1
|
Irwin RM, Thomas MA, Fahey MJ, Mayán MD, Smyth JW, Delco ML. Connexin 43 regulates intercellular mitochondrial transfer from human mesenchymal stromal cells to chondrocytes. Stem Cell Res Ther 2024; 15:359. [PMID: 39390589 PMCID: PMC11468299 DOI: 10.1186/s13287-024-03932-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND The phenomenon of intercellular mitochondrial transfer from mesenchymal stromal cells (MSCs) has shown promise for improving tissue healing after injury and has potential for treating degenerative diseases like osteoarthritis (OA). Recently MSC to chondrocyte mitochondrial transfer has been documented, but the mechanism of transfer is unknown. Full-length connexin 43 (Cx43, encoded by GJA1) and the truncated, internally translated isoform GJA1-20k have been implicated in mitochondrial transfer between highly oxidative cells, but have not been explored in orthopaedic tissues. Here, our goal was to investigate the role of Cx43 in MSC to chondrocyte mitochondrial transfer. In this study, we tested the hypotheses that (a) mitochondrial transfer from MSCs to chondrocytes is increased when chondrocytes are under oxidative stress and (b) MSC Cx43 expression mediates mitochondrial transfer to chondrocytes. METHODS Oxidative stress was induced in immortalized human chondrocytes using tert-Butyl hydroperoxide (t-BHP) and cells were evaluated for mitochondrial membrane depolarization and reactive oxygen species (ROS) production. Human bone-marrow derived MSCs were transduced for mitochondrial fluorescence using lentiviral vectors. MSC Cx43 expression was knocked down using siRNA or overexpressed (GJA1 + and GJA1-20k+) using lentiviral transduction. Chondrocytes and MSCs were co-cultured for 24 h in direct contact or separated using transwells. Mitochondrial transfer was quantified using flow cytometry. Co-cultures were fixed and stained for actin and Cx43 to visualize cell-cell interactions during transfer. RESULTS Mitochondrial transfer was significantly higher in t-BHP-stressed chondrocytes. Contact co-cultures had significantly higher mitochondrial transfer compared to transwell co-cultures. Confocal images showed direct cell contacts between MSCs and chondrocytes where Cx43 staining was enriched at the terminal ends of actin cellular extensions containing mitochondria in MSCs. MSC Cx43 expression was associated with the magnitude of mitochondrial transfer to chondrocytes; knocking down Cx43 significantly decreased transfer while Cx43 overexpression significantly increased transfer. Interestingly, GJA1-20k expression was highly correlated with incidence of mitochondrial transfer from MSCs to chondrocytes. CONCLUSIONS Overexpression of GJA1-20k in MSCs increases mitochondrial transfer to chondrocytes, highlighting GJA1-20k as a potential target for promoting mitochondrial transfer from MSCs as a regenerative therapy for cartilage tissue repair in OA.
Collapse
Affiliation(s)
- Rebecca M Irwin
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Matthew A Thomas
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Megan J Fahey
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Universidade da Coruña (UDC), A Coruña, Spain
| | - James W Smyth
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, FBRI at VTC, Roanoke, VA, 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Michelle L Delco
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
2
|
Shen Y, Wang X, Nan N, Fu X, Zeng R, Yang Y, Xian S, Shi J, Wu Q, Zhou S. SIRT3-Mediated Deacetylation of SDHA Rescues Mitochondrial Bioenergetics Contributing to Neuroprotection in Rotenone-Induced PD Models. Mol Neurobiol 2024; 61:4402-4420. [PMID: 38087172 DOI: 10.1007/s12035-023-03830-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/25/2023] [Indexed: 07/11/2024]
Abstract
Mitochondrial dysfunction is critically involved in the degeneration of dopamine (DA) neurons in the substantia nigra, a common pathological feature of Parkinson's disease (PD). Previous studies have demonstrated that the NAD+-dependent acetylase Sirtuin 3 (SIRT3) participates in maintaining mitochondrial function and is downregulated in aging-related neurodegenerative disorders. The exact mechanism of action of SIRT3 on mitochondrial bioenergetics in PD pathogenesis, however, has not been fully described. In this study, we investigated the regulatory role of SIRT3-mediated deacetylation of mitochondrial complex II (succinate dehydrogenase) subunit A (SDHA) and its effect on neuronal cell survival in rotenone (ROT)-induced rat and differentiated MN9D cell models. The results revealed that SIRT3 activity was suppressed in both in vivo and in vitro PD models. Accompanying this downregulation of SIRT3 was the hyperacetylation of SDHA, impaired activity of mitochondrial complex II, and decreased ATP production. It was found that the inhibition of SIRT3 activity was attributed to a reduction in the NAD+/NADH ratio caused by ROT-induced inhibition of mitochondrial complex I. Activation of SIRT3 by icariin and honokiol inhibited SDHA hyperacetylation and increased complex II activity, leading to increased ATP production and protection against ROT-induced neuronal damage. Furthermore, overexpression of SDHA also exerted potent protective benefits in cells treated with ROT. In addition, treatment of MN9D cells with the NAD+ precursor nicotinamide mononucleotide increased SIRT3 activity and complex II activity and promoted the survival of cells exposed to ROT. These findings unravel a regulatory SIRT3-SDHA axis, which may be closely related to PD pathology. Bioenergetic rescue through SIRT3 activation-dependent improvement of mitochondrial complex II activity may provide an effective strategy for protection from neurodegeneration.
Collapse
Affiliation(s)
- Yanhua Shen
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xueting Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Nan Nan
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiaolong Fu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ru Zeng
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yonggang Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Siting Xian
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shaoyu Zhou
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
3
|
Poudel SB, Ruff RR, Yildirim G, Miller RA, Harrison DE, Strong R, Kirsch T, Yakar S. Development of primary osteoarthritis during aging in genetically diverse UM-HET3 mice. Arthritis Res Ther 2024; 26:118. [PMID: 38851726 PMCID: PMC11161968 DOI: 10.1186/s13075-024-03349-y] [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: 01/12/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Primary osteoarthritis (OA) occurs without identifiable underlying causes such as previous injuries or specific medical conditions. Age is a major contributing factor to OA, and as one ages, various joint tissues undergo gradual change, including degeneration of the articular cartilage, alterations in subchondral bone (SCB) morphology, and inflammation of the synovium. METHODS We investigated the prevalence of primary OA in aged, genetically diverse UM-HET3 mice. Articular cartilage (AC) integrity and SCB morphology were assessed in 182 knee joints of 22-25 months old mice using the Osteoarthritis Research Society International (OARSI) scoring system and micro-CT, respectively. Additionally, we explored the effects of methylene blue (MB) and mitoquinone (MitoQ), two agents that affect mitochondrial function, on the prevalence and progression of OA during aging. RESULTS Aged UM-HET3 mice showed a high prevalence of primary OA in both sexes. Significant positive correlations were found between cumulative AC (cAC) scores and synovitis in both sexes, and osteophyte formation in female mice. Ectopic chondrogenesis did not show significant correlations with cAC scores. Significant direct correlations were found between AC scores and inflammatory markers in chondrocytes, including matrix metalloproteinase-13, inducible nitric oxide synthase, and the NLR family pyrin domain containing-3 inflammasome in both sexes, indicating a link between OA severity and inflammation. Additionally, markers of cell cycle arrest, such as p16 and β-galactosidase, also correlated with AC scores. In male mice, no significant correlations were found between SCB morphology traits and cAC scores, while in female mice, significant correlations were found between cAC scores and tibial SCB plate bone mineral density. Notably, MB and MitoQ treatments influenced the disease's progression in a sex-specific manner. MB treatment significantly reduced cAC scores at the medial knee joint, while MitoQ treatment reduced cAC scores, but these did not reach significance. CONCLUSIONS Our study provides comprehensive insights into the prevalence and progression of primary OA in aged UM-HET3 mice, highlighting the sex-specific effects of MB and MitoQ treatments. The correlations between AC scores and various pathological factors underscore the multifaceted nature of OA and its association with inflammation and subchondral bone changes.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, 345 East 24th Street, New York, NY, 10010-4086, USA
| | - Ryan R Ruff
- David B. Kriser Dental Center, Biostatistics Core, Department of Epidemiology and Health Promotion, New York University College of Dentistry, New York, NY, 10010-4086, USA
| | - Gozde Yildirim
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, 345 East 24th Street, New York, NY, 10010-4086, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, 48105, USA
| | | | - Randy Strong
- Geriatric Research, Education and Clinical Center and Research Service, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA
- Barshop Institute for Longevity and Aging Studies and Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX, 78229, USA
| | - Thorsten Kirsch
- Department of Orthopaedic Surgery, NYU Grossman School of Medicine, New York, NY, 10100, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, New York, NY, 10010, USA
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, 345 East 24th Street, New York, NY, 10010-4086, USA.
| |
Collapse
|
4
|
Da W, Chen Q, Shen B. The current insights of mitochondrial hormesis in the occurrence and treatment of bone and cartilage degeneration. Biol Res 2024; 57:37. [PMID: 38824571 PMCID: PMC11143644 DOI: 10.1186/s40659-024-00494-1] [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: 07/31/2023] [Accepted: 04/03/2024] [Indexed: 06/03/2024] Open
Abstract
It is widely acknowledged that aging, mitochondrial dysfunction, and cellular phenotypic abnormalities are intricately associated with the degeneration of bone and cartilage. Consequently, gaining a comprehensive understanding of the regulatory patterns governing mitochondrial function and its underlying mechanisms holds promise for mitigating the progression of osteoarthritis, intervertebral disc degeneration, and osteoporosis. Mitochondrial hormesis, referred to as mitohormesis, represents a cellular adaptive stress response mechanism wherein mitochondria restore homeostasis and augment resistance capabilities against stimuli by generating reactive oxygen species (ROS), orchestrating unfolded protein reactions (UPRmt), inducing mitochondrial-derived peptides (MDP), instigating mitochondrial dynamic changes, and activating mitophagy, all prompted by low doses of stressors. The varying nature, intensity, and duration of stimulus sources elicit divergent degrees of mitochondrial stress responses, subsequently activating one or more signaling pathways to initiate mitohormesis. This review focuses specifically on the effector molecules and regulatory networks associated with mitohormesis, while also scrutinizing extant mechanisms of mitochondrial dysfunction contributing to bone and cartilage degeneration through oxidative stress damage. Additionally, it underscores the potential of mechanical stimulation, intermittent dietary restrictions, hypoxic preconditioning, and low-dose toxic compounds to trigger mitohormesis, thereby alleviating bone and cartilage degeneration.
