries. Front Pharmacol 2021; 12:703847. [PMID: 34220525 PMCID: PMC8245698 DOI: 10.3389/fphar.2021.703847] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/04/2021] [Indexed: 01/10/2023] Open
Abstract
Physiological mechanical stimulation has been shown to promote chondrogenesis, but excessive mechanical loading results in cartilage degradation. Currently, the underlying mechanotransduction pathways in the context of physiological and injurious loading are not fully understood. In this study, we aim to identify the critical factors that dictate chondrocyte response to mechanical overloading, as well as to develop therapeutics that protect chondrocytes from mechanical injuries. Specifically, human chondrocytes were loaded in hyaluronic hydrogel and then subjected to dynamic compressive loading under 5% (DL-5% group) or 25% strain (DL-25% group). Compared to static culture and DL-5%, DL-25% reduced cartilage matrix formation from chondrocytes, which was accompanied by the increased senescence level, as revealed by higher expression of p21, p53, and senescence-associated beta-galactosidase (SA-β-Gal). Interestingly, mitophagy was suppressed by DL-25%, suggesting a possible role for the restoration mitophagy in reducing cartilage degeneration with mechanical overloading. Next, we treated the mechanically overloaded samples (DL-25%) with Urolithin A (UA), a natural metabolite previously shown to enhance mitophagy in other cell types. qRT-PCR, histology, and immunostaining results confirmed that UA treatment significantly increased the quantity and quality of cartilage matrix deposition. Interestingly, UA also suppressed the senescence level induced by mechanical overloading, demonstrating its senomorphic potential. Mechanistic analysis confirmed that UA functioned partially by enhancing mitophagy. In summary, our results show that mechanical overloading results in cartilage degradation partially through the impairment of mitophagy. This study also identifies UA's novel use as a compound that can protect chondrocytes from mechanical injuries, supporting high-quality cartilage formation/maintenance.
Collapse
Affiliation(s)
- Yuchen He
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Lauren Yocum
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Peter G Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
34
|
Statham P, Jones E, Jennings LM, Fermor HL. Reproducing the Biomechanical Environment of the Chondrocyte for Cartilage Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:405-420. [PMID: 33726527 DOI: 10.1089/ten.teb.2020.0373] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
It is well known that the biomechanical and tribological performance of articular cartilage is inextricably linked to its extracellular matrix (ECM) structure and zonal heterogeneity. Furthermore, it is understood that the presence of native ECM components, such as collagen II and aggrecan, promote healthy homeostasis in the resident chondrocytes. What is less frequently discussed is how chondrocyte metabolism is related to the extracellular mechanical environment, at both the macro and microscale. The chondrocyte is in immediate contact with the pericellular matrix of the chondron, which acts as a mechanocoupler, transmitting external applied loads from the ECM to the chondrocyte. Therefore, components of the pericellular matrix also play essential roles in chondrocyte mechanotransduction and metabolism. Recreating the biomechanical environment through tuning material properties of a scaffold and/or the use of external cyclic loading can induce biosynthetic responses in chondrocytes. Decellularized scaffolds, which retain the native tissue macro- and microstructure also represent an effective means of recapitulating such an environment. The use of such techniques in tissue engineering applications can ensure the regeneration of skeletally mature articular cartilage with appropriate biomechanical and tribological properties to restore joint function. Despite the pivotal role in graft maturation and performance, biomechanical and tribological properties of such interventions is often underrepresented. This review outlines the role of biomechanics in relation to native cartilage performance and chondrocyte metabolism, and how application of this theory can enhance the future development and successful translation of biomechanically relevant tissue engineering interventions. Impact statement Physiological cartilage function is a key criterion in the success of a cartilage tissue engineering solution. The in situ performance is dependent on the initial scaffold design as well as extracellular matrix deposition by endogenous or exogenous cells. Both biological and biomechanical stimuli serve as key regulators of cartilage homeostasis and maturation of the resulting tissue-engineered graft. An improved understanding of the influence of biomechanics on cellular function and consideration of the final biomechanical and tribological performance will help in the successful development and translation of tissue-engineered grafts to restore natural joint function postcartilage trauma or osteoarthritic degeneration, delaying the requirement for prosthetic intervention.
