1
|
Zila L, Tarantino R, Zastawny P, Waldman SD. Dynamic compression modulates anabolic and catabolic activity in chondrocyte seeded agarose constructs. J Biomech 2025; 183:112598. [PMID: 40023052 DOI: 10.1016/j.jbiomech.2025.112598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/15/2024] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
Mechanical stimulation is a widely used technique in the development of tissue engineered cartilage. While various regimes can enhance tissue growth and improve construct mechanical properties, existing outcome measures predominantly assess the anabolic effect of mechanical stimuli. Catabolic responses are generally overlooked, and a critical gap remains in how mechanical loading simultaneously affects both anabolic and catabolic processes. In this study, full-thickness articular cartilage was aseptically harvested from the metacarpal-phalangeal joints of skeletally mature bovine. Isolated chondrocytes were encapsulated in agarose gels and subjected to dynamic compressive strains from 0 % to 15 % for either 20 or 60 min using a custom-built mechanical stimulation device. Anabolism was assessed by [3H]-proline and [35S]-sulfate incorporation, while catabolism was evaluated by MMP-13 enzymatic activity. Long-term effects of dynamic loading were assessed through biochemical analyses and histological evaluation. Results showed that low-to-moderate strains (2.5 % and 5 %) induced high anabolic activity relative to control with minimal catabolic response. In contrast, high strains (15 %) resulted in elevated catabolic and reduced anabolic activity relative to control. The application of mechanical stimuli over the long-term elicited comparable responses with lower compressive stains leading to improved cartilaginous extracellular matrix accumulation. This study provides valuable insights into the complex interplay between anabolic and catabolic metabolism in chondrocyte-seeded agarose constructs subjected to dynamic compression. This research underscores the necessity of evaluating both responses to optimize the growth and properties of tissue-engineered cartilage.
Collapse
Affiliation(s)
- Lea Zila
- Department of Electrical, Computer, and Biomedical Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Biomedical Engineering, Science and Technology (iBEST), Unity Health and Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Roberto Tarantino
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Biomedical Engineering, Science and Technology (iBEST), Unity Health and Toronto Metropolitan University, Toronto, Ontario, Canada.
| | - Peter Zastawny
- Department of Electrical, Computer, and Biomedical Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Biomedical Engineering, Science and Technology (iBEST), Unity Health and Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Stephen D Waldman
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, Ontario, Canada; Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Biomedical Engineering, Science and Technology (iBEST), Unity Health and Toronto Metropolitan University, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Qin W, Xu S, Wei J, Li F, Zhang C, Zhang H, Liu Y. Deciphering chondrocyte diversity in diabetic osteoarthritis through single-cell transcriptomics. Comput Biol Chem 2025; 115:108356. [PMID: 39848169 DOI: 10.1016/j.compbiolchem.2025.108356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025]
Abstract
The pathophysiological distinctions between osteoarthritis (OA) and diabetic osteoarthritis (DOA) are critical yet not well delineated. In this study, we employed single-cell RNA sequencing to clarify the unique cellular and molecular mechanisms underpinning the progression of both conditions. We identified a novel subpopulation of chondrocytes in DOA, termed 'Heat Shock' chondrocytes, marked by the expression of distinct molecular markers including HSPA1A, HSPA1B, HSPB1, and HSPA8. Our comprehensive gene expression analysis revealed a pronounced upregulation of inflammatory pathways associated with oxidative stress-namely the MAPK, NF-κB, and PI3K signaling pathways-in the effector and proliferating chondrocyte subpopulations, with a predominance in DOA. Further, our investigation into cell-cell communication demonstrated a significant diminution of intercellular signaling in DOA compared to OA. These insights not only elucidate distinct cellular heterogeneities and potential pathogenic mechanisms differentiating OA from DOA but also enhance our understanding of their molecular pathophysiology, offering novel avenues for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Wei Qin
- Medical College, Jiaying University, Meizhou 514031, China; Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510000, China
| | - Shao Xu
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510000, China
| | - Jiatian Wei
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510000, China
| | - Fuxi Li
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Chuanxia Zhang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Huantian Zhang
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou 510000, China.