Collapse
Affiliation(s)
- Wacili Da
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Quan Chen
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Bin Shen
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
5
|
Poudel SB, Ruff RR, Yildirim G, Miller RA, Harrison DE, Strong R, Kirsch T, Yakar S. Development of primary osteoarthritis during aging in genetically diverse UM-HET3 mice. RESEARCH SQUARE 2024:rs.3.rs-3858256. [PMID: 38343826 PMCID: PMC10854287 DOI: 10.21203/rs.3.rs-3858256/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background Primary osteoarthritis (OA) occurs without identifiable underlying causes such as previous injuries or specific medical conditions. Age is a major contributing factor to OA, and as one ages, various joint tissues undergo gradual change, including degeneration of the articular cartilage, alterations in subchondral bone (SCB) morphology, and inflammation of the synovium. Methods We investigated the prevalence of primary OA in aged, genetically diverse UM-HET3 mice. Articular cartilage (AC) integrity and SCB morphology were assessed in 182 knee joints of 22-25 months old mice using the Osteoarthritis Research Society International (OARSI) scoring system and micro-CT, respectively. Additionally, we explored the effects of methylene blue (MB) and mitoquinone (MitoQ), two agents that affect mitochondrial function, on the prevalence and progression of OA during aging. Results Aged UM-HET3 mice showed a high prevalence of primary OA in both sexes. Significant positive correlations were found between cumulative AC (cAC) scores and synovitis in both sexes, and osteophyte formation in female mice. Ectopic chondrogenesis did not show significant correlations with cAC scores. Significant direct correlations were found between AC scores and inflammatory markers in chondrocytes, including matrix metalloproteinase-13, inducible nitric oxide synthase, and the NLR family pyrin domain containing-3 inflammasome in both sexes, indicating a link between OA severity and inflammation. Additionally, markers of cell cycle arrest, such as p16 and β-galactosidase, also correlated with AC scores. In male mice, no significant correlations were found between SCB morphology traits and cAC scores, while in female mice, significant correlations were found between cAC scores and tibial SCB plate bone mineral density. Notably, MB and MitoQ treatments influenced the disease's progression in a sex-specific manner. MB treatment significantly reduced cAC scores at the medial knee joint, while MitoQ treatment reduced cAC scores, but these did not reach significance. Conclusions Our study provides comprehensive insights into the prevalence and progression of primary OA in aged UM-HET3 mice, highlighting the sex-specific effects of MB and MitoQ treatments. The correlations between AC scores and various pathological factors underscore the multifaceted nature of OA and its association with inflammation and subchondral bone changes.
Collapse
Affiliation(s)
- Sher Bahadur Poudel
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| | - Ryan R Ruff
- David B. Kriser Dental Center, Biostatistics Core, Department of Epidemiology and Health Promotion, New York University College of Dentistry New York, NY 10010-4086
| | - Gozde Yildirim
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | | | - Randy Strong
- Geriatric Research, Education and Clinical Center and Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA; Barshop Institute for Longevity and Aging Studies and Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Thorsten Kirsch
- Department of Orthopaedic Surgery, NYU Grossman School of Medicine, and Department of Biomedical Engineering, NYU Tandon School of Engineering, New York, NY
| | - Shoshana Yakar
- David B. Kriser Dental Center, Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY
| |
Collapse
|
6
|
Poudel SB, Ruff RR, Yildirim G, Miller RA, Harrison DE, Strong R, Kirsch T, Yakar S. Development of primary osteoarthritis during aging in genetically diverse UM-HET3 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.16.571693. [PMID: 38168298 PMCID: PMC10760163 DOI: 10.1101/2023.12.16.571693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
This study investigated the prevalence and progression of primary osteoarthritis (OA) in aged UM-HET3 mice. Using the Osteoarthritis Research Society International (OARSI) scoring system, we assessed articular cartilage (AC) integrity in 182 knee joints of 22-25 months old mice. Aged UM-HET3 mice showed a high prevalence of primary OA in both sexes. Significant positive correlations were found between cumulative AC (cAC) scores and synovitis in both sexes, and osteophyte formation in female mice. Ectopic chondrogenesis did not show significant correlations with cAC scores. Significant direct correlations were found between AC scores and inflammatory markers in chondrocytes, including matrix metalloproteinase-13 (MMP-13), inducible nitric oxide synthase (iNOS), and the NLR family pyrin domain containing-3 (NLRP3) inflammasome in both sexes, indicating a link between OA severity and inflammation. Additionally, markers of cell cycle arrest, such as p16 and β-galactosidase, also correlated with AC scores. Using micro-CT, we examined the correlations between subchondral bone (SCB) morphology traits and AC scores. In male mice, no significant correlations were found between SCB morphology traits and cAC scores, while in female mice, significant correlations were found between cAC scores and tibial SCB plate bone mineral density. Finally, we explored the effects of methylene blue (MB) and mitoquinone (MitoQ), two agents that affect mitochondrial function, on the prevalence and progression of OA during aging. Notably, MB and MitoQ treatments influenced the disease's progression in a sex-specific manner. MB treatment significantly reduced cAC scores at the medial knee joint, while MitoQ treatment reduced cAC scores, but these did not reach significance. In conclusion, our study provides comprehensive insights into the prevalence and progression of primary OA in aged UM-HET3 mice, highlighting the sex-specific effects of MB and MitoQ treatments. The correlations between AC scores and various pathological factors underscore the multifaceted nature of OA and its association with inflammation and subchondral bone changes.
Collapse
|
7
|
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
|
8
|
Hou Z, Mo F, Zhou Q. Elucidating response mechanisms at the metabolic scale of Eisenia fetida in typical oil pollution sites: A native driver in influencing carbon flow. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122545. [PMID: 37716696 DOI: 10.1016/j.envpol.2023.122545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/07/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
Previous investigations on the stress response patterns of earthworms (Eisenia fetida) in practical petroleum hydrocarbon (PH) contamination systems were less focused. Therefore, this study investigated the ecotoxicological effect of PH contamination on earthworms based on metabonomics and histological observation, followed by correlation analysis between the earthworm metabolism, PH types and concentrations, soil physicochemical characteristics, and the microbial community structures (i.e., diversity and abundance) and functions. The results showed that due to the abundant PH organics, the cell metabolism of earthworms shifts under PH contamination conditions, leading them to use organic acids as alternative energy sources (i.e., gluconeogenesis pathway). Simultaneously, biomarker metabolites related to cellular uptake, stress response, and membrane disturbance were identified. In addition, when compared to the controls, considerable epicuticle and cuticle layer disruption was observed, along with PH internalization. It was demonstrated that PH pollution preferentially influences the physiological homeostasis of earthworms through indirect (i.e., microbial metabolism regulation) than direct (i.e., direct interaction with earthworms) mechanisms. Moreover, the varied CO2 releasement was verified, which highlights the potential role of earthworms in influencing carbon transformation and corresponds with the considerably enriched energy metabolism-related pathway. This study indicated that PH contamination can induce a strong stress response in earthworms through both direct and indirect mechanisms, which in turn, substantially influences carbon transformation in PH contamination sites.
Collapse
Affiliation(s)
- Zelin Hou
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, Carbon Neutrality Interdisciplinary Science Centre/College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Fan Mo
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, Carbon Neutrality Interdisciplinary Science Centre/College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Qixing Zhou
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria/Tianjin Key Laboratory of Environmental Remediation and Pollution Control, Carbon Neutrality Interdisciplinary Science Centre/College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China.
| |
Collapse
|
9
|
Mechanotransduction pathways in articular chondrocytes and the emerging role of estrogen receptor-α. Bone Res 2023; 11:13. [PMID: 36869045 PMCID: PMC9984452 DOI: 10.1038/s41413-023-00248-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/05/2022] [Accepted: 01/06/2023] [Indexed: 03/05/2023] Open
Abstract
In the synovial joint, mechanical force creates an important signal that influences chondrocyte behavior. The conversion of mechanical signals into biochemical cues relies on different elements in mechanotransduction pathways and culminates in changes in chondrocyte phenotype and extracellular matrix composition/structure. Recently, several mechanosensors, the first responders to mechanical force, have been discovered. However, we still have limited knowledge about the downstream molecules that enact alterations in the gene expression profile during mechanotransduction signaling. Recently, estrogen receptor α (ERα) has been shown to modulate the chondrocyte response to mechanical loading through a ligand-independent mechanism, in line with previous research showing that ERα exerts important mechanotransduction effects on other cell types, such as osteoblasts. In consideration of these recent discoveries, the goal of this review is to position ERα into the mechanotransduction pathways known to date. Specifically, we first summarize our most recent understanding of the mechanotransduction pathways in chondrocytes on the basis of three categories of actors, namely mechanosensors, mechanotransducers, and mechanoimpactors. Then, the specific roles played by ERα in mediating the chondrocyte response to mechanical loading are discussed, and the potential interactions of ERα with other molecules in mechanotransduction pathways are explored. Finally, we propose several future research directions that may advance our understanding of the roles played by ERα in mediating biomechanical cues under physiological and pathological conditions.
Collapse
|
10
|
Segarra-Queralt M, Piella G, Noailly J. Network-based modelling of mechano-inflammatory chondrocyte regulation in early osteoarthritis. Front Bioeng Biotechnol 2023; 11:1006066. [PMID: 36815875 PMCID: PMC9936426 DOI: 10.3389/fbioe.2023.1006066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 01/16/2023] [Indexed: 02/05/2023] Open
Abstract
Osteoarthritis (OA) is a debilitating joint disease characterized by articular cartilage degradation, inflammation and pain. An extensive range of in vivo and in vitro studies evidences that mechanical loads induce changes in chondrocyte gene expression, through a process known as mechanotransduction. It involves cascades of complex molecular interactions that convert physical signals into cellular response(s) that favor either chondroprotection or cartilage destruction. Systematic representations of those interactions can positively inform early strategies for OA management, and dynamic modelling allows semi-quantitative representations of the steady states of complex biological system according to imposed initial conditions. Yet, mechanotransduction is rarely integrated. Hence, a novel mechano-sensitive network-based model is proposed, in the form of a continuous dynamical system: an interactome of a set of 118 nodes, i.e., mechano-sensitive cellular receptors, second messengers, transcription factors and proteins, related among each other through a specific topology of 358 directed edges is developed. Results show that under physio-osmotic initial conditions, an anabolic state is reached, whereas initial perturbations caused by pro-inflammatory and injurious mechanical loads leads to a catabolic profile of node expression. More specifically, healthy chondrocyte markers (Sox9 and CITED2) are fully expressed under physio-osmotic conditions, and reduced under inflammation, or injurious loadings. In contrast, NF-κB and Runx2, characteristic of an osteoarthritic chondrocyte, become activated under inflammation or excessive loading regimes. A literature-based evaluation shows that the model can replicate 94% of the experiments tested. Sensitivity analysis based on a factorial design of a treatment shows that inflammation has the strongest influence on chondrocyte metabolism, along with a significant deleterious effect of static compressive loads. At the same time, anti-inflammatory therapies appear as the most promising ones, though the restoration of structural protein production seems to remain a major challenge even in beneficial mechanical environments. The newly developed mechano-sensitive network model for chondrocyte activity reveals a unique potential to reflect load-induced chondroprotection or articular cartilage degradation in different mechano-chemical-environments.
