401
|
Maleckar MM, Martín-Vasallo P, Giles WR, Mobasheri A. Physiological Effects of the Electrogenic Current Generated by the Na +/K + Pump in Mammalian Articular Chondrocytes. Bioelectricity 2020; 2:258-268. [PMID: 34471850 PMCID: PMC8370340 DOI: 10.1089/bioe.2020.0036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: Although the chondrocyte is a nonexcitable cell, there is strong interest in gaining detailed knowledge of its ion pumps, channels, exchangers, and transporters. In combination, these transport mechanisms set the resting potential, regulate cell volume, and strongly modulate responses of the chondrocyte to endocrine agents and physicochemical alterations in the surrounding extracellular microenvironment. Materials and Methods: Mathematical modeling was used to assess the functional roles of energy-requiring active transport, the Na+/K+ pump, in chondrocytes. Results: Our findings illustrate plausible physiological roles for the Na+/K+ pump in regulating the resting membrane potential and suggest ways in which specific molecular components of pump can respond to the unique electrochemical environment of the chondrocyte. Conclusion: This analysis provides a basis for linking chondrocyte electrophysiology to metabolism and yields insights into novel ways of manipulating or regulating responsiveness to external stimuli both under baseline conditions and in chronic diseases such as osteoarthritis.
Collapse
Affiliation(s)
| | - Pablo Martín-Vasallo
- UD of Biochemistry and Molecular Biology, Universidad de La Laguna, San Cristóbal de La Laguna, Spain.,Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Wayne R Giles
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Canada
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.,Department of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
402
|
Liu Q, Hebert JR, Shivappa N, Guo J, Tao K, Zeng C, Lei G, Lin J, Zhang Y. Inflammatory potential of diet and risk of incident knee osteoarthritis: a prospective cohort study. Arthritis Res Ther 2020; 22:209. [PMID: 32912291 PMCID: PMC7488131 DOI: 10.1186/s13075-020-02302-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/24/2020] [Indexed: 12/22/2022] Open
Abstract
Background To examine the relation between inflammatory potential of diet and incident knee osteoarthritis (OA) and the role of BMI in the association of interest. Methods In the Osteoarthritis Initiative, the energy-adjusted dietary inflammatory index (E-DII™) scores were calculated based on the Block Brief 2000 Food Frequency Questionnaire and categorized into sex-specific quartiles. Outcomes were incident (1) radiographic knee OA (ROA) (i.e., a KL grade ≥ 2) and (2) symptomatic knee OA (SxOA) (i.e., a combination of frequent knee pain and ROA). We fitted generalized estimating equation models to examine the association between E-DII scores and incident knee OA. We performed mediation analyses to assess the potential mediation by BMI in the DII-OA relation. Results Over a 48-month follow-up period, 232 and 978 knees developed ROA and SxOA, respectively. Compared with the lowest (most anti-inflammatory) E-DII quartile, the odds ratio (OR) of incident ROA for the highest (most pro-inflammatory) E-DII quartile was 1.73 (95% confidence interval (CI) 1.15 to 2.62, Ptrend = 0.007). The corresponding OR for SxOA was 1.43 (95% CI 1.16 to 1.76, Ptrend = 0.001). The DII-OA association was significantly mediated via BMI with an indirect effect of 1.08 (95% CI 1.04, 1.13) for ROA and 1.13 (95% CI 1.09, 1.16) for SxOA, accounting for 20.4% and 44.5% of the total effect, respectively. Conclusions A higher inflammatory potential of diet increased the risk of knee OA. The association was significantly mediated via BMI. Targeting the inflammatory potential of diet may be beneficial to reduce the risk of knee OA.
Collapse
Affiliation(s)
- Qiang Liu
- Arthritis Clinic and Research Center, Peking University People's Hospital, No.11 Xizhimen South Road, Xicheng District, Beijing, 100044, China.,Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St., Boston, MA, 02114, USA
| | - James R Hebert
- Cancer Prevention and Control Program, University of South Carolina, Columbia, SC, USA.,Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Nitin Shivappa
- Cancer Prevention and Control Program, University of South Carolina, Columbia, SC, USA.,Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Jianjun Guo
- Capital University of Physical Education and Sports, Beijing, China
| | - Ke Tao
- Arthritis Clinic and Research Center, Peking University People's Hospital, No.11 Xizhimen South Road, Xicheng District, Beijing, 100044, China
| | - Chao Zeng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Guanghua Lei
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Jianhao Lin
- Arthritis Clinic and Research Center, Peking University People's Hospital, No.11 Xizhimen South Road, Xicheng District, Beijing, 100044, China.