| | - Yuanxian Liu
- Department of Otolaryngology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen 518033, China.
| |
Collapse
|
3
|
Griffin TM, Komaravolu RK, Lopes EBP, Mehta-D'souza P, Conner T, Kovats T, Kovats S, Allen M, Harris P, Humphrey MB, Welhaven HD, Brahmachary P, June RK. Exercise induces dynamic changes in intra-articular metabolism and inflammation associated with remodeling of the infrapatellar fat pad in mice. Sci Rep 2025; 15:2428. [PMID: 39827311 PMCID: PMC11743197 DOI: 10.1038/s41598-025-86726-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
We hypothesized that daily exercise promotes joint health by upregulating anti-inflammatory mediators via adaptive molecular and metabolic changes in the infrapatellar fat pad (IFP). We tested this hypothesis by conducting time-resolved analyses between 1 and 14 days of voluntary wheel running exercise in C57BL/6J mice. IFP structure and cellularity were evaluated by histomorphology, picrosirius red collagen staining, and flow cytometry analysis of stromal vascular fraction cells. Joint inflammation and metabolism were evaluated by multiplex gene expression analysis of synovium-IFP tissue and synovial fluid metabolomics, respectively. Exercise transiently increased cytokine and chemokine gene expression in synovium-IFP tissue, resolving within the first 5 days of exercise. The acute inflammatory response was associated with decreased adipocyte size and elevated CD45+Gr1+ myeloid cells, increased collagen content, and oxidized phospholipids. Exercise acutely altered synovial fluid metabolites, characterized by increased amino acids, peptides, bile acids, sphingolipids, dicarboxylic acids, and straight medium chain fatty acids and decreased hydroxy fatty acids and diacylglycerols. Between 5 and 14 days of exercise, inflammation, collagen, and adipocyte size returned to pre-exercise levels, and CD206+ immuno-regulatory macrophages increased. Thus, although the onset of new daily exercise transiently induced synovium-IFP inflammation and altered tissue structure, sustained daily exercise promoted IFP homeostasis.
Collapse
Affiliation(s)
- Timothy M Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA.
- Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA.
- Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| | - Ravi K Komaravolu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
| | - Erika Barboza Prado Lopes
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
| | - Padmaja Mehta-D'souza
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
| | - Taylor Conner
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
| | - Tessa Kovats
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, 73104, USA
| | - Susan Kovats
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Madeline Allen
- Department of Health and Exercise Science, University of Oklahoma, Norman, OK, 73019, USA
| | - Peyton Harris
- Department of Biology, University of Oklahoma, Norman, OK, 73019, USA
| | - Mary Beth Humphrey
- Veterans Affairs Medical Center, Oklahoma City, OK, 73104, USA
- Department of Medicine, Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Hope D Welhaven
- Department of Chemistry & Biochemistry, Montana State University, Bozeman, MT, 59717, USA
| | - Priyanka Brahmachary
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, 59717, USA
| | - Ronald K June
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, MT, 59717, USA
| |
Collapse
|
4
|
Cicek H, Tuhanioglu U, Cicek F. Comparison of the bioactivity and apoptotic responses of hyaline cartilage chondrocytes and fibrocartilage chondrocytes obtained by bone marrow stimulation in rats. ACTA ORTHOPAEDICA ET TRAUMATOLOGICA TURCICA 2024; 58:250-254. [PMID: 39560637 PMCID: PMC11583943 DOI: 10.5152/j.aott.2024.22172] [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: 10/31/2022] [Accepted: 09/16/2024] [Indexed: 11/20/2024]
Abstract