Collapse
|
11
|
Li Y, Chen G. Upconversion Nanoparticles for Cancer Therapy. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Yang Li
- School of Chemistry and Chemical Engineering Harbin Institute of Technology Harbin 150001 China
| | - Guanying Chen
- School of Chemistry and Chemical Engineering Harbin Institute of Technology Harbin 150001 China
| |
Collapse
|
12
|
Silva Santos Ribeiro P, Willemen HLDM, Eijkelkamp N. Mitochondria and sensory processing in inflammatory and neuropathic pain. FRONTIERS IN PAIN RESEARCH 2022; 3:1013577. [PMID: 36324872 PMCID: PMC9619239 DOI: 10.3389/fpain.2022.1013577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/26/2022] [Indexed: 01/24/2023] Open
Abstract
Rheumatic diseases, such as osteoarthritis and rheumatoid arthritis, affect over 750 million people worldwide and contribute to approximately 40% of chronic pain cases. Inflammation and tissue damage contribute to pain in rheumatic diseases, but pain often persists even when inflammation/damage is resolved. Mechanisms that cause this persistent pain are still unclear. Mitochondria are essential for a myriad of cellular processes and regulate neuronal functions. Mitochondrial dysfunction has been implicated in multiple neurological disorders, but its role in sensory processing and pain in rheumatic diseases is relatively unexplored. This review provides a comprehensive understanding of how mitochondrial dysfunction connects inflammation and damage-associated pathways to neuronal sensitization and persistent pain. To provide an overall framework on how mitochondria control pain, we explored recent evidence in inflammatory and neuropathic pain conditions. Mitochondria have intrinsic quality control mechanisms to prevent functional deficits and cellular damage. We will discuss the link between neuronal activity, mitochondrial dysfunction and chronic pain. Lastly, pharmacological strategies aimed at reestablishing mitochondrial functions or boosting mitochondrial dynamics as therapeutic interventions for chronic pain are discussed. The evidence presented in this review shows that mitochondria dysfunction may play a role in rheumatic pain. The dysfunction is not restricted to neuronal cells in the peripheral and central nervous system, but also includes blood cells and cells at the joint level that may affect pain pathways indirectly. Pre-clinical and clinical data suggest that modulation of mitochondrial functions can be used to attenuate or eliminate pain, which could be beneficial for multiple rheumatic diseases.
Collapse
Affiliation(s)
| | | | - Niels Eijkelkamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Kan S, Duan M, Liu Y, Wang C, Xie J. Role of Mitochondria in Physiology of Chondrocytes and Diseases of Osteoarthritis and Rheumatoid Arthritis. Cartilage 2021; 13:1102S-1121S. [PMID: 34894777 PMCID: PMC8804744 DOI: 10.1177/19476035211063858] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE OF REVIEW Mitochondria are recognized to be one of the most important organelles in chondrocytes for their role in triphosphate (ATP) generation through aerobic phosphorylation. Mitochondria also participate in many intracellular processes involving modulating reactive oxygen species (ROS), responding to instantaneous hypoxia stress, regulating cytoplasmic transport of calcium ion, and directing mitophagy to maintain the homeostasis of individual chondrocytes. DESIGNS To summarize the specific role of mitochondria in chondrocytes, we screened related papers in PubMed database and the search strategy is ((mitochondria) AND (chondrocyte)) AND (English [Language]). The articles published in the past 5 years were included and 130 papers were studied. RESULTS In recent years, the integrity of mitochondrial structure has been regarded as a prerequisite for normal chondrocyte survival and defect in mitochondrial function has been found in cartilage-related diseases, such as osteoarthritis (OA) and rheumatoid arthritis (RA). However, the understanding of mitochondria in cartilage is still largely limited. The mechanism on how the changes in mitochondrial structure and function directly lead to the occurrence and development of cartilage-related diseases remains to be elusive. CONCLUSION This review aims to summarize the role of mitochondria in chondrocytes under the physiological and pathological changes from ATP generation, calcium homeostasis, redox regulation, mitophagy modulation, mitochondria biogenesis to immune response activation. The enhanced understanding of molecular mechanisms in mitochondria might offer some new cues for cartilage remodeling and pathological intervention.
Collapse
Affiliation(s)
- Shiyi Kan
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mengmeng Duan
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Liu
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chunli Wang
- “111” Project Laboratory of
Biomechanics and Tissue Repair, Bioengineering College, Chongqing University,
Chongqing, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases,
West China Hospital of Stomatology, Sichuan University, Chengdu, China,“111” Project Laboratory of
Biomechanics and Tissue Repair, Bioengineering College, Chongqing University,
Chongqing, China,Lab of Bone & Joint Disease, State
Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan
University, Chengdu, China,Jing Xie, Lab of Bone & Joint Disease,
State Key Laboratory of Oral Diseases, West China Hospital of Stomatology,
Sichuan University, Chengdu 610064, Sichuan, China.
| |
Collapse
|
16
|
Quarterman JC, Naguib YW, Chakka JL, Seol D, Martin JA, Salem AK. HPLC-UV Method Validation for Amobarbital and Pharmaceutical Stability Evaluation When Dispersed in a Hyaluronic Acid Hydrogel: A New Concept for Post-Traumatic Osteoarthritis Prevention. J Pharm Sci 2021; 111:1379-1390. [PMID: 34563533 DOI: 10.1016/j.xphs.2021.09.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/17/2021] [Accepted: 09/17/2021] [Indexed: 11/17/2022]
Abstract
A mitochondrial electron transport chain member complex I inhibitor, amobarbital, can reduce oxidative damage and chondrocyte death, eventually preventing post-traumatic osteoarthritis (PTOA). Viscosupplementation using a crosslinked hyaluronic acid (HA) hydrogel is currently applied clinically for knee OA pain relief. In this work, we utilized the HA hydrogel as a drug delivery vehicle to improve the long-term efficacy of amobarbital. Here we evaluated the pharmaceutic stability of amobarbital when dispersed in a crosslinked HA hydrogel formulated in proportions intended for clinical use. We validated a high-performance liquid chromatography with an ultraviolet detector (HPLC-UV) method following International Conference for Harmonization Q2(R1) guidelines to ensure its suitability for amobarbital detection. The feasibility of this formulation's drug delivery capability was proven by measuring the release, solubility, and drug uniformity. The amobarbital/HA hydrogel showed comparable amobarbital stability in different biological fluids compared to amobarbital solution. In addition, the amobarbital/HA hydrogel imparted significantly greater drug stability when stored at 70°C for 24 hours. In conclusion, we confirmed the pharmaceutical stability of the amobarbital/HA hydrogel in various conditions and biological fluids using a validated HPLC-UV method. This data provides essential evidence in support of the use of this amobarbital/HA formulation in future clinical trials for PTOA treatment.
Collapse
Affiliation(s)
- Juliana C Quarterman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 115 S Grand Avenue, 201 Pharmacy Building, Iowa City, IA 52242, USA
| | - Youssef W Naguib
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 115 S Grand Avenue, 201 Pharmacy Building, Iowa City, IA 52242, USA; Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Jaidev L Chakka
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 115 S Grand Avenue, 201 Pharmacy Building, Iowa City, IA 52242, USA
| | - Dongrim Seol
- Department of Orthopedics and Rehabilitation, College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - James A Martin
- Department of Orthopedics and Rehabilitation, College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, 115 S Grand Avenue, 201 Pharmacy Building, Iowa City, IA 52242, USA.
| |
Collapse
|
17
|
Tudorachi NB, Totu EE, Fifere A, Ardeleanu V, Mocanu V, Mircea C, Isildak I, Smilkov K, Cărăuşu EM. The Implication of Reactive Oxygen Species and Antioxidants in Knee Osteoarthritis. Antioxidants (Basel) 2021; 10:985. [PMID: 34205576 PMCID: PMC8233827 DOI: 10.3390/antiox10060985] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/11/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022] Open
Abstract
Knee osteoarthritis (KOA) is a chronic multifactorial pathology and a current and essential challenge for public health, with a negative impact on the geriatric patient's quality of life. The pathophysiology is not fully known; therefore, no specific treatment has been found to date. The increase in the number of newly diagnosed cases of KOA is worrying, and it is essential to reduce the risk factors and detect those with a protective role in this context. The destructive effects of free radicals consist of the acceleration of chondrosenescence and apoptosis. Among other risk factors, the influence of redox imbalance on the homeostasis of the osteoarticular system is highlighted. The evolution of KOA can be correlated with oxidative stress markers or antioxidant status. These factors reveal the importance of maintaining a redox balance for the joints and the whole body's health, emphasizing the importance of an individualized therapeutic approach based on antioxidant effects. This paper aims to present an updated picture of the implications of reactive oxygen species (ROS) in KOA from pathophysiological and biochemical perspectives, focusing on antioxidant systems that could establish the premises for appropriate treatment to restore the redox balance and improve the condition of patients with KOA.
Collapse
Affiliation(s)
- Nicoleta Bianca Tudorachi
- Faculty of Medicine, “Ovidius” University of Constanța, Mamaia Boulevard 124, 900527 Constanța, Romania; (N.B.T.); (V.A.)
| | - Eugenia Eftimie Totu
- Faculty of Applied Chemistry and Material Science, University Politehnica of Bucharest, 1–5 Polizu Street, 011061 Bucharest, Romania
| | - Adrian Fifere
- Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, “Petru Poni” Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487 Iasi, Romania
| | - Valeriu Ardeleanu
- Faculty of Medicine, “Ovidius” University of Constanța, Mamaia Boulevard 124, 900527 Constanța, Romania; (N.B.T.); (V.A.)
| | - Veronica Mocanu
- Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (V.M.); (C.M.)
| | - Cornelia Mircea
- Faculty of Pharmacy, Grigore T. Popa University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (V.M.); (C.M.)
| | - Ibrahim Isildak
- Faculty of Chemistry-Metallurgy, Department of Bioengineering, Yildiz Technical University, Istanbul 34220, Turkey;
| | - Katarina Smilkov
- Faculty of Medical Sciences, Division of Pharmacy, Department of Applied Pharmacy, Goce Delcev University, Krste Misirkov Street, No. 10-A, 2000 Stip, North Macedonia;
| | - Elena Mihaela Cărăuşu
- Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, “Nicolae Leon” Building, 13 Grigore Ghica Street, 700259 Iasi, Romania;
| |
Collapse
|
18
|
Osteocyte Dysfunction in Joint Homeostasis and Osteoarthritis. Int J Mol Sci 2021; 22:ijms22126522. [PMID: 34204587 PMCID: PMC8233862 DOI: 10.3390/ijms22126522] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 01/29/2023] Open
Abstract
Structural disturbances of the subchondral bone are a hallmark of osteoarthritis (OA), including sclerotic changes, cystic lesions, and osteophyte formation. Osteocytes act as mechanosensory units for the micro-cracks in response to mechanical loading. Once stimulated, osteocytes initiate the reparative process by recruiting bone-resorbing cells and bone-forming cells to maintain bone homeostasis. Osteocyte-expressed sclerostin is known as a negative regulator of bone formation through Wnt signaling and the RANKL pathway. In this review, we will summarize current understandings of osteocytes at the crossroad of allometry and mechanobiology to exploit the relationship between osteocyte morphology and function in the context of joint aging and osteoarthritis. We also aimed to summarize the osteocyte dysfunction and its link with structural and functional disturbances of the osteoarthritic subchondral bone at the molecular level. Compared with normal bones, the osteoarthritic subchondral bone is characterized by a higher bone volume fraction, a larger trabecular bone number in the load-bearing region, and an increase in thickness of pre-existing trabeculae. This may relate to the aberrant expressions of sclerostin, periostin, dentin matrix protein 1, matrix extracellular phosphoglycoprotein, insulin-like growth factor 1, and transforming growth factor-beta, among others. The number of osteocyte lacunae embedded in OA bone is also significantly higher, yet the volume of individual lacuna is relatively smaller, which could suggest abnormal metabolism in association with allometry. The remarkably lower percentage of sclerostin-positive osteocytes, together with clustering of Runx-2 positive pre-osteoblasts, may suggest altered regulation of osteoblast differentiation and osteoblast-osteocyte transformation affected by both signaling molecules and the extracellular matrix. Aberrant osteocyte morphology and function, along with anomalies in molecular signaling mechanisms, might explain in part, if not all, the pre-osteoblast clustering and the uncoupled bone remodeling in OA subchondral bone.