| | - Yuqing Zhang
- Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St., Boston, MA, 02114, USA.
| |
Collapse
|
403
|
Robertson-Plouch C, Stille JR, Liu P, Smith C, Brown D, Warner M, Hu L, Fisher MJ. A randomized clinical efficacy study targeting mPGES1 or EP4 in dogs with spontaneous osteoarthritis. Sci Transl Med 2020; 11:11/516/eaaw9993. [PMID: 31666405 DOI: 10.1126/scitranslmed.aaw9993] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022]
Abstract
Canine studies of spontaneous osteoarthritis (OA) pain add valuable data supporting drug treatment mechanisms that may translate to humans. A multicenter, randomized, double-blind, placebo- and active-controlled study was conducted in client-owned dogs with moderate OA pain to evaluate efficacy of LYA, an inhibitor of microsomal prostaglandin E synthase-1 (mPGES1), an EP4 antagonist (LYB), and carprofen, versus placebo. Of 255 dogs screened, 163 were randomized (placebo/LYA/LYB/carprofen: n = 43/39/42/39) and 158 completed treatment. Efficacy versus placebo was assessed using Bayesian mixed-effect model for repeated measure analyses of the Canine Brief Pain Inventory (CBPI) pain interference score (PIS; primary endpoint), pain severity score, and overall impression, as well as the Liverpool Osteoarthritis in Dogs (LOAD) mobility score. The posterior probability that the difference to placebo was <0 at week 2 was 80% for LYA and 54% for LYB for CBPI PIS (both <95% predefined threshold). For secondary endpoints, the posterior probability that the difference to placebo was <0 at week 2 ranged from 89 to 96% for LYA and from 56 to 89% for LYB. The posterior probabilities comparing carprofen to placebo groups were ≥90% for all efficacy endpoints. The proportion of dogs with one or more adverse event was not significantly different from placebo (32.6%) for LYA (35.9%) or carprofen (25.6%), but the rate for LYB (59.5%) was higher versus placebo (P = 0.017). LYA treatment demonstrated consistent improvement in all efficacy measures, suggesting that inhibition of mPGES1 may be an effective treatment for chronic pain associated with OA.
Collapse
Affiliation(s)
| | - John R Stille
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA.
| | - Peng Liu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Claire Smith
- Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| | - Dorothy Brown
- Elanco, Eli Lilly and Company, Indianapolis, IN 46140, USA.,School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Margaret Warner
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Leijun Hu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Matthew J Fisher
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN 46285, USA
| |
Collapse
|
404
|
Hu B, Gao F, Li C, Zhang B, An M, Lu M, Liu Y, Liu Y. Rhein laden pH-responsive polymeric nanoparticles for treatment of osteoarthritis. AMB Express 2020; 10:158. [PMID: 32865763 PMCID: PMC7459081 DOI: 10.1186/s13568-020-01095-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/19/2020] [Indexed: 12/16/2022] Open
Abstract
Osteoarthritis (OA) is a condition associated with severe inflammation, cartilage destruction and degeneration of joints. Rhein (Rh) is an effective anti-inflammatory drug with proven efficacy in in-vitro and in-vivo models. pH sensitive Rh and NH4HCO3 laden poly (lactic-co-glycolic acid (PLGA) nanoparticles (NPs) (Rh-PLGA-NPs@NH4) are developed for an effective treatment of OA. The Rh-PLGA-NPs@NH4 are prepared along with Rh-PLGA-NPs as a control by double emulsion method. Rh-PLGA-NPs@NH4 was characterized for their size, shape, morphology and encapsulation efficiency (EE). The effect of pH on release of Rh from Rh-PLGA-NPs@NH4 was studied at different pH. Further, the cytotoxicity effect of Rh-PLGA-NPs@NH4 on THP-1 cells were evaluated. Anti-inflammatory efficacy was evaluated on LPS stimulated THP-1 cells and the release of pro-inflammatory cytokines was evaluated and compared with control. The size of Rh-PLGA-NPs@NH4 and Rh-PLGA-NPs was found to be 190.7 ± 1.2 nm and 134.6 ± 2.4 nm respectively with poly dispersity (PDI) 0.14 and 0.15. The zeta potential of Rh-PLGA-NPs@NH4 was found to be -22 ± 1.12 mV. Rh-PLGA-NPs@NH4 were uniform, smooth and spherical shape as confirmed using electron microscopy analysis. Rh-PLGA-NPs@NH4 release the Rh more effectively in the low pH of synovial fluid environment (SFE). Rh-PLGA-NPs@NH4 also significantly affect inflammatory cytokines TNF-α and IL-1β and reduced their release in LPS stimulated THP-1 cells. Reactive oxygen species (ROS), a mediator responsible for the cartilage collapse was also found to be reduced. Results proposes that Rh-PLGA-NPs could provide therapeutic solution to those patients who suffer from chronic joint ailments by reducing the progression of OA.