Objective The microfracture technique is often considered the standard therapy for treating cartilage defects. The aim of the treatment is the migration of mesenchymal stem cells from the bone marrow into the defect area. However, this regeneration process often results in the formation of fibrocartilage instead of natural hyaline cartilage, due to cellular mechanisms whose causes are not well understood. Therefore, in this study, the differences in bioactivity and apoptosis of fibrocartilage tissue-derived chondrocytes (FCh) obtained by bone marrow stimulation and natural hyaline cartilage tissue-derived chondrocytes (HCh) in the knee joint of rats were compared. Methods A total of 24 Wistar albino rats were used in this study, and one hind leg of each animal was operated on, while the other served as a control. A 2-step surgical procedure was performed: First, a microfracture was generated at a 2 mm × 2 mm cartilage defect area in the medial condyle of the femur after a left knee arthrotomy. Second, 6 weeks later, after a double knee arthrotomy, fibrocartilage from the microfracture area of the left knee and 2 × 2 mm of hyaline cartilage from the medial femoral condyle of the right knee were harvested. Chondrocytes were isolated and grouped as HCh or FCh, and cell viability and apoptosis were determined by MTT (4,5-dimethylthiazol -2-yl)-2,5-diphenyltetrazolium bromide) and caspase-3 assays using enzyme-linked immunosorbent assay (ELISA) kits. Results Fibrocartilage tissue-derived chondrocytes showed similar bioactivity and apoptotic response as HCh under physiological conditions. However, low-dose H2O2 decreased viability (bioactivity) and increased apoptosis in FCh without affecting HCh. High-dose H2O2 reduced the bioactivity of both cell types and induced apoptosis, while the response of FCh to oxidative stress was more aggressive than that of HCh. Conclusion The different response patterns in oxidative stress may provide a basis for understanding the limited survival time of bone marrow-derived fibrocartilage tissue induced by microfractures. Level of Evidence N/A.
Collapse
Affiliation(s)
- Hakan Cicek
- Adana City Hospital, Orthopedics and Traumatology Clinics, Adana, Türkiye
| | - Umit Tuhanioglu
- Adana City Hospital, Orthopedics and Traumatology Clinics, Adana, Türkiye
| | - Figen Cicek
- Department of Biophysics, Cukurova University Faculty of Medicine, Adana, Türkiye
| |
Collapse
|
5
|
Capuana E, Marino D, Di Gesù R, La Carrubba V, Brucato V, Tuan RS, Gottardi R. A High-Throughput Mechanical Activator for Cartilage Engineering Enables Rapid Screening of in vitro Response of Tissue Models to Physiological and Supra-Physiological Loads. Cells Tissues Organs 2023; 211:670-688. [PMID: 34261061 PMCID: PMC9843549 DOI: 10.1159/000514985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/02/2021] [Indexed: 01/25/2023] Open
Abstract
Articular cartilage is crucially influenced by loading during development, health, and disease. However, our knowledge of the mechanical conditions that promote engineered cartilage maturation or tissue repair is still incomplete. Current in vitro models that allow precise control of the local mechanical environment have been dramatically limited by very low throughput, usually just a few specimens per experiment. To overcome this constraint, we have developed a new device for the high throughput compressive loading of tissue constructs: the High Throughput Mechanical Activator for Cartilage Engineering (HiT-MACE), which allows the mechanoactivation of 6 times more samples than current technologies. With HiT-MACE we were able to apply cyclic loads in the physiological (e.g., equivalent to walking and normal daily activity) and supra-physiological range (e.g., injurious impacts or extensive overloading) to up to 24 samples in one single run. In this report, we compared the early response of cartilage to physiological and supra-physiological mechanical loading to the response to IL-1β exposure, a common but rudimentary in vitro model of cartilage osteoarthritis. Physiological loading rapidly upregulated gene expression of anabolic markers along the TGF-β1 pathway. Notably, TGF-β1 or serum was not included in the medium. Supra-physiological loading caused a mild catabolic response while IL-1β exposure drove a rapid anabolic shift. This aligns well with recent findings suggesting that overloading is a more realistic and biomimetic model of cartilage degeneration. Taken together, these findings showed that the application of HiT-MACE allowed the use of larger number of samples to generate higher volume of data to effectively explore cartilage mechanobiology, which will enable the design of more effective repair and rehabilitation strategies for degenerative cartilage pathologies.