Collapse
|
19
|
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: 25] [Impact Index Per Article: 8.3] [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
|
20
|
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
|
21
|
Mitochondrial DNA from osteoarthritic patients drives functional impairment of mitochondrial activity: a study on transmitochondrial cybrids. Cytotherapy 2021; 23:399-410. [PMID: 33727013 DOI: 10.1016/j.jcyt.2020.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 06/05/2020] [Accepted: 08/20/2020] [Indexed: 11/23/2022]
Abstract
With the redefinition of osteoarthritis (OA) and the understanding that the joint behaves as an organ, OA is now considered a systemic illness with a low grade of chronic inflammation. Mitochondrial dysfunction is well documented in OA and has the capacity to alter chondrocyte and synoviocyte function. Transmitochondrial cybrids are suggested as a useful cellular model to study mitochondrial biology in vitro, as they carry different mitochondrial variants with the same nuclear background. The aim of this work was to study mitochondrial and metabolic function of cybrids with mitochondrial DNA from healthy (N) and OA donors. In this work, the authors demonstrate that cybrids from OA patients behave differently from cybrids from N donors in several mitochondrial parameters. Furthermore, OA cybrids behave similarly to OA chondrocytes. These results enhance our understanding of the role of mitochondria in the degeneration process of OA and present cybrids as a useful model to study OA pathogenesis.
Collapse
|
22
|
Celik C, Franco-Obregón A, Lee EH, Hui JH, Yang Z. Directionalities of magnetic fields and topographic scaffolds synergise to enhance MSC chondrogenesis. Acta Biomater 2021; 119:169-183. [PMID: 33130304 DOI: 10.1016/j.actbio.2020.10.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/15/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cell (MSC) chondrogenesis is modulated by diverse biophysical cues. We have previously shown that brief, low-amplitude pulsed electromagnetic fields (PEMFs) differentially enhance MSC chondrogenesis in scaffold-free pellet cultures versus conventional tissue culture plastic (TCP), indicating an interplay between magnetism and micromechanical environment. Here, we examined the influence of PEMF directionality over the chondrogenic differentiation of MSCs laden on electrospun fibrous scaffolds of either random (RND) or aligned (ALN) orientations. Correlating MSCs' chondrogenic outcome to pFAK activation and YAP localisation, MSCs on the RND scaffolds experienced the least amount of resting mechanical stress and underwent greatest chondrogenic differentiation in response to brief PEMF exposure (10 min at 1 mT) perpendicular to the dominant plane of the scaffolds (Z-directed). By contrast, in MSC-impregnated RND scaffolds, greatest mitochondrial respiration resulted from X-directed PEMF exposure (parallel to the scaffold plane), and was associated with curtailed chondrogenesis. MSCs on TCP or the ALN scaffolds exhibited greater resting mechanical stress and accordingly, were unresponsive, or negatively responsive, to PEMF exposure from all directions. The efficacy of PEMF-induced MSC chondrogenesis is hence regulated in a multifaceted manner involving focal adhesion dynamics, as well as mitochondrial responses, culminating in a final cellular response. The combined contributions of micromechanical environment and magnetic field orientation hence will need to be considered when designing magnetic exposure paradigms.
Collapse
Affiliation(s)
- Cenk Celik
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228
| | - Alfredo Franco-Obregón
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228; BioIonic Currents Electromagnetic Pulsing Systems Laboratory, BICEPS, National University of Singapore, Singapore, 117599; Institute for Health Innovation & Technology, iHealthtech, National University of Singapore, Singapore, 117599; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117593.
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228; Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510
| | - James Hp Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228; Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228; Tissue Engineering Program (NUSTEP), Life Sciences Institute, National University of Singapore, DSO (Kent Ridge) Building, #04-01, 27 Medical Drive, Singapore, 117510.
| |
Collapse
|
23
|
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: 4.8] [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
|
24
|
He Y, Wu Z, Xu L, Xu K, Chen Z, Ran J, Wu L. The role of SIRT3-mediated mitochondrial homeostasis in osteoarthritis. Cell Mol Life Sci 2020; 77:3729-3743. [PMID: 32468094 PMCID: PMC11105031 DOI: 10.1007/s00018-020-03497-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 02/07/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
Osteoarthritis is the most common degenerative joint disease and causes major pain and disability in adults. It has been reported that mitochondrial dysfunction in chondrocytes is associated with osteoarthritis. Sirtuins are a family of nicotinamide adenine dinucleotide-dependent histone deacetylases that have the ability to deacetylate protein targets and play an important role in the regulation of cell physiological and pathological processes. Among sirtuin family members, sirtuin 3, which is mainly located in mitochondria, can exert its deacetylation activity to regulate mitochondrial function, regeneration, and dynamics; these processes are presently recognized to maintain redox homeostasis to prevent oxidative stress in cell metabolism. In this review, we provide present opinions on the effect of mitochondrial dysfunction in osteoarthritis. Furthermore, the potential protective mechanism of SIRT3-mediated mitochondrial homeostasis in the progression of osteoarthritis is discussed.
Collapse
Affiliation(s)
- Yuzhe He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhipeng Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Langhai Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhonggai Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jisheng Ran
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lidong Wu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
25
|
Crisol M, Wu K, Laouar L, Elliott JAW, Jomha NM. Antioxidant additives reduce reactive oxygen species production in articular cartilage during exposure to cryoprotective agents. Cryobiology 2020; 96:114-121. [PMID: 32777334 DOI: 10.1016/j.cryobiol.2020.07.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023]
Abstract
High concentrations of cryoprotective agents (CPA) are required during articular cartilage cryopreservation but these CPAs can be toxic to chondrocytes. Reactive oxygen species have been linked to cell death due to oxidative stress. Addition of antioxidants has shown beneficial effects on chondrocyte survival and functions after cryopreservation. The objectives of this study were to investigate (1) oxidative stress experienced by chondrocytes and (2) the effect of antioxidants on cellular reactive oxygen species production during articular cartilage exposure to high concentrations of CPAs. Porcine cartilage dowels were exposed to a multi-CPA solution supplemented with either 0.1 mg/mL chondroitin sulfate or 2000 μM ascorbic acid, at 4 °C for 180 min (N = 7). Reactive oxygen species production was measured with 5 μM dihydroethidium, a fluorescent probe that targets reactive oxygen species. The cell viability was quantified with a dual cell membrane integrity stain containing 6.25 μM Syto 13 + 9 μM propidium iodide using confocal microscopy. Supplementation of CPA solutions with chondroitin sulfate or ascorbic acid resulted in significantly lower dihydroethidium counts (p < 0.01), and a lower decrease in the percentage of viable cells (p < 0.01) compared to the CPA-treated group without additives. These results indicated that reactive oxygen species production is induced when articular cartilage is exposed to high CPA concentrations, and correlated with the amount of dead cells. Both chondroitin sulfate and ascorbic acid treatments significantly reduced reactive oxygen species production and improved chondrocyte viability when articular cartilage was exposed to high concentrations of CPAs.
Collapse
Affiliation(s)
- Mary Crisol
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Kezhou Wu
- Department of Surgery, University of Alberta, Edmonton, AB, Canada; Department of Orthopedic Surgery, First Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, China
| | - Leila Laouar
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Janet A W Elliott
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Nadr M Jomha
- Department of Surgery, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
26
|
Insulin Resistance in Osteoarthritis: Similar Mechanisms to Type 2 Diabetes Mellitus. J Nutr Metab 2020; 2020:4143802. [PMID: 32566279 PMCID: PMC7261331 DOI: 10.1155/2020/4143802] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/01/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) and type 2 diabetes mellitus (T2D) are two of the most widespread chronic diseases. OA and T2D have common epidemiologic traits, are considered heterogenic multifactorial pathologies that develop through the interaction of genetic and environmental factors, and have common risk factors. In addition, both of these diseases often manifest in a single patient. Despite differences in clinical manifestations, both diseases are characterized by disturbances in cellular metabolism and by an insulin-resistant state primarily associated with the production and utilization of energy. However, currently, the primary cause of OA development and progression is not clear. In addition, although OA is manifested as a joint disease, evidence has accumulated that it affects the whole body. As pathological insulin resistance is viewed as a driving force of T2D development, now, we present evidence that the molecular and cellular metabolic disturbances associated with OA are linked to an insulin-resistant state similar to T2D. Moreover, the alterations in cellular energy requirements associated with insulin resistance could affect many metabolic changes in the body that eventually result in pathology and could serve as a unified mechanism that also functions in many metabolic diseases. However, these issues have not been comprehensively described. Therefore, here, we discuss the basic molecular mechanisms underlying the pathological processes associated with the development of insulin resistance; the major inducers, regulators, and metabolic consequences of insulin resistance; and instruments for controlling insulin resistance as a new approach to therapy.
Collapse
|
27
|
Pathomechanisms of Posttraumatic Osteoarthritis: Chondrocyte Behavior and Fate in a Precarious Environment. Int J Mol Sci 2020; 21:ijms21051560. [PMID: 32106481 PMCID: PMC7084733 DOI: 10.3390/ijms21051560] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/18/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
Traumatic injuries of the knee joint result in a wide variety of pathomechanisms, which contribute to the development of so-called posttraumatic osteoarthritis (PTOA). These pathogenetic processes include oxidative stress, excessive expression of catabolic enzymes, release of damage-associated molecular patterns (DAMPs), and synovial inflammation. The present review focuses on the underlying pathomechanisms of PTOA and in particular the behavior and fate of the surviving chondrocytes, comprising chondrocyte metabolism, regulated cell death, and phenotypical changes comprising hypertrophy and senescence. Moreover, possible therapeutic strategies, such as chondroanabolic stimulation, anti-oxidative and anti-inflammatory treatment, as well as novel therapeutic targets are discussed.