Collapse
Affiliation(s)
- Patrick Statham
- Institute of Medical and Biological Engineering, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds, United Kingdom
| | - Louise M Jennings
- Institute of Medical and Biological Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, United Kingdom
| | - Hazel L Fermor
- Institute of Medical and Biological Engineering, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
35
|
Jiang W, Liu H, Wan R, Wu Y, Shi Z, Huang W. Mechanisms linking mitochondrial mechanotransduction and chondrocyte biology in the pathogenesis of osteoarthritis. Ageing Res Rev 2021; 67:101315. [PMID: 33684550 DOI: 10.1016/j.arr.2021.101315] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 02/12/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022]
Abstract
Mechanical loading is essential for chondrocyte health. Chondrocytes can sense and respond to various extracellular mechanical signals through an integrated set of mechanisms. Recently, it has been found that mitochondria, acting as critical mechanotransducers, are at the intersection between extracellular mechanical signals and chondrocyte biology. Much attention has been focused on identifying how mechanical loading-induced mitochondrial dysfunction contributes to the pathogenesis of osteoarthritis. In contrast, little is known regarding the mechanisms underlying functional alterations in mitochondria induced by mechanical stimulation. In this review, we describe how chondrocytes perceive environmental mechanical signals. We discuss how mechanical load induces mitochondrial functional alterations and highlight the major unanswered questions in this field. We speculate that AMP-activated protein kinase (AMPK), a master regulator of energy homeostasis, may play an important role in coupling force transmission to mitochondrial health and intracellular biological responses.
Collapse
|
36
|
Gomez-Contreras PC, Kluz PN, Hines MR, Coleman MC. Intersections Between Mitochondrial Metabolism and Redox Biology Mediate Posttraumatic Osteoarthritis. Curr Rheumatol Rep 2021; 23:32. [PMID: 33893892 DOI: 10.1007/s11926-021-00994-z] [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] [Accepted: 02/23/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW This review will cover foundational studies and recent findings that established key concepts for understanding the importance of redox biology to chondrocyte mitochondrial function and osteoarthritis pathophysiology after injury. RECENT FINDINGS Articular chondrocyte mitochondria can be protected with a wide variety of antioxidants that will be discussed within a framework suggested by classic studies. These agents not only underscore the importance of thiol metabolism and associated redox function for chondrocyte mitochondria but also suggest complex interactions with signal transduction pathways and other molecular features of osteoarthritis that require more thorough investigation. Emerging evidence also indicates that reductive stress could occur alongside oxidative stress. Recent studies have shed new light on historic paradoxes in chondrocyte redox and mitochondrial physiology, leading to the development of promising disease-modifying therapies for posttraumatic osteoarthritis.
Collapse
Affiliation(s)
| | - Paige N Kluz
- University of Iowa, 1182 Biomedical Laboratories, 500 Newton Road, Iowa City, 52242, USA
| | - Madeline R Hines
- University of Iowa, 1182 Biomedical Laboratories, 500 Newton Road, Iowa City, 52242, USA
| | - Mitchell C Coleman
- University of Iowa, 1182 Biomedical Laboratories, 500 Newton Road, Iowa City, 52242, USA.
| |
Collapse
|
37
|
Zhen G, Guo Q, Li Y, Wu C, Zhu S, Wang R, Guo XE, Kim BC, Huang J, Hu Y, Dan Y, Wan M, Ha T, An S, Cao X. Mechanical stress determines the configuration of TGFβ activation in articular cartilage. Nat Commun 2021; 12:1706. [PMID: 33731712 PMCID: PMC7969741 DOI: 10.1038/s41467-021-21948-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/19/2021] [Indexed: 01/18/2023] Open
Abstract
Our incomplete understanding of osteoarthritis (OA) pathogenesis has significantly hindered the development of disease-modifying therapy. The functional relationship between subchondral bone (SB) and articular cartilage (AC) is unclear. Here, we found that the changes of SB architecture altered the distribution of mechanical stress on AC. Importantly, the latter is well aligned with the pattern of transforming growth factor beta (TGFβ) activity in AC, which is essential in the regulation of AC homeostasis. Specifically, TGFβ activity is concentrated in the areas of AC with high mechanical stress. A high level of TGFβ disrupts the cartilage homeostasis and impairs the metabolic activity of chondrocytes. Mechanical stress stimulates talin-centered cytoskeletal reorganization and the consequent increase of cell contractile forces and cell stiffness of chondrocytes, which triggers αV integrin-mediated TGFβ activation. Knockout of αV integrin in chondrocytes reversed the alteration of TGFβ activation and subsequent metabolic abnormalities in AC and attenuated cartilage degeneration in an OA mouse model. Thus, SB structure determines the patterns of mechanical stress and the configuration of TGFβ activation in AC, which subsequently regulates chondrocyte metabolism and AC homeostasis.