Collapse
|
405
|
Pain-related behavior is associated with increased joint innervation, ipsilateral dorsal horn gliosis, and dorsal root ganglia activating transcription factor 3 expression in a rat ankle joint model of osteoarthritis. Pain Rep 2020; 5:e846. [PMID: 33490841 PMCID: PMC7808682 DOI: 10.1097/pr9.0000000000000846] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/18/2020] [Accepted: 07/17/2020] [Indexed: 12/02/2022] Open
Abstract
Supplemental Digital Content is Available in the Text. In a rat model of osteoarthritis, we found increased joint sensory and sympathetic innervation and glia changes in dorsal horn, accompanying pain-related behavior onset. Introduction: Osteoarthritis (OA)-associated pain is often poorly managed, as our understanding of the underlying pain mechanisms remains limited. The known variability from patient to patient in pain control could be a consequence of a neuropathic component in OA. Methods: We used a rat monoiodoacetate model of the ankle joint to study the time-course of the development of pain-related behavior and pathological changes in the joint, dorsal root ganglia (DRG), and spinal cord, and to investigate drug treatments effects. Results: Mechanical hypersensitivity and loss of mobility (as assessed by treadmill) were detected from 4 weeks after monoiodoacetate. Cold allodynia was detected from 5 weeks. Using histology and x-ray microtomography, we confirmed significant cartilage and bone degeneration at 5 and 10 weeks. We detected increased nociceptive peptidergic and sympathetic fiber innervation in the subchondral bone and synovium at 5 and 10 weeks. Sympathetic blockade at 5 weeks reduced pain-related behavior. At 5 weeks, we observed, ipsilaterally only, DRG neurons expressing anti-activating transcription factor 3, a neuronal stress marker. In the spinal cord, there was microgliosis at 5 and 10 weeks, and astrocytosis at 10 weeks only. Inhibition of glia at 5 weeks with minocycline and fluorocitrate alleviated mechanical allodynia. Conclusion: Besides a detailed time-course of pathology in this OA model, we show evidence of contributions of the sympathetic nervous system and dorsal horn glia to pain mechanisms. In addition, late activating transcription factor 3 expression in the DRG that coincides with these changes provides evidence in support of a neuropathic component in OA pain.
Collapse
|
406
|
Li C, Zheng Z. Identification of Novel Targets of Knee Osteoarthritis Shared by Cartilage and Synovial Tissue. Int J Mol Sci 2020; 21:6033. [PMID: 32842604 PMCID: PMC7504179 DOI: 10.3390/ijms21176033] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
Arthritis is the leading cause of disability among adults, while osteoarthritis (OA) is the most common form of arthritis that results in cartilage loss. However, accumulating evidence suggests that the protective hyaline cartilage should not be the sole focus of OA treatment. Particularly, synovium also plays essential roles in OA's initiation and progression and warrants serious consideration when battling against OA. Thus, biomarkers with similar OA-responsive expressions in cartilage and synovium should be the potential targets for OA treatment. On the other hand, molecules with a distinguished response during OA in cartilage and synovium should be ruled out as OA therapeutic(s) to avoid controversial effects in different tissues. Here, to pave the path for developing a new generation of OA therapeutics, two published transcriptome datasets of knee articular cartilage and synovium were analyzed in-depth. Genes with statistically significantly different expression in OA and healthy cartilage were compared with those in the synovium. Thirty-five genes with similar OA-responsive expression in both tissues were identified while recognizing three genes with opposite OA-responsive alteration trends in cartilage and synovium. These genes were clustered based on the currently available knowledge, and the potential impacts of these clusters in OA were explored.