Collapse
|
28
|
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: 0.8] [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
|
29
|
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: 69] [Impact Index Per Article: 13.8] [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
|
30
|
Osteoarthritis year in review 2018: biology. Osteoarthritis Cartilage 2019; 27:365-370. [PMID: 30808484 DOI: 10.1016/j.joca.2018.10.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/05/2018] [Accepted: 10/11/2018] [Indexed: 02/02/2023]
Abstract
This Year in Review highlights a selection of articles published between the 2017 and 2018 Osteoarthritis Research Society International (OARSI) World Congress meetings within the field of osteoarthritis biology, presented at OARSI 2018. Selected articles were obtained from a PubMed search covering cartilage, subchondral bone, inflammation, ageing, pain and animal models. Studies focused on biomechanics, biomarkers, genetics and epigenetics, imaging and clinical studies were excluded due to their coverage in other articles within the OARSI Year in Review series. Significant themes including the role of progenitor cells in cartilage homeostasis and repair, novel signalling mechanisms controlling chondrocyte phenotypic stability and the influence of disrupted or senescent chondrocytes were identified and are discussed in this review. Overarching conclusions derived from these study areas indicate that promising avenues of intervention are on the horizon, however further understanding is required in order to target therapeutic treatments to suitable patient subgroups and disease stages.
Collapse
|
31
|
Mechanosensitive channels and their functions in stem cell differentiation. Exp Cell Res 2018; 374:259-265. [PMID: 30500393 DOI: 10.1016/j.yexcr.2018.11.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/17/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022]
Abstract
Stem cells continuously perceive and respond to various environmental signals during development, tissue homeostasis, and pathological conditions. Mechanical force, one of the fundamental signals in the physical world, plays a vital role in the regulation of multiple functions of stem cells. The importance of cell adhesion to the extracellular matrix (ECM), cell-cell junctions, and a mechanoresponsive cell cytoskeleton has been under intensive study in the fields of stem cell biology and mechanobiology. However, the involvement of mechanosensitive (MS) ion channels in the mechanical regulation of stem cell activity has just begun to be realized. Here, we review the diversity and importance of mechanosensitive channels (MSCs), and discuss recently discovered functions of MSCs in stem cell regulation, especially in the determination of cell fate.
Collapse
|
32
|
Koike M, Nojiri H, Kanazawa H, Yamaguchi H, Miyagawa K, Nagura N, Banno S, Iwase Y, Kurosawa H, Kaneko K. Superoxide dismutase activity is significantly lower in end-stage osteoarthritic cartilage than non-osteoarthritic cartilage. PLoS One 2018; 13:e0203944. [PMID: 30222787 PMCID: PMC6141073 DOI: 10.1371/journal.pone.0203944] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 08/30/2018] [Indexed: 12/31/2022] Open
Abstract
Recent studies have shown that superoxide dismutase 1 (SOD1), SOD2, and SOD3 are significantly decreased in human osteoarthritic cartilage. SOD activity is a marker that can be used to comprehensively evaluate the enzymatic capacities of SOD1, SOD2, and SOD3; however, the trend of SOD activity in end-stage osteoarthritic tissues remains unknown. In the present study, we found that SOD activity in end-stage osteoarthritic synovium of the knee was significantly lower than that in control synovium without the influence of age. The SOD activity was significantly lower in the end-stage knee osteoarthritic cartilage than in the control, but a weak negative correlation was observed between aging and SOD activity. However, SOD activity in end-stage hip osteoarthritic cartilage was significantly lower than that in control cartilage without the influence of aging. The relationship between osteoarthritis and SOD activity was stronger than the relationship between aging and SOD activity. These results indicate that direct regulation of SOD activity in joint tissues may lead to suppression of osteoarthritis progression.
Collapse
Affiliation(s)
- Masato Koike
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Hidetoshi Nojiri
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
- * E-mail:
| | - Hiroaki Kanazawa
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Hiroto Yamaguchi
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Kei Miyagawa
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Nana Nagura
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Sammy Banno
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Yoshiyuki Iwase
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Hisashi Kurosawa
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo, Japan
| | - Kazuo Kaneko
- Department of Orthopaedic Surgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
33
|
Seol D, Tochigi Y, Bogner AM, Song I, Fredericks DC, Kurriger GL, Smith SM, Goetz JE, Buckwalter JA, Martin JA. Effects of knockout of the receptor for advanced glycation end-products on bone mineral density and synovitis in mice with intra-articular fractures. J Orthop Res 2018; 36:2439-2449. [PMID: 29667227 PMCID: PMC6128287 DOI: 10.1002/jor.24021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 04/12/2018] [Indexed: 02/04/2023]
Abstract
Our group employed the mouse closed intra-articular fracture (IAF) model to test the hypothesis that the innate immune system plays a role in initiating synovitis and post-traumatic osteoarthritis (PTOA) in fractured joints. A transgenic strategy featuring knockout of the receptor for advanced glycation end-products (RAGE -/- ) was pursued. The 42 and 84 mJ impacts used to create fractures were in the range previously reported to cause PTOA at 60 days post-fracture. MicroCT (μCT) was used to assess fracture patterns and epiphyseal and metaphyseal bone loss at 30 and 60 days post-fracture. Cartilage degeneration, synovitis, and matrix metalloproteinase (MMP-3, -13) expression were evaluated by histologic analyses. In wild-type mice, μCT imaging showed that 84 mJ impacts led to significant bone loss at 30 days (p < 0.05), but recovered to normal at 60 days. Bone losses did not occur in RAGE-/- mice. Synovitis was significantly elevated in 84 mJ impact wild-type mice at both endpoints (30 day, p = 0.001; 60 day, p = 0.05), whereas in RAGE-/- mice synovitis was elevated only at 30 days (p = 0.02). Mankin scores were slightly elevated in both mouse strains at 30 days, but not at 60 days. Immunohistochemistry revealed significant fracture-related increases in MMP-3 and -13 expression at 30 days (p < 0.05), with no significant difference between genotypes. These findings indicated that while RAGE -/- accelerated recovery from fracture and diminished synovitis, arthritic changes were temporary and too modest to detect an effect on the pathogenesis of PTOA. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:2439-2449, 2018.
Collapse
Affiliation(s)
- Dongrim Seol
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
| | - Yuki Tochigi
- Department of Orthopaedic Surgery, Dokkyo Medical University Koshigaya Hospital, Saitama, Japan
| | - Ashley M. Bogner
- Bone Healing Research Laboratory and Iowa Spine Research Laboratory, University of Iowa, Coralville, IA 52241, USA
| | - Ino Song
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA,Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Douglas C. Fredericks
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA,Bone Healing Research Laboratory and Iowa Spine Research Laboratory, University of Iowa, Coralville, IA 52241, USA
| | - Gail L. Kurriger
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
| | - Sonja M. Smith
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
| | - Jessica E. Goetz
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
| | - Joseph A. Buckwalter
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA,Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | - James A. Martin
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA,Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA,Correspondence to James A. Martin (Address: 1182 ML, University of Iowa, Iowa City, IA 52242; T: +1-319-335-5810; F: +1-319-335-5631; )
| |
Collapse
|
34
|
Issa R, Boeving M, Kinter M, Griffin TM. Effect of biomechanical stress on endogenous antioxidant networks in bovine articular cartilage. J Orthop Res 2018; 36:760-769. [PMID: 28892196 PMCID: PMC5839935 DOI: 10.1002/jor.23728] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/31/2017] [Indexed: 02/04/2023]
Abstract
Mechanosensitve pathways in chondrocytes are essential for maintaining articular cartilage homeostasis. Traumatic loading increases cartilage oxidation and causes cell death and osteoarthritis. However, sub-lethal doses of the pro-oxidant molecule tert-Butyl hydroperoxide (tBHP) protects against loading-induced chondrocyte death. We hypothesized that compressive cyclic loading at moderate strains (<20%) causes sub-lethal cartilage oxidation that induces an adaptive increase in the endogenous antioxidant defense network. We tested this hypothesis by subjecting healthy bovine articular cartilage explants to in vitro static or cyclic (1 Hz) compressive loading at 50 kPa (15% strain, "physiologic") versus 300 kPa (40% strain, "hyper-physiologic") for 12 h per day for 2 days. We also treated unloaded explants with 100 μM tBHP for 12 h per day for 2 days to differentiate between biomechanical and chemical pro-oxidant stimulation. All loading conditions induced glutathione oxidation relative to unloaded controls, but only the 50 kPa cyclic loading condition increased total glutathione content (twofold). This increase was associated with a greater expression of glutamate-cysteine ligase, the rate-limiting step in glutathione synthesis, compared to 300 kPa cyclic loading. 50 kPa cyclic loading also increased the expression of superoxide dismutase-1 and peroxiredoxin-3. Like 50 kPa loading, tBHP treatment also increased total glutathione content. However, tBHP treatment and 50 kPa cyclic loading differed in their effect on the expression of genes regulating antioxidant defense and cartilage matrix synthesis and degradation. These findings suggest that glutathione metabolism is a mechanosensitive antioxidant defense pathway in chondrocytes and that intermittent pro-oxidant treatment alone is insufficient to account for all changes in mediators of cartilage homeostasis associated with cyclic loading. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:760-769, 2018.
Collapse
Affiliation(s)
- Rita Issa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael Boeving
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA,Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Timothy M. Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA,Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA,Department of Biochemistry and Molecular Biology and Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
35
|
Coleman MC, Brouillette MJ, Andresen NS, Oberley-Deegan RE, Martin JM. Differential Effects of Superoxide Dismutase Mimetics after Mechanical Overload of Articular Cartilage. Antioxidants (Basel) 2017; 6:E98. [PMID: 29189731 PMCID: PMC5745508 DOI: 10.3390/antiox6040098] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/20/2017] [Accepted: 11/28/2017] [Indexed: 01/17/2023] Open
Abstract
Post-traumatic osteoarthritis can develop as a result of the initial mechanical impact causing the injury and also as a result of chronic changes in mechanical loading of the joint. Aberrant mechanical loading initiates excessive production of reactive oxygen species, oxidative damage, and stress that appears to damage mitochondria in the surviving chondrocytes. To probe the benefits of increasing superoxide removal with small molecular weight superoxide dismutase mimetics under severe loads, we applied both impact and overload injury scenarios to bovine osteochondral explants using characterized mechanical platforms with and without GC4403, MnTE-2-PyP, and MnTnBuOE-2-PyP. In impact scenarios, each of these mimetics provides some dose-dependent protection from cell death and loss of mitochondrial content while in repeated overloading scenarios only MnTnBuOE-2-PyP provided a clear benefit to chondrocytes. These results support the hypothesis that superoxide is generated in excess after impact injuries and suggest that superoxide production within the lipid compartment may be a critical mediator of responses to chronic overload. This is an important nuance distinguishing roles of superoxide, and thus superoxide dismutases, in mediating damage to cellular machinery in hyper-acute impact scenarios compared to chronic scenarios.