Collapse
Affiliation(s)
- Gehua Zhen
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Qiaoyue Guo
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Yusheng Li
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Chuanlong Wu
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Shouan Zhu
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Ruomei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - X Edward Guo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Byoung Choul Kim
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University, Baltimore, MD, USA
| | - Jessie Huang
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, NJ, USA
| | - Yizhong Hu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yang Dan
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Taekjip Ha
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University, Baltimore, MD, USA
| | - Steven An
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, NJ, USA
- Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, USA
| | - Xu Cao
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
38
|
Molecular mechanisms of mechanical load-induced osteoarthritis. INTERNATIONAL ORTHOPAEDICS 2021; 45:1125-1136. [PMID: 33459826 DOI: 10.1007/s00264-021-04938-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 01/07/2021] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Mechanical loading enhances the progression of osteoarthritis. However, its molecular mechanisms have not been established. OBJECTIVE The aim of this review was to summarize the probable mechanisms of mechanical load-induced osteoarthritis. METHODS A comprehensive search strategy was used to search PubMed and EMBASE databases (from the 15th of January 2015 to the 20th of October 2020). Search terms included "osteoarthritis", "mechanical load", and "mechanism". RESULTS Abnormal mechanical loading activates the interleukin-1β, tumour necrosis factor-α, nuclear factor kappa-B, Wnt, transforming growth factor-β, microRNAs pathways, and the oxidative stress pathway. These pathways induce the pathological progression of osteoarthritis. Mechanical stress signal receptors such as integrin, ion channel receptors, hydrogen peroxide-inducible clone-5, Gremlin-1, and transient receptor potential channel 4 are present in the articular cartilages. CONCLUSION This review highlights the molecular mechanisms of mechanical loading in inducing chondrocyte apoptosis and extracellular matrix degradation. These mechanisms provide potential targets for osteoarthritis prevention and treatment.
Collapse
|
39
|
He Y, Makarczyk MJ, Lin H. Role of mitochondria in mediating chondrocyte response to mechanical stimuli. Life Sci 2020; 263:118602. [PMID: 33086121 PMCID: PMC7736591 DOI: 10.1016/j.lfs.2020.118602] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/22/2020] [Accepted: 10/11/2020] [Indexed: 12/21/2022]
Abstract
As the most common form of arthritis, osteoarthritis (OA) has become a major cause of severe joint pain, physical disability, and quality of life impairment in the affected population. To date, precise pathogenesis of OA has not been fully clarified, which leads to significant obstacles in developing efficacious treatments such as failures in finding disease-modifying OA drugs (DMOADs) in the last decades. Given that diarthrodial joints primarily display the weight-bearing and movement-supporting function, it is not surprising that mechanical stress represents one of the major risk factors for OA. However, the inner connection between mechanical stress and OA onset/progression has yet to be explored. Mitochondrion, a widespread organelle involved in complex biological regulation processes such as adenosine triphosphate (ATP) synthesis and cellular metabolism, is believed to have a controlling role in the survival and function implement of chondrocytes, the singular cell type within cartilage. Mitochondrial dysfunction has also been observed in osteoarthritic chondrocytes. In this review, we systemically summarize mitochondrial alterations in chondrocytes during OA progression and discuss our recent progress in understanding the potential role of mitochondria in mediating mechanical stress-associated osteoarthritic alterations of chondrocytes. In particular, we propose the potential signaling pathways that may regulate this process, which provide new views and therapeutic targets for the prevention and treatment of mechanical stress-associated OA.
Collapse
Affiliation(s)
- Yuchen He
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Meagan J Makarczyk
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
40
|
Walsh SK, Shelley JC, Henak CR. Mechanobiology of Cartilage Impact Via Real-Time Metabolic Imaging. J Biomech Eng 2020; 142:100802. [PMID: 32542333 DOI: 10.1115/1.4047534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Indexed: 11/08/2022]
Abstract
Cartilage loading is important in both structural and biological contexts, with overloading known to cause osteoarthritis (OA). Cellular metabolism, which can be evaluated through the relative measures of glycolysis and oxidative phosphorylation, is important in disease processes across tissues. Details of structural damage coupled with cellular metabolism in cartilage have not been evaluated. Therefore, the aim of this study was to characterize the time- and location-dependent metabolic response to traumatic impact loading in articular cartilage. Cartilage samples from porcine femoral condyles underwent a single traumatic injury that created cracks in most samples. Before and up to 30 min after loading, samples underwent optical metabolic imaging. Optical metabolic imaging measures the fluorescent intensity of byproducts of the two metabolic pathways, flavin adenine dinucleotide for oxidative phosphorylation and nicotinamide adenine dinucleotide ± phosphate for glycolysis, as well as the redox ratio between them. Images were taken at varied distances from the center of the impact. Shortly after impact, fluorescence intensity in both channels decreased, while redox ratio was unchanged. The most dramatic metabolic response was measured closest to the impact center, with suppressed fluorescence in both channels relative to baseline. Redox ratio varied nonlinearly as a function of distance from the impact. Finally, both lower and higher magnitude loading reduced flavin adenine dinucleotide fluorescence, whereas reduced nicotinamide adenine dinucleotide ± phosphate fluorescence was associated only with low strain loads and high contact pressure loads, respectively. In conclusion, this study performed novel analysis of metabolic activity following induction of cartilage damage and demonstrated time-, distance-, and load-dependent response to traumatic impact loading.