Collapse
Affiliation(s)
- Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Zhong Zheng
- Section of Orthodontics, Dental and Craniofacial Research Institute and Division of Growth and Development, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
407
|
Long-Term Effectiveness of Polymerized-Type I Collagen Intra-Articular Injections in Patients with Symptomatic Knee Osteoarthritis: Clinical and Radiographic Evaluation in a Cohort Study. Adv Orthop 2020; 2020:9398274. [PMID: 32802520 PMCID: PMC7416232 DOI: 10.1155/2020/9398274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/09/2020] [Indexed: 01/07/2023] Open
Abstract
Objective Polymerized-type I collagen (polymerized-collagen) is a downregulator of inflammation and a tissue regenerator. The aim was to evaluate the effect of intra-articular injections (IAIs) of polymerized-collagen among patients with symptomatic knee osteoarthritis (OA) in delaying or preventing joint replacement surgery. Patients and Methods. This was a cohort study of 309 patients with knee OA. Patients with mild-to-moderate disease were treated weekly with IAIs of 2 mL of polymerized-collagen for six weeks (n = 309). Follow-up was for 6–60 months. The primary endpoints included the following determinations: (1) therapeutic effect; (2) survival from total knee replacement surgery (TKR); (3) Western Ontario and McMaster University Osteoarthritis Index (WOMAC) and pain (visual analogue scale, VAS). Clinical improvement was defined as a decrease in pain exceeding 20 mm on the VAS and the achievement of at least 20% improvement from baseline with respect to the WOMAC score. Radiographic analysis was performed at baseline and 60 months. The joint space width in the medial, lateral, and patellofemoral compartments was calculated. Results Patients who received IAIs of polymerized-collagen had a statistically significant improvement in the primary criteria (p < 0.05). Kaplan–Meier survival analysis of the therapeutic effect demonstrated 98.8% survival at 60 months with TKR as the endpoint. There was no significant reduction in joint space in any compartment based on the analyzed radiographs. No serious adverse events were recorded. Conclusion Polymerized-collagen increased the time to TKR by at least 60 months, modifying the disease course, improving functional disability, and decreasing pain.
Collapse
|
408
|
Degenerative osteoarthritis a reversible chronic disease. Regen Ther 2020; 15:149-160. [PMID: 33426213 PMCID: PMC7770340 DOI: 10.1016/j.reth.2020.07.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/09/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most common chronic musculoskeletal disorder. It can affect any joint and is the most frequent single cause of disability in older adults. OA is a progressive degenerative disease involving the entire joint structure in a vicious circle that includes the capsule-bursa tissue inflammation, synovial fluid modifications, cartilage breakdown and erosions, osteochondral inflammatory damage leading to bone erosion and distortion. Research has identified the initial inflammatory-immunologic process that starts this vicious cycle leading to so-called early OA. Research has also identified the role played in the disease advancement by synoviocytes type A and B, chondrocytes, extracellular matrix, local immune-inflammatory mediators and proteases. This article investigates the joint-resident MSCs that play an essential local homeostatic role and regulate cell turn over and tissue repair. Resident MSCs establish and maintain a local regenerative microenvironment. The understanding of OA physiopathology clarifies the core mechanisms by which minimally invasive interventions might be able to halt and reverse the course of early stage OA. Interventions employing PRP, MSCs and exosomes are considered in this article.
Collapse
|
409
|
Yeh YT, Liang CC, Chang CL, Hsu CY, Li PC. Increased risk of knee osteoarthritis in patients using oral N-acetylcysteine: a nationwide cohort study. BMC Musculoskelet Disord 2020; 21:531. [PMID: 32778089 PMCID: PMC7418329 DOI: 10.1186/s12891-020-03562-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022] Open
Abstract
Background Knee osteoarthritis (OA) is known to be a progressive degenerative disorder; however, recent evidence suggests that inflammatory mediators contribute to cartilage degradation. Studies have reported that N-acetylcysteine (NAC) had a promising effect on the reduction of the synthesis of proinflammatory and structural mediators by synovial cells. Given the lack of relevant clinical trials, we conducted this study to determine the relationship between NAC use and risk of knee OA. Methods We designed a retrospective cohort study from 2000 to 2013. Patients who received oral NAC over 28 days within 1 year after the first prescription were defined as the case group, whereas those without NAC use were considered as candidates of the control group. We adopted 1:4 propensity-score matching by age, sex, index year, and comorbidities to obtain the control group. The primary outcome was a new diagnosis of knee OA during the follow-up period. Results Our study sample comprised 12,928 people who used NAC and 51,715 NAC nonusers. NAC users had a significantly higher incidence of osteoarthritis (adjusted hazard ratio: 1.42, P < .001) than did NAC nonusers. Also, in analyses stratified by age group and sex, all subgroups exhibited a significantly higher incidence of knee osteoarthritis (P < .0001) among NAC users than among NAC nonusers. The use of oral NAC was associated with nearly four-fold increased the risk of knee OA in the young age group. Conclusions Long-term use of oral NAC is associated with a higher risk of knee OA.