Collapse
Affiliation(s)
- Elisa Capuana
- Department of Engineering, University of Palermo, Palermo, Italy,Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Davide Marino
- Department of Engineering, University of Palermo, Palermo, Italy,Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Roberto Di Gesù
- Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Children's Hospital of Philadelphia, and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA,Fondazione Ri.MED, Palermo, Italy
| | - Vincenzo La Carrubba
- Department of Engineering, University of Palermo, Palermo, Italy,INSTM, Palermo Research Unit, Palermo, Italy
| | - Valerio Brucato
- Department of Engineering, University of Palermo, Palermo, Italy
| | - Rocky S. Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Riccardo Gottardi
- Center for Cellular and Molecular Engineering, Department of Orthopedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Children's Hospital of Philadelphia, and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA,Fondazione Ri.MED, Palermo, Italy,*Riccardo Gottardi,
| |
Collapse
|
6
|
Liu L, Luo P, Yang M, Wang J, Hou W, Xu P. The role of oxidative stress in the development of knee osteoarthritis: A comprehensive research review. Front Mol Biosci 2022; 9:1001212. [PMID: 36203877 PMCID: PMC9532006 DOI: 10.3389/fmolb.2022.1001212] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Knee osteoarthritis (KOA) is one of the most common degenerative diseases, and its core feature is the degeneration and damage of articular cartilage. The cartilage degeneration of KOA is due to the destruction of dynamic balance caused by the activation of chondrocytes by various factors, with oxidative stress playing an important role in the pathogenesis of KOA. The overproduction of reactive oxygen species (ROS) is a result of oxidative stress, which is caused by a redox process that goes awry in the inherent antioxidant defence system of the human body. Superoxide dismutase (SOD) inside and outside chondrocytes plays a key role in regulating ROS in cartilage. Additionally, synovitis is a key factor in the development of KOA. In an inflammatory environment, hypoxia in synovial cells leads to mitochondrial damage, which leads to an increase in ROS levels, which further aggravates synovitis. In addition, oxidative stress significantly accelerates the telomere shortening and ageing of chondrocytes, while ageing promotes the development of KOA, damages the regulation of redox of mitochondria in cartilage, and stimulates ROS production to further aggravate KOA. At present, there are many drugs to regulate the level of ROS, but these drugs still need to be developed and verified in animal models of KOA. We discuss mainly how oxidative stress plays a part in the development of KOA. Although the current research has achieved some results, more research is needed.
Collapse
|
7
|
Inter-limb asymmetry of kinetic and electromyographic during walking in patients with chronic ankle instability. Sci Rep 2022; 12:3928. [PMID: 35273300 PMCID: PMC8913811 DOI: 10.1038/s41598-022-07975-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/23/2022] [Indexed: 12/26/2022] Open
Abstract
After an initial ankle sprain, a relevant number of participants develop chronic ankle instability (CAI). Compensatory strategies in patients with CAI may change the inter-limb symmetry needed for absorbing movement-related forces. Accordingly, an increased risk of injury can occur. The present study aimed to compare the inter-limb asymmetry of kinetic and electromyography between individuals with CAI and without a history of an ankle sprain (Non-CAI) during walking. In this cross-sectional study, fifty-six athletes (28 CAI; 28 Non-CAI) participated. Participants walked at a comfortable pace over level ground while vertical ground reaction force (vGRF) and muscle activity of the tibialis anterior, peroneus longus, medial gastrocnemius, and gluteus medius were recorded. Inter-limb asymmetry during walking was calculated for each of the variables. Patients with CAI exhibited a greater inter-limb asymmetry of the first peak of vGRF, time to peak vGRF, and loading rate (P < 0.001), as well as presenting a greater inter-limb asymmetry of peroneus longus activity (contact phase) (P = 0.003) and gluteus medius activity (midstance/propulsion phase) (P = 0.010) compared to the Non-CAI group. No other differences in vGRF or muscles activity were observed between the groups. Our findings indicate that patients with CAI walk with greater inter-limb asymmetry in vGRF and muscle activity in different phases of the gait cycle compared to Non-CAI group. Our results could inform future studies on gait training aimed to reduce asymmetry during walking in patients with CAI.