Collapse
Affiliation(s)
- Shannon K Walsh
- Comparative Biomedical Sciences Program, University of Wisconsin-Madison, Madison, WI 53706
| | - Joshua C Shelley
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53706
| | - Corinne R Henak
- Department of Mechanical Engineering, University of Wisconsin-Madison, 3031 Mechanical Engineering Building, 1513 University Ave. Madison, WI 53706; Department of Biomedical Engineering, University of Wisconsin-Madison, 3031 Mechanical Engineering Building, 1513 University Ave. Madison, WI 53706; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 3031 Mechanical Engineering Building, 1513 University Ave. Madison, WI 53705
| |
Collapse
|
41
|
Chen L, Zheng JJY, Li G, Yuan J, Ebert JR, Li H, Papadimitriou J, Wang Q, Wood D, Jones CW, Zheng M. Pathogenesis and clinical management of obesity-related knee osteoarthritis: Impact of mechanical loading. J Orthop Translat 2020; 24:66-75. [PMID: 32695606 PMCID: PMC7349942 DOI: 10.1016/j.jot.2020.05.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/28/2020] [Accepted: 05/02/2020] [Indexed: 12/16/2022] Open
Abstract
Obesity-related osteoarthritis (OA) is a complex, multifactorial condition that can cause significant impact on patients' quality of life. Whilst chronic inflammation, adipocytokines and metabolic factors are considered to be important pathogenic factors in obesity related OA, there has been limited investigation into the biomechanical impact of obesity on OA development. This review aims to demonstrate that mechanical factors are the major pathological cause of obesity-related OA. The effect of obesity on pathological changes to the osteochondral unit and surrounding connective tissues in OA is summarized, as well as the impact of obesity-related excessive and abnormal joint loading, concomitant joint malalignment and muscle weakness. An integrated therapeutic strategy based on this multi-factorial presentation is presented, to assist in the management of obesity related OA. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE Despite the high prevalence of obesity-related OA, there is no specific guideline available for obesity-related OA management. In this review, we demonstrated the pathological changes of obesity-related OA and summarized the impact of biomechanical factors by proposing a hypothetical model of obesity-related OA change. Therapeutic strategies based on adjusting abnormal mechanical effects are presented to assist in the management of obesity-related OA.
Collapse
Affiliation(s)
- Lianzhi Chen
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | | | - Guangyi Li
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jun Yuan
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Perth, Western Australia, Australia
| | - Jay R. Ebert
- School of Human Sciences (Exercise and Sport Science), University of Western Australia, Perth, Western Australia, Australia
| | - Hengyuan Li
- Department of Orthopedics, Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang, China
| | - John Papadimitriou
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia, Australia
- Pathwest Laboratories, Perth, Western Australia, Australia
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - David Wood
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia, Australia
| | - Christopher W. Jones
- Fiona Stanley Hospital Group, Perth, Western Australia, Australia
- Curtin University Medical School, Perth, Western Australia, Australia
| | - Minghao Zheng
- Centre for Orthopaedic Research, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Perth, Western Australia, Australia
| |
Collapse
|
42
|
Gratal P, Lamuedra A, Medina JP, Bermejo-Álvarez I, Largo R, Herrero-Beaumont G, Mediero A. Purinergic System Signaling in Metainflammation-Associated Osteoarthritis. Front Med (Lausanne) 2020; 7:506. [PMID: 32984382 PMCID: PMC7485330 DOI: 10.3389/fmed.2020.00506] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022] Open
Abstract
Inflammation triggered by metabolic imbalance, also called metainflammation, is low-grade inflammation caused by the components involved in metabolic syndrome (MetS), including central obesity and impaired glucose tolerance. This phenomenon is mainly due to excess nutrients and energy, and it contributes to the pathogenesis of osteoarthritis (OA). OA is characterized by the progressive degeneration of articular cartilage, which suffers erosion and progressively becomes thinner. Purinergic signaling is involved in several physiological and pathological processes, such as cell proliferation in development and tissue regeneration, neurotransmission and inflammation. Adenosine and ATP receptors, and other members of the signaling pathway, such as AMP-activated protein kinase (AMPK), are involved in obesity, type 2 diabetes (T2D) and OA progression. In this review, we focus on purinergic regulation in osteoarthritic cartilage and how different components of MetS, such as obesity and T2D, modulate the purinergic system in OA. In that regard, we describe the critical role in this disease of receptors, such as adenosine A2A receptor (A2AR) and ATP P2X7 receptor. Finally, we also assess how nucleotides regulate the inflammasome in OA.