Collapse
Affiliation(s)
- Mitchell C Coleman
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA.
| | - Marc J Brouillette
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA.
| | - Nicholas S Andresen
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA.
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - James M Martin
- Department of Orthopedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
36
|
Zhang B, Hou R, Zou Z, Luo T, Zhang Y, Wang L, Wang B. Mechanically induced autophagy is associated with ATP metabolism and cellular viability in osteocytes in vitro. Redox Biol 2017; 14:492-498. [PMID: 29096322 PMCID: PMC5680519 DOI: 10.1016/j.redox.2017.10.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/17/2017] [Accepted: 10/25/2017] [Indexed: 12/16/2022] Open
Abstract
Both mechanical loading and intracellular autophagy play important roles in bone homeostasis; however, their relationship remains largely unexplored. The objectives of this study were to determine whether osteocytes undergo autophagy upon fluid shear stress (FSS) loading and to determine the correlation between mechanically induced autophagy and ATP metabolism. Autophagic vacuoles were observed by transmission electron microscopy (TEM) in osteocyte-like MLO-Y4 cells subjected to FSS. Increased autophagic flux was further confirmed by the increased amount of the LC3-II isoform and the degradation of p62. Fluorescent puncta distributed in the cytoplasm were observed in the GFP-LC3 transformed cells subjected to FSS. Furthermore, FSS-induced ATP release and synthesis in osteocytes were attenuated by inhibiting autophagy with 3-MA. After FSS exposure, a high ratio of cell death was observed in cultures pretreated with 3-MA, an autophagy inhibitor, with no significantly different Caspase 3/7 activity. Our results indicated that FSS induces protective autophagy in osteocytes and that mechanically induced autophagy is associated with ATP metabolism and osteocyte survival. From the clinical perspective, it may be possible to enhance skeletal cell survival with drugs that modulate the autophagic state, and the autophagy-related pathway could be a potential target for the prevention of ageing-related bone disorders. Fluid flow shear stress (FSS) induces activation of autophagic flux in MLO-Y4 osteocytes. FSS-induced autophagy promoted ATP metabolism in MLO-Y4 osteocytes. Inhibited autophagy decreased FSS-induced ATP release. FSS-induced autophagy was beneficial to the osteocyte survival after FSS exposure.
Collapse
Affiliation(s)
- Bingbing Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Rutao Hou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Zhen Zou
- Department of Medical Laboratory Technology, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Tiantian Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yang Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, USA
| | - Bin Wang
- Department of Medical Laboratory Technology, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
37
|
Li P, Hou G, Zhang R, Gan Y, Xu Y, Song L, Zhou Q. High-magnitude compression accelerates the premature senescence of nucleus pulposus cells via the p38 MAPK-ROS pathway. Arthritis Res Ther 2017; 19:209. [PMID: 28923094 PMCID: PMC5604423 DOI: 10.1186/s13075-017-1384-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 07/07/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Mechanical overloading can lead to disc degeneration. Nucleus pulposus (NP) cell senescence is aggravated within the degenerated disc. This study was designed to investigate the effects of high compression on NP cell senescence and the underlying molecular mechanism of this process. METHODS Rat NP cells seeded in decalcified bone matrix were subjected to non-compression (control) or compression (2% or 20% deformation, 1.0 Hz, 6 hours/day). The reactive oxygen species (ROS) scavenger N-acetylcysteine (NAC) and the p38 MAPK inhibitor SB203580 were used to investigate the roles of the ROS and p38 MAPK pathway under high-magnitude compression. Additionally, we studied the effects of compression (0.1 or 1.3 MPa, 1.0 Hz, 6 hours/day) in a rat disc organ culture. RESULTS Both in scaffold and organ cultures, high-magnitude compression (20% deformation or 1.3 MPa) increased senescence-associated β-galactosidase (SA-β-Gal) activity, senescence marker (p16 and p53) expression, G1 cell cycle arrest, and ROS generation, and decreased cell proliferation, telomerase activity and matrix (aggrecan and collagen II) synthesis. Further analysis of the 20% deformation group showed that NAC inhibited NP cell senescence but had no obvious effect on phospho-p38 MAPK expression and that SB203580 significantly attenuated ROS generation and NP cell senescence. CONCLUSIONS High-magnitude compression can accelerate NP cell senescence through the p38 MAPK-ROS pathway.
Collapse
Affiliation(s)
- Pei Li
- Department of Orthopaedic Surgery, No. 89 hospital of PLA, Weifang, Shandong, 261026, China.,Department of Orthopaedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Gang Hou
- Department of Orthopaedics, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510700, China
| | - Ruijie Zhang
- Department of Respiratory Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China.
| | - Yibo Gan
- Department of Orthopaedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yuan Xu
- Department of Orthopaedic Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Lei Song
- Department of Orthopaedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Qiang Zhou
- Department of Orthopaedic Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
38
|
Martin JA, Anderson DD, Goetz JE, Fredericks D, Pedersen DR, Ayati BP, Marsh JL, Buckwalter JA. Complementary models reveal cellular responses to contact stresses that contribute to post-traumatic osteoarthritis. J Orthop Res 2017; 35:515-523. [PMID: 27509320 PMCID: PMC5303196 DOI: 10.1002/jor.23389] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 08/05/2016] [Indexed: 02/04/2023]
Abstract
Two categories of joint overloading cause post-traumatic osteoarthritis (PTOA): single acute traumatic loads/impactions and repetitive overloading due to incongruity/instability. We developed and refined three classes of complementary models to define relationships between joint overloading and progressive cartilage loss across the spectrum of acute injuries and chronic joint abnormalities: explant and whole joint models that allow probing of cellular responses to mechanical injury and contact stresses, animal models that enable study of PTOA pathways in living joints and pre-clinical testing of treatments, and patient-specific computational models that define the overloading that causes OA in humans. We coordinated methodologies across models so that results from each informed the others, maximizing the benefit of this complementary approach. We are incorporating results from these investigations into biomathematical models to provide predictions of PTOA risk and guide treatment. Each approach has limitations, but each provides opportunities to elucidate PTOA pathogenesis. Taken together, they help define levels of joint overloading that cause cartilage destruction, show that both forms of overloading can act through the same biologic pathways, and create a framework for initiating clinical interventions that decrease PTOA risk. Considered collectively, studies extending from explants to humans show that thresholds of joint overloading that cause cartilage loss can be defined, that to at least some extent both forms of joint overloading act through the same biologic pathways, and interventions that interrupt these pathways prevent cartilage damage. These observations suggest that treatments that decrease the risk of all forms of OA progression can be discovered. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:515-523, 2017.
Collapse
Affiliation(s)
- James A. Martin
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City Iowa,Departments of Biomedical Engineering, University of Iowa, Iowa City Iowa
| | - Donald D. Anderson
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City Iowa,Departments of Biomedical Engineering, University of Iowa, Iowa City Iowa
| | - Jessica E. Goetz
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City Iowa,Departments of Biomedical Engineering, University of Iowa, Iowa City Iowa
| | - Douglas Fredericks
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City Iowa
| | - Douglas R. Pedersen
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City Iowa,Departments of Biomedical Engineering, University of Iowa, Iowa City Iowa
| | - Bruce P. Ayati
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City Iowa,Departments of Mathematics, University of Iowa, Iowa City Iowa
| | - J. Lawrence Marsh
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City Iowa
| | - Joseph A. Buckwalter
- Departments of Orthopedics and Rehabilitation, University of Iowa, Iowa City Iowa,Iowa City Veterans Administration Medical Center
| |
Collapse
|
39
|
Ayati BP, Kapitanov GI, Coleman MC, Anderson DD, Martin JA. Mathematics as a conduit for translational research in post-traumatic osteoarthritis. J Orthop Res 2017; 35:566-572. [PMID: 27653021 PMCID: PMC5352510 DOI: 10.1002/jor.23439] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 09/15/2016] [Indexed: 02/04/2023]
Abstract
Biomathematical models offer a powerful method of clarifying complex temporal interactions and the relationships among multiple variables in a system. We present a coupled in silico biomathematical model of articular cartilage degeneration in response to impact and/or aberrant loading such as would be associated with injury to an articular joint. The model incorporates fundamental biological and mechanical information obtained from explant and small animal studies to predict post-traumatic osteoarthritis (PTOA) progression, with an eye toward eventual application in human patients. In this sense, we refer to the mathematics as a "conduit of translation." The new in silico framework presented in this paper involves a biomathematical model for the cellular and biochemical response to strains computed using finite element analysis. The model predicts qualitative responses presently, utilizing system parameter values largely taken from the literature. To contribute to accurate predictions, models need to be accurately parameterized with values that are based on solid science. We discuss a parameter identification protocol that will enable us to make increasingly accurate predictions of PTOA progression using additional data from smaller scale explant and small animal assays as they become available. By distilling the data from the explant and animal assays into parameters for biomathematical models, mathematics can translate experimental data to clinically relevant knowledge. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:566-572, 2017.
Collapse
Affiliation(s)
- Bruce P. Ayati
- Departments of Mathematics, Orthopedics & Rehabilitation, and Program in Applied Mathematical and Computational Sciences, University of Iowa
| | | | | | - Donald D. Anderson
- Departments of Orthopedics & Rehabilitation and Biomedical Engineering, University of Iowa
| | - James A. Martin
- Departments of Orthopedics & Rehabilitation and Biomedical Engineering, University of Iowa
| |
Collapse
|
40
|
Low-Intensity Ultrasound Decreases α-Synuclein Aggregation via Attenuation of Mitochondrial Reactive Oxygen Species in MPP(+)-Treated PC12 Cells. Mol Neurobiol 2016; 54:6235-6244. [PMID: 27714630 DOI: 10.1007/s12035-016-0104-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/06/2016] [Indexed: 12/23/2022]
Abstract
Many studies have shown that mitochondrial dysfunction and the subsequent oxidative stress caused by excessive reactive oxygen species (ROS) generation play a central role in the pathogenesis of Parkinson's disease (PD). We have previously shown that low-intensity ultrasound (LIUS) could reduce ROS generation by L-buthionine-(S,R)-sulfoximine (BSO) in retinal pigment epithelial cells. In this study, we studied the effects of LIUS stimulation on the ROS-dependent α-synuclein aggregation in 1-methyl-4-phenylpyridinium ion (MPP+)-treated PC12 cells. We found that LIUS stimulation suppressed the MPP+-induced ROS generation and inhibition of mitochondrial complex I activity in PC12 cells in an intensity-dependent manner at 30, 50, and 100 mW/cm2. Furthermore, LIUS stimulation at 100 mW/cm2 suppressed inhibition of mitochondrial complex activity by MPP+ and actually resulted in a decrease of α-synuclein phosphorylation and aggregation induced by MMP+ treatment in PC12 cells. LIUS stimulation also inhibited expression of casein kinase 2 (CK2) that appears to mediate ROS-dependent α-synuclein aggregation. Finally, LIUS stimulation alleviated the death of PC12 cells by MPP+ treatment in an intensity-dependent manner. We, hence, suggest that LIUS stimulation inhibits ROS generation by MPP+ treatment, thereby suppressing α-synuclein aggregation in PC12 cells.