Collapse
Affiliation(s)
- Ying-Ting Yeh
- Department of Physical Medicine and Rehabilitation Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chung-Chao Liang
- Department of Physical Medicine and Rehabilitation Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chia-Ling Chang
- Management Office for Health Data, China Medical University Hospital, Taichung, Taiwan
| | - Chung-Y Hsu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Pei-Chen Li
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.
| |
Collapse
|
410
|
Impaired chondrocyte U3 snoRNA expression in osteoarthritis impacts the chondrocyte protein translation apparatus. Sci Rep 2020; 10:13426. [PMID: 32778764 PMCID: PMC7417995 DOI: 10.1038/s41598-020-70453-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
Although pathways controlling ribosome activity have been described to regulate chondrocyte homeostasis in osteoarthritis, ribosome biogenesis in osteoarthritis is unexplored. We hypothesized that U3 snoRNA, a non-coding RNA involved in ribosomal RNA maturation, is critical for chondrocyte protein translation capacity in osteoarthritis. U3 snoRNA was one of a number of snoRNAs with decreased expression in osteoarthritic cartilage and osteoarthritic chondrocytes. OA synovial fluid impacted U3 snoRNA expression by affecting U3 snoRNA gene promoter activity, while BMP7 was able to increase its expression. Altering U3 snoRNA expression resulted in changes in chondrocyte phenotype. Interference with U3 snoRNA expression led to reduction of rRNA levels and translational capacity, whilst induced expression of U3 snoRNA was accompanied by increased 18S and 28S rRNA levels and elevated protein translation. Whole proteome analysis revealed a global impact of reduced U3 snoRNA expression on protein translational processes and inflammatory pathways. For the first time we demonstrate implications of a snoRNA in osteoarthritis chondrocyte biology and investigated its role in the chondrocyte differentiation status, rRNA levels and protein translational capacity.
Collapse
|
411
|
Chakrabarti S, Ai M, Henson FM, Smith ESJ. Peripheral mechanisms of arthritic pain: A proposal to leverage large animals for in vitro studies. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2020; 8:100051. [PMID: 32817908 PMCID: PMC7426561 DOI: 10.1016/j.ynpai.2020.100051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 04/14/2023]
Abstract
Pain arising from musculoskeletal disorders such as arthritis is one of the leading causes of disability. Whereas the past 20-years has seen an increase in targeted therapies for rheumatoid arthritis (RA), other arthritis conditions, especially osteoarthritis, remain poorly treated. Although modulation of central pain pathways occurs in chronic arthritis, multiple lines of evidence indicate that peripherally driven pain is important in arthritic pain. To understand the peripheral mechanisms of arthritic pain, various in vitro and in vivo models have been developed, largely in rodents. Although rodent models provide numerous advantages for studying arthritis pathogenesis and treatment, the anatomy and biomechanics of rodent joints differ considerably to those of humans. By contrast, the anatomy and biomechanics of joints in larger animals, such as dogs, show greater similarity to human joints and thus studying them can provide novel insight for arthritis research. The purpose of this article is firstly to review models of arthritis and behavioral outcomes commonly used in large animals. Secondly, we review the existing in vitro models and assays used to study arthritic pain, primarily in rodents, and discuss the potential for adopting these strategies, as well as likely limitations, in large animals. We believe that exploring peripheral mechanisms of arthritic pain in vitro in large animals has the potential to reduce the veterinary burden of arthritis in commonly afflicted species like dogs, as well as to improve translatability of pain research into the clinic.