Collapse
Affiliation(s)
- Paula Gratal
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Ana Lamuedra
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | - Juan Pablo Medina
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | | | - Raquel Largo
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| | | | - Aránzazu Mediero
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz UAM, Madrid, Spain
| |
Collapse
|
43
|
Wang Z, Ai S, Tian F, Liow MHL, Wang S, Zhao J, Tsai TY. Higher Body Mass Index Is Associated With Biochemical Changes in Knee Articular Cartilage After Marathon Running: A Quantitative T2-Relaxation MRI Study. Orthop J Sports Med 2020; 8:2325967120943874. [PMID: 32851106 PMCID: PMC7427140 DOI: 10.1177/2325967120943874] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/25/2020] [Indexed: 11/23/2022] Open
Abstract
Background: More than 30 million individuals participate in marathon running every year worldwide. As the popularity of marathon running continues to increase, it is essential for the purposes of injury prevention to understand the effects of marathon running on the knee cartilage. Purpose: To investigate the immediate effects of marathon running on knee articular cartilage and to determine the relationship between body mass index and cartilage biochemical composition. Study Design: Descriptive laboratory study. Methods: T2-relaxation magnetic resonance imaging (MRI) of knees in 18 nonprofessional marathoners (mean age, 35.6 ± 6.4 years) was performed before and after a full-length marathon. Three-dimensional models of the knee articular cartilage were reconstructed and divided into different regions of interest. The 3-dimensional models were then applied to corresponding T2-relaxation MRI maps to calculate T2 values in each region of interest. The mean values of the T2-relaxation times in each region of interest before and after the marathon were compared by use of the paired Student t test. The Pearson correlation coefficient between T2 change and runner body mass index (BMI) was calculated. Results: Postmarathon T2-relaxation times were significantly higher than premarathon values for patellofemoral cartilage (32.6 ± 12.1 vs 34.1 ± 10.9 ms; P < .01) and medial tibial cartilage (35.6 ± 11.7 vs 34.6 ± 12.0 ms; P = .01). The greatest increase was observed in the anterior part of the medial tibial cartilage. No statistically significant changes were seen in the T2-relaxation times of the lateral tibial and femoral cartilage. Postmarathon T2-relaxation elevation in the anteromedial knee tibiofemoral joint cartilage strongly correlated with body weight (R = 0.6746; P = .03) and BMI (R = 0.6989; P = .001). Changes in T2-relaxation times did not correlate with marathon time, height, age, or sex in any regions of interest. Conclusion: Marathon running leads to immediate postmarathon elevated T2-relaxation values within knee articular cartilage, suggesting biochemical content alteration. Additionally, runners with higher BMI may have greater changes in cartilage biochemical composition after a marathon. Further studies should investigate whether these changes are sustained over time to determine the relationship between immediate biochemical changes in cartilage composition and cartilage degeneration. Clinical Relevance: Runners with a higher BMI may carry a higher risk of anteromedial tibiofemoral cartilage degeneration compared with runners with lower BMI.
Collapse
Affiliation(s)
- Zhongzheng Wang
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University; Engineering Research Center of Digital Medicine and Clinical Translation, Ministry of Education, China.,Shanghai Key Laboratory of Orthopaedic Implants & Clinical Translation R&D Center of 3D Printing Technology, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Songtao Ai
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University; Engineering Research Center of Digital Medicine and Clinical Translation, Ministry of Education, China.,Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Tian
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | | | - Shaobai Wang
- Key Laboratory of Exercise and Health Sciences of Ministry of Education, School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Jinzhong Zhao
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Tsung-Yuan Tsai
- School of Biomedical Engineering & Med-X Research Institute, Shanghai Jiao Tong University; Engineering Research Center of Digital Medicine and Clinical Translation, Ministry of Education, China.,Shanghai Key Laboratory of Orthopaedic Implants & Clinical Translation R&D Center of 3D Printing Technology, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
44
|
Zheng L, Wang Y, Qiu P, Xia C, Fang Y, Mei S, Fang C, Shi Y, Wu K, Chen Z, Fan S, He D, Lin X, Chen P. Primary chondrocyte exosomes mediate osteoarthritis progression by regulating mitochondrion and immune reactivity. Nanomedicine (Lond) 2020; 14:3193-3212. [PMID: 31855117 DOI: 10.2217/nnm-2018-0498] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: We aimed to investigate the proteomics of primary chondrocyte exosomes and the effect of exosomes in osteoarthritis (OA) treatment. Materials & methods: We isolated exosomes from primary chondrocytes cultured in normal (D0) and inflammatory environments induced by IL-1β and determined the proteomics of these exosomes. Next, we investigated what effect and mechanism D0 chondrocytes exosomes have in OA treatment. Results: There were more proteins that belonged to mitochondrion and were involved in immune system processes in D0 exosomes. Notably, intra-articular administration of D0 exosomes successfully prevented the development of OA. D0 chondrocyte exosomes could restore mitochondrial dysfunction and polarize macrophage response toward an M2 phenotype. Conclusion: Our findings demonstrated that primary chondrocyte exosomes are efficient in OA treatment.