Collapse
|
41
|
Coleman MC, Ramakrishnan PS, Brouillette MJ, Martin JA. Injurious Loading of Articular Cartilage Compromises Chondrocyte Respiratory Function. Arthritis Rheumatol 2016; 68:662-71. [PMID: 26473613 DOI: 10.1002/art.39460] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 09/29/2015] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To determine whether repeatedly overloading healthy cartilage disrupts mitochondrial function in a manner similar to that associated with osteoarthritis (OA) pathogenesis. METHODS We exposed normal articular cartilage on bovine osteochondral explants to 1 day or 7 consecutive days of cyclic axial compression (0.25 MPa or 1.0 MPa at 0.5 Hz for 3 hours) and evaluated the effects on chondrocyte viability, ATP concentration, reactive oxygen species (ROS) production, indicators of oxidative stress, respiration, and mitochondrial membrane potential. RESULTS Neither 0.25 MPa nor 1.0 MPa of cyclic compression caused extensive chondrocyte death, macroscopic tissue damage, or overt changes in stress-strain behavior. After 1 day of loading, differences in respiratory activities between the 0.25 MPa and 1.0 MPa groups were minimal; however, after 7 days of loading, respiratory activity and ATP levels were suppressed in the 1.0 MPa group relative to the 0.25 MPa group, an effect prevented by pretreatment with 10 mM N-acetylcysteine. These changes were accompanied by increased proton leakage and decreased mitochondrial membrane potential, as well as by increased ROS formation, as indicated by dihydroethidium staining and glutathione oxidation. CONCLUSION Repeated overloading leads to chondrocyte oxidant-dependent mitochondrial dysfunction. This mitochondrial dysfunction may contribute to destabilization of cartilage during various stages of OA in distinct ways by disrupting chondrocyte anabolic responses to mechanical stimuli.
Collapse
|
42
|
Arulkumaran N, Deutschman CS, Pinsky MR, Zuckerbraun B, Schumacker PT, Gomez H, Gomez A, Murray P, Kellum JA. MITOCHONDRIAL FUNCTION IN SEPSIS. Shock 2016; 45:271-81. [PMID: 26871665 PMCID: PMC4755359 DOI: 10.1097/shk.0000000000000463] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mitochondria are an essential part of the cellular infrastructure, being the primary site for high-energy adenosine triphosphate production through oxidative phosphorylation. Clearly, in severe systemic inflammatory states, like sepsis, cellular metabolism is usually altered, and end organ dysfunction is not only common, but also predictive of long-term morbidity and mortality. Clearly, interest is mitochondrial function both as a target for intracellular injury and response to extrinsic stress have been a major focus of basic science and clinical research into the pathophysiology of acute illness. However, mitochondria have multiple metabolic and signaling functions that may be central in both the expression of sepsis and its ultimate outcome. In this review, the authors address five primary questions centered on the role of mitochondria in sepsis. This review should be used both as a summary source in placing mitochondrial physiology within the context of acute illness and as a focal point for addressing new research into diagnostic and treatment opportunities these insights provide.
Collapse
Affiliation(s)
| | - Clifford S. Deutschman
- Department of Pediatrics and Molecular Medicine, Hofstra-North Shore-Long Island Jewish School of Medicine
| | - Michael R. Pinsky
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine
| | | | - Paul T. Schumacker
- Departments of Pediatrics-Neonatology, Cell and Molecular Biology and Medicine, Northwestern University Feinberg School of Medicine
| | - Hernando Gomez
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh PA
| | - Alonso Gomez
- Academia Colombiana de Medicina Critica (ACOMEC)
- Division of Critical Care Medicine, Clínica Palermo, Bogotá, Colombia
| | | | - John A. Kellum
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine
- Center for Critical Care Nephrology, University of Pittsburgh, Pittsburgh PA
| |
Collapse
|
43
|
Lim NH, Vincent TL, Nissim A. In vivo optical imaging of early osteoarthritis using an antibody specific to damaged arthritic cartilage. Arthritis Res Ther 2015; 17:376. [PMID: 26703223 PMCID: PMC4718036 DOI: 10.1186/s13075-015-0898-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/10/2015] [Indexed: 02/07/2023] Open
Abstract
Background The lack of specific and sensitive serum and radiographic biomarkers for early diagnosis of osteoarthritis (OA) as well as for monitoring subtle changes in disease activity in clinical trials has hampered the development of treatments for OA. We previously showed that 1-11E, a human single chain fragment variable (scFv) specific to collagen type II that has been post-translationally modified by reactive oxidants (ROS-CII), binds exclusively to arthritic cartilage. Here we test the validity of 1-11E as a radiographic biomarker for early disease in experimental OA. Methods Murine OA was induced by destabilisation of the medial meniscus (DMM) in adult male mice. Immunohistochemistry of destabilised or sham-operated knees was performed from 2 to 8 weeks post-surgery with Cy5.5-labelled 1-11E and negative control scFv, C7. Prospective in vivo optical images were taken 4 and 8 weeks post-DMM following intra-articular injection of Cy5.5-labelled scFvs, or intravenous injection of Cy5.5-labelled full length monoclonal antibodies (mAbs). Results Specific cartilage staining with 1-11E was apparent as early as 4 weeks post-DMM at the time of earlier cartilage degradation assessed by histology. Prospective in vivo optical images taken 4 and 8 weeks post-DMM following local intra-articular injection of Cy5.5-labelled scFv (n = 7) showed specific in vivo retention of Cys5.5-1-11E scFv following local administration into the knee joint (tissue half-life >78 hours, n = 7, signal to noise ratio (SNR) > 2.1). Specific localization of Cys-5.5-1-11E-mAb to DMM knees (SNR >1.65) was also observed (p < 0.01, n = 8, SNR >1.65). In both cases the SNR increased with time post-DMM. Conclusions 1-11E binds specifically to early osteoarthritic cartilage and can be used as a radiographic biomarker following local or systemic delivery to facilitate early diagnosis and monitor disease progression in OA.
Collapse
Affiliation(s)
- Ngee Han Lim
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
| | - Tonia L Vincent
- Arthritis Research UK Centre for OA Pathogenesis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7FY, UK.
| | - Ahuva Nissim
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK.
| |
Collapse
|
44
|
Le Clanche S, Bonnefont-Rousselot D, Sari-Ali E, Rannou F, Borderie D. Inter-relations between osteoarthritis and metabolic syndrome: A common link? Biochimie 2015; 121:238-52. [PMID: 26700146 DOI: 10.1016/j.biochi.2015.12.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 12/05/2015] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a degenerative disorder of the joint, principally occurring during aging, and characterized by a focal degradation of cartilage. It is the most prevalent rheumatic disease in industrialized countries and represents the second cause of disability in France. However, the etiology of OA remains unclear. There is only one cell type found in cartilage, chondrocyte, which is responsible for its repair and the synthesis of the elements of the extra-cellular matrix. A dysfunction of these cells results in an imbalance between repair and degradation in cartilage, leading to its destruction. Recently, a link between OA and metabolic syndrome (MetS) has been suggested, introducing a notion of metabolic OA, and a new vision of the disease. MetS is characterized by a cluster of factors (insulin resistance, hypertension, dyslipidemia, visceral obesity), although there is still no clear definition of it. During the 20th century, MetS dramatically increased with changes in population lifestyle, becoming a major health issue in industrialized countries. MetS concerns 10-30% of the worldwide population, but is prevalent in 59% of OA patients. Patients with both OA and MetS have more severe symptoms, occurring sooner than in the general population. Indeed, OA is generally a disease concerning the population over 65 years old, but with an associated MetS the target population is around 50 years old. In this review, we will focus on common factors in OA and MetS, such as hypertension, obesity, dyslipidemia, mitochondrial dysfunction and hyperglycemia, linking one disease to the other.
Collapse
Affiliation(s)
- S Le Clanche
- UMR-S 1124 INSERM Toxicologie, Pharmacologie et Signalisation Cellulaire, CUSP, Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France; Unité pédagogique de Biochimie, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France.
| | - D Bonnefont-Rousselot
- Unité pédagogique de Biochimie, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France; UMR-S 1166 INSERM ICAN, Université Pierre et Marie Curie, Paris 6, 75013 Paris, France; Service de Biochimie Métabolique, Groupe hospitalier Pitié-Salpêtrière-Charles Foix, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris Cedex 13, France.
| | - E Sari-Ali
- Groupe de Recherche En Orthopédie de la Pitié-Salpêtrière (GREOPS), Hôpital de la Pitié-Salpêtrière, 47-83 boulevard de l'hôpital, 75013 Paris, France.
| | - F Rannou
- UMR-S 1124 INSERM Toxicologie, Pharmacologie et Signalisation Cellulaire, CUSP, Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France; Service de rééducation, Hôpital Cochin (AP-HP), Université Paris Descartes, 27 rue du faubourg Saint Jacques, 75679 Paris Cedex 14, France.
| | - D Borderie
- UMR-S 1124 INSERM Toxicologie, Pharmacologie et Signalisation Cellulaire, CUSP, Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France; Unité pédagogique de Biochimie, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France; Service de Diagnostic Biologique Automatisé, Hôpital Cochin (AP-HP), 27 rue du faubourg Saint Jacques, 75679 Paris Cedex 14, France.
| |
Collapse
|
45
|
Koike M, Nojiri H, Ozawa Y, Watanabe K, Muramatsu Y, Kaneko H, Morikawa D, Kobayashi K, Saita Y, Sasho T, Shirasawa T, Yokote K, Kaneko K, Shimizu T. Mechanical overloading causes mitochondrial superoxide and SOD2 imbalance in chondrocytes resulting in cartilage degeneration. Sci Rep 2015; 5:11722. [PMID: 26108578 PMCID: PMC4480010 DOI: 10.1038/srep11722] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/03/2015] [Indexed: 12/31/2022] Open
Abstract
Mechanical stress and aging are major risk factors of cartilage degeneration. Human studies have previously reported that oxidative damage increased, while SOD2 protein was reciprocally downregulated in osteoarthritic degenerated cartilage. However, it remains unclear whether mitochondrial superoxide imbalance in chondrocytes causes cartilage degeneration. We herein demonstrate that mechanical loading promoted mitochondrial superoxide generation and selective Sod2 downregulation in chondrocytes in vivo and that mitochondrial superoxide inducer also downregulated Sod2 expression in chondrocytes in vitro. A genetically manipulated model revealed that Sod2 deficiency in chondrocytes also resulted in mitochondrial superoxide overproduction and dysfunction, thus leading to cartilage degeneration. Intra-articular injection of a permeable antioxidant effectively suppressed the mechanical loading-induced mitochondrial superoxide generation and cartilage degeneration in mice. Our findings demonstrate that mitochondrial superoxide plays a pivotal role in the development and progression of osteoarthritis, and the mitochondrial superoxide balance may therefore be a promising target for the treatment of cartilage degeneration.