Collapse
|
8
|
Zhang L, Virgous C, Si H. How Does Ginsenoside Rh2 Mitigate Adipogenesis in Cultured Cells and Obese Mice? Molecules 2020; 25:E2412. [PMID: 32455850 PMCID: PMC7287807 DOI: 10.3390/molecules25102412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/02/2020] [Accepted: 05/19/2020] [Indexed: 02/06/2023] Open
Abstract
Ginsenoside Rh2, an intermediate metabolite of ginseng, but not naturally occurring, has recently drawn attention because of its anticancer effect. However, it is not clear if and how Rh2 inhibits preadipocytes differentiation. In the present study, we hypothesized that ginsenoside Rh2 attenuates adipogenesis through regulating the peroxisome proliferator-activated receptor gamma (PPAR-γ) pathway both in cells and obese mice. Different concentrations of Rh2 were applied both in 3T3-L1 cells and human primary preadipocytes to determine if Rh2 inhibits cell differentiation. Dietary Rh2 was administered to obese mice to determine if Rh2 prevents obesity in vivo. The mRNA and protein expression of PPAR-γ pathway molecules in cells and tissues were measured by real-time polymerase chain reaction (RT-PCR) and Western blot, respectively. Our results show that Rh2 dose-dependently (30-60 μM) inhibited cell differentiation in 3T3-L1 cells (44.5% ± 7.8% of control at 60 μM). This inhibitory effect is accompanied by the attenuation of the protein and/or mRNA expression of adipogenic markers including PPAR-γ and CCAAT/enhancer binding protein alpha, fatty acid synthase, fatty acid binding protein 4, and perilipin significantly (p < 0.05). Moreover, Rh2 significantly (p < 0.05) inhibited differentiation in human primary preadipocytes at much lower concentrations (5-15 μM). Furthermore, dietary intake of Rh2 (0.1 g Rh2/kg diet, w/w for eight weeks) significantly (p < 0.05) reduced protein PPAR-γ expression in liver and hepatic glutathione reductase and lowered fasting blood glucose. These results suggest that ginsenoside Rh2 dose-dependently inhibits adipogenesis through down-regulating the PPAR-γ pathway, and Rh2 may be a potential agent in preventing obesity in vivo.
Collapse
Affiliation(s)
- Longyun Zhang
- Department of Human Sciences, Tennessee State University, Nashville, TN 37209, USA;
| | - Carlos Virgous
- Animal Care Facility, Meharry Medical College, Nashville, TN 37208, USA;
| | - Hongwei Si
- Department of Human Sciences, Tennessee State University, Nashville, TN 37209, USA;
| |
Collapse
|
9
|
Zhu S, Makosa D, Miller BF, Griffin TM. Glutathione as a mediator of cartilage oxidative stress resistance and resilience during aging and osteoarthritis. Connect Tissue Res 2020; 61:34-47. [PMID: 31522568 PMCID: PMC6884680 DOI: 10.1080/03008207.2019.1665035] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose: An underlying cause of osteoarthritis (OA) is the inability of chondrocytes to maintain homeostasis in response to changing stress conditions. The purpose of this article was to review and experimentally evaluate oxidative stress resistance and resilience concepts in cartilage using glutathione redox homeostasis as an example. This framework may help identify novel approaches for promoting chondrocyte homeostasis during aging and obesity.Materials and Methods: Changes in glutathione content and redox ratio were evaluated in three models of chondrocyte stress: (1) age- and tissue-specific changes in joint tissues of 10 and 30-month old F344BN rats, including ex vivo patella culture experiments to evaluate N-acetylcysteine dependent resistance to interleukin-1beta; (2) effect of different durations and patterns of cyclic compressive loading in bovine cartilage on glutathione stress resistance and resilience pathways; (3) time-dependent changes in GSH:GSSG in primary chondrocytes from wild-type and Sirt3 deficient mice challenged with the pro-oxidant menadione.Results: Glutathione was more abundant in cartilage than meniscus or infrapatellar fat pad, although cartilage was also more susceptible to age-related glutathione oxidation. Glutathione redox homeostasis was sensitive to the duration of compressive loading such that load-induced oxidation required unloaded periods to recover and increase total antioxidant capacity. Exposure to a pro-oxidant stress enhanced stress resistance by increasing glutathione content and GSH:GSSG ratio, especially in Sirt3 deficient cells. However, the rate of recovery, a marker of resilience, was delayed without Sirt3.Conclusions: OA-related models of cartilage stress reveal multiple mechanisms by which glutathione provides oxidative stress resistance and resilience.