Collapse
Affiliation(s)
- Lin Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China.,Department of Orthopedics, 5th Affiliated Hospital, Lishui Municipal Central Hospital, Wenzhou Medical University, Lishui, PR China
| | - Yiyun Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Pengcheng Qiu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Chen Xia
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Yifan Fang
- Hangzhou Foreign Languages School, Hangzhou, PR China
| | - Sheng Mei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Chen Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Yiling Shi
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Kaiwei Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Zhijun Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Dengwei He
- Department of Orthopedics, 5th Affiliated Hospital, Lishui Municipal Central Hospital, Wenzhou Medical University, Lishui, PR China
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| | - Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, PR China.,Key Laboratory of Musculoskeletal System Degeneration & Regeneration Translational Research of Zhejiang Province, Hangzhou, PR China
| |
Collapse
|
45
|
Corciulo C, Cronstein BN. Signaling of the Purinergic System in the Joint. Front Pharmacol 2020; 10:1591. [PMID: 32038258 PMCID: PMC6993121 DOI: 10.3389/fphar.2019.01591] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022] Open
Abstract
The joint is a complex anatomical structure consisting of different tissues, each with a particular feature, playing together to give mobility and stability at the body. All the joints have a similar composition including cartilage for reducing the friction of the movement and protecting the underlying bone, a synovial membrane that produces synovial fluid to lubricate the joint, ligaments to limit joint movement, and tendons for the interaction with muscles. Direct or indirect damage of one or more of the tissues forming the joint is the foundation of different pathological conditions. Many molecular mechanisms are involved in maintaining the joint homeostasis as well as in triggering disease development. The molecular pathway activated by the purinergic system is one of them.The purinergic signaling defines a group of receptors and intermembrane channels activated by adenosine, adenosine diphosphate, adenosine 5’-triphosphate, uridine triphosphate, and uridine diphosphate. It has been largely described as a modulator of many physiological and pathological conditions including rheumatic diseases. Here we will give an overview of the purinergic system in the joint describing its expression and function in the synovium, cartilage, ligament, tendon, and bone with a therapeutic perspective.
Collapse
Affiliation(s)
- Carmen Corciulo
- Division of Translational Medicine, Department of Medicine, NYU School of Medicine, New York, NY, United States.,Krefting Research Centre-Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Bruce N Cronstein
- Division of Translational Medicine, Department of Medicine, NYU School of Medicine, New York, NY, United States.,Division of Rheumatology, Department of Medicine, NYU School of Medicine, New York, NY, United States
| |
Collapse
|
46
|
Zhu S, Makosa D, Miller BF, Griffin TM. Glutathione as a mediator of cartilage oxidative stress resistance and resilience during aging and osteoarthritis. Connect Tissue Res 2020; 61:34-47. [PMID: 31522568 PMCID: PMC6884680 DOI: 10.1080/03008207.2019.1665035] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: An underlying cause of osteoarthritis (OA) is the inability of chondrocytes to maintain homeostasis in response to changing stress conditions. The purpose of this article was to review and experimentally evaluate oxidative stress resistance and resilience concepts in cartilage using glutathione redox homeostasis as an example. This framework may help identify novel approaches for promoting chondrocyte homeostasis during aging and obesity.Materials and Methods: Changes in glutathione content and redox ratio were evaluated in three models of chondrocyte stress: (1) age- and tissue-specific changes in joint tissues of 10 and 30-month old F344BN rats, including ex vivo patella culture experiments to evaluate N-acetylcysteine dependent resistance to interleukin-1beta; (2) effect of different durations and patterns of cyclic compressive loading in bovine cartilage on glutathione stress resistance and resilience pathways; (3) time-dependent changes in GSH:GSSG in primary chondrocytes from wild-type and Sirt3 deficient mice challenged with the pro-oxidant menadione.Results: Glutathione was more abundant in cartilage than meniscus or infrapatellar fat pad, although cartilage was also more susceptible to age-related glutathione oxidation. Glutathione redox homeostasis was sensitive to the duration of compressive loading such that load-induced oxidation required unloaded periods to recover and increase total antioxidant capacity. Exposure to a pro-oxidant stress enhanced stress resistance by increasing glutathione content and GSH:GSSG ratio, especially in Sirt3 deficient cells. However, the rate of recovery, a marker of resilience, was delayed without Sirt3.Conclusions: OA-related models of cartilage stress reveal multiple mechanisms by which glutathione provides oxidative stress resistance and resilience.