Collapse
Affiliation(s)
- Sampurna Chakrabarti
- Department of Neuroscience, Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
- Department of Pharmacology, University of Cambridge, UK
| | - Minji Ai
- Department of Veterinary Medicine, University of Cambridge, UK
| | | | | |
Collapse
|
412
|
Folkesson E, Turkiewicz A, Ali N, Rydén M, Hughes H, Tjörnstrand J, Önnerfjord P, Englund M. Proteomic comparison of osteoarthritic and reference human menisci using data-independent acquisition mass spectrometry. Osteoarthritis Cartilage 2020; 28:1092-1101. [PMID: 32407894 PMCID: PMC7397514 DOI: 10.1016/j.joca.2020.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/09/2020] [Accepted: 05/01/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Recent research in knee osteoarthritis (OA) highlights the role of the meniscus in OA pathology. Our aim was to compare the proteomes of medial and lateral menisci from end-stage medial compartment knee OA patients, with reference menisci from knee-healthy deceased donors, using mass spectrometry. DESIGN Tissue plugs of Ø3 mm were obtained from the posterior horns of the lateral and medial menisci from one knee of 10 knee-healthy deceased donors and 10 patients undergoing knee replacement. Proteins were extracted and prepared for mass spectrometric analysis. Statistical analysis was conducted on abundance data that was log2-transformed, using a linear mixed effects model and evaluated using pathway analysis. RESULTS We identified a total of 835 proteins in all samples, of which 331 were included in the statistical analysis. The largest differences could be seen between the medial menisci from OA patients and references, with most proteins showing higher intensities in the medial menisci from OA patients. Several matrix proteins, e.g., matrix metalloproteinase 3 (MMP3) (4.3 times higher values [95%CI 1.8, 10.6]), TIMP1 (3.5 [1.4, 8.5]), asporin (4.1 [1.7, 10.0]) and versican (4.4 [1.8, 10.9]), all showed higher abundance in medial menisci from OA patients compared to medial reference menisci. OA medial menisci also showed increased activation of several pathways involved in inflammation. CONCLUSION An increase in protein abundance for proteins such as MMP and TIMP1 in the medial menisci from OA patients suggests simultaneous activation of both catabolic and anabolic processes that warrants further attention.
Collapse
Affiliation(s)
- E. Folkesson
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Lund, Sweden,Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Rheumatology and Molecular Skeletal Biology, Lund, Sweden,Address correspondence and reprint requests to: E. Folkesson, Department of Clinical Sciences Lund, Lund University Molecular Skeletal Biology - BMC-C12, Klinikgatan 28, 221 00, Lund, Sweden.
| | - A. Turkiewicz
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Lund, Sweden
| | - N. Ali
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Lund, Sweden
| | - M. Rydén
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Lund, Sweden
| | - H.V. Hughes
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Lund, Sweden
| | - J. Tjörnstrand
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Lund, Sweden,Department of Orthopaedics, Skåne University Hospital, Lund, Sweden
| | - P. Önnerfjord
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Rheumatology and Molecular Skeletal Biology, Lund, Sweden
| | - M. Englund
- Lund University, Faculty of Medicine, Department of Clinical Sciences Lund, Orthopaedics, Clinical Epidemiology Unit, Lund, Sweden,Clinical Epidemiology Research and Training Unit, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
413
|
Szwedowski D, Szczepanek J, Paczesny Ł, Pękała P, Zabrzyński J, Kruczyński J. Genetics in Cartilage Lesions: Basic Science and Therapy Approaches. Int J Mol Sci 2020; 21:E5430. [PMID: 32751537 PMCID: PMC7432875 DOI: 10.3390/ijms21155430] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/31/2022] Open
Abstract
Cartilage lesions have a multifactorial nature, and genetic factors are their strongest determinants. As biochemical and genetic studies have dramatically progressed over the past decade, the molecular basis of cartilage pathologies has become clearer. Several homeostasis abnormalities within cartilaginous tissue have been found, including various structural changes, differential gene expression patterns, as well as altered epigenetic regulation. However, the efficient treatment of cartilage pathologies represents a substantial challenge. Understanding the complex genetic background pertaining to cartilage pathologies is useful primarily in the context of seeking new pathways leading to disease progression as well as in developing new targeted therapies. A technology utilizing gene transfer to deliver therapeutic genes to the site of injury is quickly becoming an emerging approach in cartilage renewal. The goal of this work is to provide an overview of the genetic basis of chondral lesions and the different approaches of the most recent systems exploiting therapeutic gene transfer in cartilage repair. The integration of tissue engineering with viral gene vectors is a novel and active area of research. However, despite promising preclinical data, this therapeutic concept needs to be supported by the growing body of clinical trials.