Collapse
Affiliation(s)
- Shouan Zhu
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Dawid Makosa
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA,Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Benjamin F. Miller
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Timothy M. Griffin
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA,Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| |
Collapse
|
10
|
Glutamine Metabolism Is Essential for Stemness of Bone Marrow Mesenchymal Stem Cells and Bone Homeostasis. Stem Cells Int 2019; 2019:8928934. [PMID: 31611919 PMCID: PMC6757285 DOI: 10.1155/2019/8928934] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023] Open
Abstract
Skeleton has emerged as an endocrine organ which is both capable of regulating energy metabolism and being a target for it. Glutamine is the most bountiful and flexible amino acid in the body which provides adenosine 5′-triphosphate (ATP) demands for cells. Emerging evidences support that glutamine which acts as the second metabolic regulator after glucose exerts crucial roles in bone homeostasis at cellular level, including the lineage allocation and proliferation of bone mesenchymal stem cells (BMSCs), the matrix mineralization of osteoblasts, and the biosynthesis in chondrocytes. The integrated mechanism consisting of WNT, mammalian target of rapamycin (mTOR), and reactive oxygen species (ROS) signaling pathway in a glutamine-dependent pattern is responsible to regulate the complex intrinsic biological process, despite more extensive molecules are deserved to be elucidated in glutamine metabolism further. Indeed, dysfunctional glutamine metabolism enhances the development of degenerative bone diseases, such as osteoporosis and osteoarthritis, and glutamine or glutamine progenitor supplementation can partially restore bone defects which may promote treatment of bone diseases, although the mechanisms are not quite clear. In this review, we will summarize and update the latest research findings and clinical trials on the crucial regulatory roles of glutamine metabolism in BMSCs and BMSC-derived bone cells, also followed with the osteoclasts which are important in bone resorption.
Collapse
|
11
|
Bolduc JA, Collins JA, Loeser RF. Reactive oxygen species, aging and articular cartilage homeostasis. Free Radic Biol Med 2019; 132:73-82. [PMID: 30176344 PMCID: PMC6342625 DOI: 10.1016/j.freeradbiomed.2018.08.038] [Citation(s) in RCA: 416] [Impact Index Per Article: 69.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/07/2018] [Accepted: 08/30/2018] [Indexed: 01/08/2023]
Abstract
Chondrocytes are responsible for the maintenance of the articular cartilage. A loss of homeostasis in cartilage contributes to the development of osteoarthritis (OA) when the synthetic capacity of chondrocytes is overwhelmed by processes that promote matrix degradation. There is evidence for an age-related imbalance in reactive oxygen species (ROS) production relative to the anti-oxidant capacity of chondrocytes that plays a role in cartilage degradation as well as chondrocyte cell death. The ROS produced by chondrocytes that have received the most attention include superoxide, hydrogen peroxide, the reactive nitrogen species nitric oxide, and the nitric oxide derived product peroxynitrite. Excess levels of these ROS not only cause oxidative-damage but, perhaps more importantly, cause a disruption in cell signaling pathways that are redox-regulated, including Akt and MAP kinase signaling. Age-related mitochondrial dysfunction and reduced activity of the mitochondrial superoxide dismutase (SOD2) are associated with an increase in mitochondrial-derived ROS and are in part responsible for the increase in chondrocyte ROS with age. Peroxiredoxins (Prxs) are a key family of peroxidases responsible for removal of H2O2, as well as for regulating redox-signaling events. Prxs are inactivated by hyperoxidation. An age-related increase in chondrocyte Prx hyperoxidation and an increase in OA cartilage has been noted. The finding in mice that deletion of SOD2 or the anti-oxidant gene transcriptional regulator nuclear factor-erythroid 2- related factor (Nrf2) result in more severe OA, while overexpression or treatment with mitochondrial targeted anti-oxidants reduces OA, further support a role for excessive ROS in the pathogenesis of OA. Therefore, new therapeutic strategies targeting specific anti-oxidant systems including mitochondrial ROS may be of value in reducing the progression of age-related OA.