Collapse
Affiliation(s)
- Shouan Zhu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Dawid Makosa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA,Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Benjamin F. Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Timothy M. Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
47
|
Real-time optical redox imaging of cartilage metabolic response to mechanical loading. Osteoarthritis Cartilage 2019; 27:1841-1850. [PMID: 31513919 DOI: 10.1016/j.joca.2019.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Metabolic dysregulation has recently been identified as a key feature of osteoarthritis. Mechanical overloading has been postulated as a primary cause of this metabolic response. Current methods of real-time metabolic activity analysis in cartilage are limited and challenging. However, optical redox imaging leverages the autofluorescence of co-enzymes NAD(P)H and FAD to provide dye-free real-time analysis of metabolic activity. This technique has not yet been applied to cartilage. This study aimed to assess the effects of a compressive load on cartilage using optical redox imaging. METHOD Cartilage samples were excised from porcine femoral condyles. To validate this imaging modality in cartilage, glycolysis was inhibited via 2-deoxy-D-glucose (2DG) and oxidative phosphorylation was inhibited by rotenone. Optical redox images were collected pre- and post-inhibition. To assess the effects of mechanical loading, samples were subjected to a compressive load and imaged for approximately 30 min. Load and strain parameters were determined using high-speed camera images in Matlab. A range of loading magnitudes and rates were applied across samples. RESULTS 2DG and rotenone demonstrated the expected inhibitory effects on fluorescence intensity in the channels corresponding to NAD(P)H and FAD, respectively. Mechanical loading induced an increase in NAD(P)H channel fluorescence which subsided by 30 min post-loading. Magnitude of loading parameters had mixed effects on metabolites. CONCLUSIONS Optical redox imaging provides an opportunity to assess real-time metabolic activity in cartilage. This approach revealed a metabolic response to a single load and can be used to provide insight into the role of metabolism in mechanically-mediated cartilage degradation.
Collapse
|
48
|
Coleman MC, Goetz JE, Brouillette MJ, Seol D, Willey MC, Petersen EB, Anderson HD, Hendrickson NR, Compton J, Khorsand B, Morris AS, Salem AK, Fredericks DC, McKinley TO, Martin JA. Targeting mitochondrial responses to intra-articular fracture to prevent posttraumatic osteoarthritis. Sci Transl Med 2019; 10:10/427/eaan5372. [PMID: 29437147 DOI: 10.1126/scitranslmed.aan5372] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/19/2017] [Accepted: 01/05/2018] [Indexed: 12/16/2022]
Abstract
We tested whether inhibiting mechanically responsive articular chondrocyte mitochondria after severe traumatic injury and preventing oxidative damage represent a viable paradigm for posttraumatic osteoarthritis (PTOA) prevention. We used a porcine hock intra-articular fracture (IAF) model well suited to human-like surgical techniques and with excellent anatomic similarities to human ankles. After IAF, amobarbital or N-acetylcysteine (NAC) was injected to inhibit chondrocyte electron transport or downstream oxidative stress, respectively. Effects were confirmed via spectrophotometric enzyme assays or glutathione/glutathione disulfide assays and immunohistochemical measures of oxidative stress. Amobarbital or NAC delivered after IAF provided substantial protection against PTOA at 6 months, including maintenance of proteoglycan content, decreased histological disease scores, and normalized chondrocyte metabolic function. These data support the therapeutic potential of targeting chondrocyte metabolism after injury and suggest a strong role for mitochondria in mediating PTOA.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Todd O McKinley
- Indiana University Health Methodist Hospital Orthopaedic Trauma Service, Indianapolis, IN 46202, USA
| | | |
Collapse
|
49
|
Stojanović B, Bauer C, Stotter C, Klestil T, Nehrer S, Franek F, Rodríguez Ripoll M. Tribocorrosion of a CoCrMo alloy sliding against articular cartilage and the impact of metal ion release on chondrocytes. Acta Biomater 2019; 94:597-609. [PMID: 31226479 DOI: 10.1016/j.actbio.2019.06.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 01/22/2023]
Abstract
Partial knee replacement and hemiarthroplasty are some of the orthopedic procedures resulting in a metal on cartilage interface. As metal implant material, CoCrMo based alloys are commonly used. The aim of the present study is to assess the role of biotribocorrosion on the CoCrMo-cartilage interface with an emphasis on metal release during sliding contact. The biotribocorrosion experiments were performed under controlled electrochemical conditions using a floating cell with a three electrode set up coupled to a microtribometer. Throughout the experiment the coefficient of friction and the open circuit potential were monitored. Analyses of the electrolyte after the experiment show that metal release can occur during sliding contact of CoCrMo alloy against articular cartilage despite the extraordinary low coefficient of friction measured. Metal release is attributed to changes in passive layer caused at the onset of sliding. The released metal was found to be forming compounds with potential cytotoxicity. Since the presence of metal ions in the cartilage matrix can potentially lead to cell apoptosis, the metabolic activity of human osteoarthritic chondrocytes (2D-cultures) was investigated in the presence of phosphate buffered saline containing metal ions using XTT-assay. The experiments indicate that critical concentrations of Co ions lead to a significant decrease in chondrocyte metabolic activity. Therefore, biotribocorrosion is a mechanism that can occur in partial replacements and lead to chondrocyte apoptosis thus playing a role in the observed accelerated degradation of the remaining cartilage tissue after the mentioned orthopedic procedures. STATEMENT OF SIGNIFICANCE: Partial replacements provide an alternative to total joint replacements. This procedure is less invasive, allows a faster rehabilitation and provides a better function of the joint. However, the remaining native cartilage experiences accelerated degradation when in contact with metallic implant components. This work investigates the role of tribocorrosion at the metal-cartilage interface during sliding. Tribocorrosion is a degradation process that can alter significantly the wear rates experienced by metallic implants and lead to the release of metal ions and particles. The released metal can form compounds with potential cytotoxicity on cartilage tissue. The knowledge gained in this work will serve to understand the mechanisms behind the failure of partial replacements and develop future biomaterials with an enhanced lifetime.