Collapse
Affiliation(s)
- Masato Koike
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.,Department of Orthopaedics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hidetoshi Nojiri
- Department of Orthopaedics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yusuke Ozawa
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kenji Watanabe
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yuta Muramatsu
- Department of Orthopaedics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Haruka Kaneko
- Department of Orthopaedics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Daichi Morikawa
- Department of Orthopaedics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Keiji Kobayashi
- Department of Orthopaedics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshitomo Saita
- Department of Orthopaedics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takahisa Sasho
- Department of Orthopaedics, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takuji Shirasawa
- Department of Aging Control Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine' Chiba University Graduate School of Medicine, Chiba, Japan
| | - Kazuo Kaneko
- Department of Orthopaedics, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takahiko Shimizu
- Department of Advanced Aging Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
46
|
Emerging targets in osteoarthritis therapy. Curr Opin Pharmacol 2015; 22:51-63. [PMID: 25863583 DOI: 10.1016/j.coph.2015.03.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/17/2015] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is a destructive joint disease in which the initiation may be attributed to direct injury and mechanical disruption of joint tissues, but the progressive changes are dependent on active cell-mediated processes that can be observed or inferred during the generally long time-course of the disease. Based on clinical observations and experimental studies, it is now recognized a that it is possible for individual patients to exhibit common sets of symptoms and structural abnormalities due to distinct pathophysiological pathways that act independently or in combination. Recent research that has focused on the underlying mechanisms involving biochemical cross talk among the cartilage, synovium, bone, and other joint tissues within a background of poorly characterized genetic factors will be addressed in this review.
Collapse
|
47
|
Akasaki Y, Alvarez-Garcia O, Saito M, Caramés B, Iwamoto Y, Lotz MK. FoxO transcription factors support oxidative stress resistance in human chondrocytes. Arthritis Rheumatol 2015; 66:3349-58. [PMID: 25186470 DOI: 10.1002/art.38868] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Accepted: 08/28/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVE A major signaling pathway that regulates cellular aging is the insulin/insulin-like growth factor 1 (IGF-1)/phosphatidylinositol 3-kinase (PI3K)/Akt/FoxO transcription factor axis. We previously observed that FoxO transcription factors are dysregulated in aged and OA cartilage. The objective of this study was to investigate the impact of down-regulated FoxO transcription factors on chondrocytes. METHODS Small interfering RNAs (siRNAs) targeting FOXO1 (siFOXO1) and FOXO3 (siFOXO3) were transfected into human articular chondrocytes. Cell viability following treatment with the oxidant tert-butyl-hydroperoxide (tBHP) was measured by MTT assay. Caspase 3/7 activation and apoptotic cells were examined. Gene and protein expression of antioxidant proteins and autophagy-related proteins and changes in inflammatory mediators following treatment with interleukin-1β were assessed. Cells transfected with FOXO plasmids were also analyzed. RESULTS Cell viability was significantly reduced by siFOXO after treatment with tBHP. Apoptosis accompanied by caspase activation was significantly increased in siFOXO-transfected chondrocytes. Knockdown of FOXO1 and FOXO1+3 resulted in significant reductions in levels of glutathione peroxidase 1 (GPX-1), catalase, light chain 3 (LC3), Beclin1, and sirtuin 1 (SIRT-1) proteins following treatment with tBHP. In contrast, the constitutive active form of FOXO3 increased cell viability while inducing GPX-1, Beclin1, and LC3 in response to tBHP. Expression and production of ADAMTS-4 and chemerin were significantly increased in siFOXO-transfected chondrocytes. CONCLUSION Reduced expression of FoxO transcription factors in chondrocytes increased susceptibility to cell death induced by oxidative stress. This was associated with reduced levels of antioxidant proteins and autophagy-related proteins. Our data provide evidence for a key role of FoxO transcription factors as regulators of chondrocyte oxidative stress resistance and tissue homeostasis.
Collapse
Affiliation(s)
- Yukio Akasaki
- The Scripps Research Institute, La Jolla, California
| | | | | | | | | | | |
Collapse
|
48
|
Zhao X, Petursson F, Viollet B, Lotz M, Terkeltaub R, Liu-Bryan R. Peroxisome proliferator-activated receptor γ coactivator 1α and FoxO3A mediate chondroprotection by AMP-activated protein kinase. Arthritis Rheumatol 2015; 66:3073-82. [PMID: 25047750 DOI: 10.1002/art.38791] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 07/17/2014] [Indexed: 12/19/2022]
Abstract
OBJECTIVE AMP-activated protein kinase (AMPK) inhibits chondrocyte procatabolic responses to inflammation and biomechanical injury. This study was undertaken to test the hypothesis that peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and FoxO3A, 2 major AMPK downstream targets, mediate the chondroprotective effect of AMPK activation. METHODS We assessed the activity of AMPKα (threonine 172 phosphorylation) and the expression of PGC-1α and FoxO3A in human chondrocytes and AMPKα1- or AMPKα2-knockout mouse chondrocytes by Western blotting, and in mouse knee cartilage by immunohistochemistry. We also knocked down or overexpressed PGC-1α and FoxO3A by small interfering RNA or plasmid DNA transfection, respectively. We assessed mitochondrial superoxide generation using MitoSOX Red. RESULTS Expression of PGC-1α and FoxO3A was enhanced by pharmacologic AMPK activator A-769662 but impaired in AMPKα1(-/-) or AMPKα2(-/-) mouse chondrocytes. Reduced expression of PGC-1α and FoxO3A was observed in mouse knee instability-induced osteoarthritis (OA) cartilage and in aged C57BL/6 mouse knee cartilage. Knockdown of PGC-1α and FoxO3A enhanced, but limited the ability of A-769662 to inhibit, phosphorylation of p65 NF-κB (Ser(536) ) and procatabolic responses induced by inflammatory cytokines. Forced expression of PGC-1α and FoxO3A induced increased expression of superoxide dismutase 2 (SOD2) and catalase, but A-769662 failed to increase the expression of SOD2 and catalase in either PGC-1α- or FoxO3A-knockdown chondrocytes. Last, menadione-induced superoxide generation was inhibited by AMPK pharmacologic activators and by overexpression of PGC-1α or FoxO3A. CONCLUSION PGC-1α and FoxO3A limit oxidative stress and at least partially mediate the capacity of AMPK activity to block procatabolic responses in chondrocytes, and therefore have the potential to inhibit the progression of cartilage damage in OA.
Collapse
Affiliation(s)
- Xianling Zhao
- VA San Diego Medical Center and University of California, San Diego
| | | | | | | | | | | |
Collapse
|
49
|
Akasaki Y, Alvarez-Garcia O, Saito M, Caramés B, Iwamoto Y, Lotz MK. FoxO transcription factors support oxidative stress resistance in human chondrocytes. ARTHRITIS & RHEUMATOLOGY (HOBOKEN, N.J.) 2014. [PMID: 25186470 DOI: 10.1002/art.38868.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE A major signaling pathway that regulates cellular aging is the insulin/insulin-like growth factor 1 (IGF-1)/phosphatidylinositol 3-kinase (PI3K)/Akt/FoxO transcription factor axis. We previously observed that FoxO transcription factors are dysregulated in aged and OA cartilage. The objective of this study was to investigate the impact of down-regulated FoxO transcription factors on chondrocytes. METHODS Small interfering RNAs (siRNAs) targeting FOXO1 (siFOXO1) and FOXO3 (siFOXO3) were transfected into human articular chondrocytes. Cell viability following treatment with the oxidant tert-butyl-hydroperoxide (tBHP) was measured by MTT assay. Caspase 3/7 activation and apoptotic cells were examined. Gene and protein expression of antioxidant proteins and autophagy-related proteins and changes in inflammatory mediators following treatment with interleukin-1β were assessed. Cells transfected with FOXO plasmids were also analyzed. RESULTS Cell viability was significantly reduced by siFOXO after treatment with tBHP. Apoptosis accompanied by caspase activation was significantly increased in siFOXO-transfected chondrocytes. Knockdown of FOXO1 and FOXO1+3 resulted in significant reductions in levels of glutathione peroxidase 1 (GPX-1), catalase, light chain 3 (LC3), Beclin1, and sirtuin 1 (SIRT-1) proteins following treatment with tBHP. In contrast, the constitutive active form of FOXO3 increased cell viability while inducing GPX-1, Beclin1, and LC3 in response to tBHP. Expression and production of ADAMTS-4 and chemerin were significantly increased in siFOXO-transfected chondrocytes. CONCLUSION Reduced expression of FoxO transcription factors in chondrocytes increased susceptibility to cell death induced by oxidative stress. This was associated with reduced levels of antioxidant proteins and autophagy-related proteins. Our data provide evidence for a key role of FoxO transcription factors as regulators of chondrocyte oxidative stress resistance and tissue homeostasis.
Collapse
Affiliation(s)
- Yukio Akasaki
- The Scripps Research Institute, La Jolla, California
| | | | | | | | | | | |
Collapse
|
50
|
Reed KN, Wilson G, Pearsall A, Grishko VI. The role of mitochondrial reactive oxygen species in cartilage matrix destruction. Mol Cell Biochem 2014; 397:195-201. [PMID: 25129057 DOI: 10.1007/s11010-014-2187-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/08/2014] [Indexed: 10/24/2022]
Abstract
Upregulation of matrix metalloproteinases (MMPs) is a hallmark of osteoarthritis progression; along with the role reactive oxygen species (ROS) may play in this process. Moreover, mitochondrial DNA damage and dysfunction are also present in osteoarthritic chondrocytes. However, there are no studies published investigating the direct relationship between mitochondrial ROS, mitochondrial DNA damage, and MMP expression. Therefore, the purpose of the present study was to evaluate whether mitochondrial DNA damage and mitochondrial-originated oxidative stress modulates matrix destruction through the upregulation of MMP protein levels. MitoSox red was utilized to observe mitochondrial ROS production while a Quantitative Southern blot technique was conducted to analyze mitochondrial DNA damage. Additionally, Western blot analysis was used to determine MMP protein levels. The results of the present study show that menadione augmented mitochondrial-generated ROS and increased mitochondrial DNA damage. This increase in mitochondrial-generated ROS led to an increase in MMP levels. When a mitochondrial ROS scavenger was added, there was a subsequent reduction in MMP levels. These studies reveal that mitochondrial integrity is essential for maintaining the cartilage matrix by altering MMP levels. This provides new and important insights into the role of mitochondria in chondrocyte function and its potential importance in therapeutic approaches.
Collapse
Affiliation(s)
- Kendra N Reed
- Department of Cell Biology and Neuroscience, University of South Alabama, 307 University Boulevard N., MSB 1201, Mobile, AL, 36688-0002, USA
| | | | | | | |
Collapse
|