Collapse
Affiliation(s)
- Dawid Szwedowski
- Orthopedic Arthroscopic Surgery International (O.A.S.I.) Bioresearch Foundation, Gobbi N.P.O., 20133 Milan, Italy;
- Department of Orthopaedics and Trauma Surgery, Provincial Polyclinical Hospital, 87100 Torun, Poland
| | - Joanna Szczepanek
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, 87100 Torun, Poland
| | - Łukasz Paczesny
- Orvit Clinic, Citomed Healthcare Center, 87100 Torun, Poland; (Ł.P.); (J.Z.)
| | - Przemysław Pękała
- Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, 30705 Krakow, Poland;
| | - Jan Zabrzyński
- Orvit Clinic, Citomed Healthcare Center, 87100 Torun, Poland; (Ł.P.); (J.Z.)
| | - Jacek Kruczyński
- Department of General Orthopaedics, Musculoskeletal Oncology and Trauma Surgery, Poznan University of Medical Sciences, 60512 Poznań, Poland;
| |
Collapse
|
414
|
Greif DN, Kouroupis D, Murdock CJ, Griswold AJ, Kaplan LD, Best TM, Correa D. Infrapatellar Fat Pad/Synovium Complex in Early-Stage Knee Osteoarthritis: Potential New Target and Source of Therapeutic Mesenchymal Stem/Stromal Cells. Front Bioeng Biotechnol 2020; 8:860. [PMID: 32850724 PMCID: PMC7399076 DOI: 10.3389/fbioe.2020.00860] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022] Open
Abstract
The infrapatellar fat pad (IFP) has until recently been viewed as a densely vascular and innervated intracapsular/extrasynovial tissue with biomechanical roles in the anterior compartment of the knee. Over the last decade, secondary to the proposition that the IFP and synovium function as a single unit, its recognized tight molecular crosstalk with emerging roles in the pathophysiology of joint disease, and the characterization of immune-related resident cells with varying phenotypes (e.g., pro and anti-inflammatory macrophages), this structural complex has gained increasing attention as a potential therapeutic target in patients with various knee pathologies including osteoarthritis (KOA). Furthermore, the description of the presence of mesenchymal stem/stromal cells (MSC) as perivascular cells within the IFP (IFP-MSC), exhibiting immunomodulatory, anti-fibrotic and neutralizing activities over key local mediators, has promoted the IFP as an alternative source of MSC for cell-based therapy protocols. These complementary concepts have supported the growing notion of immune and inflammatory events participating in the pathogenesis of KOA, with the IFP/synovium complex engaging not only in amplifying local pathological responses, but also as a reservoir of potential therapeutic cell-based products. Consequently, the aim of this review is to outline the latest discoveries related with the IFP/synovium complex as both an active participant during KOA initiation and progression thus emerging as a potential target, and a source of therapeutic IFP-MSCs. Finally, we discuss how these notions may help the design of novel treatments for KOA through modulation of local cellular and molecular cascades that ultimately lead to joint destruction.
Collapse
Affiliation(s)
- Dylan N Greif
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Christopher J Murdock
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Lee D Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
415
|
TissueGene-C promotes an anti-inflammatory micro-environment in a rat monoiodoacetate model of osteoarthritis via polarization of M2 macrophages leading to pain relief and structural improvement. Inflammopharmacology 2020; 28:1237-1252. [PMID: 32696209 PMCID: PMC7524813 DOI: 10.1007/s10787-020-00738-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/23/2020] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) is the most common form of arthritis, characterized by cartilage destruction, pain and inflammation in the joints. Existing medications can provide relief from the symptoms, but their effects on the progression of the disease are limited. TissueGene-C (TG-C) is a novel cell and gene therapy for the treatment of OA, comprising a mixture of human allogeneic chondrocytes and irradiated cells engineered to overexpress transforming growth factor-β1 (TGF-β1). This study aims to investigate the efficacy and mechanism of action of TG-C in a rat model of OA. Using the monosodium-iodoacetate (MIA) model of OA, we examined whether TG-C could improve OA symptoms and cartilage structure in rats. Our results showed that TG-C provided pain relief and cartilage structural improvement in the MIA OA model over 56 days. In parallel with these long-term effects, cytokine profiles obtained on day 4 revealed increased expression of interleukin-10 (IL-10), an anti-inflammatory cytokine, in the synovial lavage fluid. Moreover, the increased levels of TGF-β1 and IL-10 caused by TG-C induced the expression of arginase 1, a marker of M2 macrophages, and decreased the expression of CD86, a marker of M1 macrophages. These results suggest that TG-C exerts a beneficial effect on OA by inducing a M2 macrophage-dominant micro-environment. Cell therapy using TG-C may be a promising strategy for targeting the underlying pathogenic mechanisms of OA, reducing pain, improving function, and creating a pro-anabolic micro-environment. This environment supports cartilage structure regeneration and is worthy of further evaluation in future clinical trials.