Collapse
Affiliation(s)
- B Stojanović
- AC2T research GmbH, Viktor Kaplan-Straße 2/C, A-2700 Wiener Neustadt, Austria
| | - C Bauer
- Danube University Krems, Faculty of Health and Medicine, Department for Health Sciences and Biomedicine, Center for Regenerative Medicine and Orthopedics, Dr. Karl-Dorrek-Str. 30, A-3500 Krems, Austria
| | - C Stotter
- Danube University Krems, Faculty of Health and Medicine, Department for Health Sciences and Biomedicine, Center for Regenerative Medicine and Orthopedics, Dr. Karl-Dorrek-Str. 30, A-3500 Krems, Austria; LK Baden-Mödling-Hainburg, Department of Orthopedics and Traumatology, Waltersdorferstraße 75, A-2500 Baden, Austria
| | - T Klestil
- LK Baden-Mödling-Hainburg, Department of Orthopedics and Traumatology, Waltersdorferstraße 75, A-2500 Baden, Austria; Danube University Krems, Faculty of Health and Medicine, Department for Health Sciences and Biomedicine, Center for Medical Specializations, Dr. Karl-Dorrek-Str. 30, A-3500 Krems, Austria
| | - S Nehrer
- Danube University Krems, Faculty of Health and Medicine, Department for Health Sciences and Biomedicine, Center for Regenerative Medicine and Orthopedics, Dr. Karl-Dorrek-Str. 30, A-3500 Krems, Austria
| | - F Franek
- AC2T research GmbH, Viktor Kaplan-Straße 2/C, A-2700 Wiener Neustadt, Austria
| | - M Rodríguez Ripoll
- AC2T research GmbH, Viktor Kaplan-Straße 2/C, A-2700 Wiener Neustadt, Austria.
| |
Collapse
|
50
|
Salinas D, Mumey B, June RK. Physiological dynamic compression regulates central energy metabolism in primary human chondrocytes. Biomech Model Mechanobiol 2019; 18:69-77. [PMID: 30097814 PMCID: PMC9851408 DOI: 10.1007/s10237-018-1068-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 08/01/2018] [Indexed: 01/21/2023]
Abstract
Chondrocytes use the pathways of central metabolism to synthesize molecular building blocks and energy for cartilage homeostasis. An interesting feature of the in vivo chondrocyte environment is the cyclical loading generated in various activities (e.g., walking). However, it is unknown whether central metabolism is altered by mechanical loading. We hypothesized that physiological dynamic compression alters central metabolism in chondrocytes to promote production of amino acid precursors for matrix synthesis. We measured the expression of central metabolites (e.g., glucose, its derivatives, and relevant co-factors) for primary human osteoarthritic chondrocytes in response to 0-30 minutes of compression. To analyze the data, we used principal components analysis and ANOVA-simultaneous components analysis, as well as metabolic flux analysis. Compression-induced metabolic responses consistent with our hypothesis. Additionally, these data show that chondrocyte samples from different patient donors exhibit different sensitivity to compression. Most importantly, we find that grade IV osteoarthritic chondrocytes are capable of synthesizing non-essential amino acids and precursors in response to mechanical loading. These results suggest that further advances in metabolic engineering of chondrocyte mechanotransduction may yield novel translational strategies for cartilage repair.
Collapse
Affiliation(s)
- Daniel Salinas
- Department of Computer Science, Montana State University, PO Box 173800, Bozeman, MT 59717-3800
| | - Brendan Mumey
- Department of Computer Science, Montana State University, PO Box 173800, Bozeman, MT 59717-3800
| | - Ronald K. June
- Department of Mechanical and Industrial Engineering, Montana State University, PO Box 173800, Bozeman, MT 59717-3800
| |
Collapse
|