Collapse
Affiliation(s)
- Jesalyn A Bolduc
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina, Chapel Hill, NC, USA; Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - John A Collins
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina, Chapel Hill, NC, USA; Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Richard F Loeser
- Division of Rheumatology, Allergy, and Immunology, University of North Carolina, Chapel Hill, NC, USA; Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Xue J, Min Z, Xia Z, Cheng B, Lan B, Zhang F, Han Y, Wang K, Sun J. The hsa-miR-181a-5p reduces oxidation resistance by controlling SECISBP2 in osteoarthritis. BMC Musculoskelet Disord 2018; 19:355. [PMID: 30286747 PMCID: PMC6172777 DOI: 10.1186/s12891-018-2273-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 09/23/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The phenotypes of osteoarthritis (OA) consist of cartilage extracellular matrix (ECM) metabolism disorder and the breakdown of cartilage homeostasis, which are induced by pro-inflammatory factors and oxidative stress. Selenoproteins regulated by selenocysteine insertion sequence binding protein 2 (SBP2) are highly effective antioxidants, but their regulatory mechanisms, particularly the involvement of miRNAs, are not fully understood. METHODS To explore whether miR-181a-5p and SBP2 are involved in OA pathogenesis, we established an IL-1β model using the chondrocyte SW1353 cell line. Next, we up- or down-regulated SBP2 and miRNA-181a-5p expression in the cells. Finally, we measured the expression of miRNA-181a-5p, SBP2 and three selenoproteins in OA cartilage and peripheral blood. RESULTS The results showed that IL-1β increased hsa-miR-181a-5p and decreased SBP2 in a time- and dose-dependent manner. GPX1 and GPX4, which encode crucial glutathione peroxidase antioxidant enzymes, were up-regulated along with SBP2 and miR-181a-5p. Furthermore, SBP2 showed a significant negative correlation with miR-181a-5p during induced ATDC5 cell differentiation. There was lower GPX1 and GPX4 mRNA expression and SBP2 protein expression in damaged cartilage than in smooth cartilage from the same OA sample, and hsa-miR-181a-5p expression on the contrary. Similar results were observed in peripheral blood. In conclusion, we have reported a novel pathway in which pro-inflammatory factors, miRNA, SBP2 and selenoproteins are associated with oxidation resistance in cartilage. CONCLUSION Overall, this study provides the first comprehensive evidence that pro-inflammatory factors cause changes in the cartilage antioxidant network and describes the discovery of novel mediators of cartilage oxidative stress and OA pathophysiology. Our data suggest that miR-181a-5p may be used to develop novel early-stage diagnostic and therapeutic strategies for OA.
Collapse
Affiliation(s)
- Jianli Xue
- Department of Orthopaedics, The Second Affiliated Hospital, Xi'an Jiaotong University Health Science Center, 157 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Zixin Min
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Zhuqing Xia
- Beaurau of healthcare, Shaanxi Health and Family Planning Commission, Xi'an, Shaanxi, 710000, People's Republic of China
| | - Bin Cheng
- Department of Orthopaedics, The Second Affiliated Hospital, Xi'an Jiaotong University Health Science Center, 157 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Binshang Lan
- Department of Orthopaedics, The Second Affiliated Hospital, Xi'an Jiaotong University Health Science Center, 157 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Fujun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yan Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Kunzheng Wang
- Department of Orthopaedics, The Second Affiliated Hospital, Xi'an Jiaotong University Health Science Center, 157 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China.
| | - Jian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, People's Republic of China
| |
Collapse
|
13
|
Guo XE, Hung CT, Sandell LJ, Silva MJ. Musculoskeletal mechanobiology: A new era for MechanoMedicine. J Orthop Res 2018; 36:531-532. [PMID: 29409134 DOI: 10.1002/jor.23789] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- X Edward Guo
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY 10027
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY 10027
| | - Linda J Sandell
- Department of Orthopaedic Surgery, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110
| |
Collapse
|