Collapse
|
416
|
Whyte A, Boeddinghaus R, Bartley A, Vijeyaendra R. Imaging of the temporomandibular joint. Clin Radiol 2020; 76:76.e21-76.e35. [PMID: 32709388 DOI: 10.1016/j.crad.2020.06.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022]
Abstract
Temporomandibular disorders are common, especially in young to middle-aged women, and most settle with supportive treatment. Imaging is indicated for the small percentage of cases that do not respond to conservative management and when the diagnosis is no doubt. The temporomandibular joint (TMJ) is a bilateral synovial articulation between the mandible and skull base. It has an intra-articular disc dividing the joint into superior and inferior compartments and the articular surfaces are lined with fibrocartilage. The normal imaging anatomy of the TMJ is described and illustrated. Different movements occur in each joint compartments: a hinge movement in the inferior joint space and translation or gliding in the superior joint space. Internal derangement is the commonest disorder affecting the TMJ and is most commonly due to disc displacement, followed by osteoarthritis and inflammatory arthritides. The imaging findings, primarily on magnetic resonance imaging (MRI) and computed tomography (CT), of internal derangement and less common disorders of the joint, are reviewed and illustrated. Optimal imaging protocols are discussed with detailed reporting guidelines.
Collapse
Affiliation(s)
- A Whyte
- Perth Radiological Clinic, 127 Hamersley Rd, Subiaco WA 6008, Australia; Department of Dentistry, University of Western Australia, Nedlands, WA 6009, Australia; Department of Medicine and Radiology, University of Melbourne, Carlton, Victoria 3000, Australia.
| | - R Boeddinghaus
- Perth Radiological Clinic, 127 Hamersley Rd, Subiaco WA 6008, Australia; Department of Surgery, University of Western Australia, Nedlands WA 6009, Australia
| | - A Bartley
- Perth Radiological Clinic, 127 Hamersley Rd, Subiaco WA 6008, Australia; Medical Imaging, Perth Children's Hospital, 15 Hospital Avenue, Nedlands WA 6009, Australia
| | - R Vijeyaendra
- Irwin Dental Clinic - Army Barracks, Samichon Road, Karrakatta WA 6010, Australia
| |
Collapse
|
417
|
Cheng HJ, Hsu WT, Chen CN, Li C. Activation of NOTCH1 by Shear Force Elicits Immediate Cytokine Expression in Human Chondrocytes. Int J Mol Sci 2020; 21:ijms21144958. [PMID: 32674293 PMCID: PMC7404062 DOI: 10.3390/ijms21144958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/04/2020] [Accepted: 07/10/2020] [Indexed: 01/01/2023] Open
Abstract
Osteoarthritis is caused by overloading of joints and is characterized by inflammation-induced disruption of cartilage structure. Current treatment strategy aims to relieve inflammation and prevent further deterioration of joint function. However, how mechanical force leads to inflammation and deterioration of chondrocyte function still remains incompletely understood. To explore the force-regulated molecular mechanism, an in vitro hydraulic shear force experiment to simulate the condition of force loading was required. The result demonstrated that multiple cytokines and immune regulators, including interleukin 8, interferon β, TRAF1 and TNFAIP3, were significantly increased by shear force within two hours of treatment. Moreover, JAG1 and HES1 were drastically upregulated as well, suggesting that NOTCH1 signaling is activated by shear force. Short-term expression of NOTCH1 intracellular domain activated a similar set of cytokines, indicating that NOTCH1 responds to shear force and activates downstream genes. When incubated under the medium conditioned by NOTCH1-activated chondrocyte, osteoblasts expressed higher levels of interferon β and interferon λ. Together, our results indicated that NOTCH1 functions as a force sensor and promotes expression of cytokines and immune regulators from shear-force bearing chondrocytes.
Collapse
Affiliation(s)
- Hao-Jen Cheng
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 621, Taiwan; (H.-J.C.); (W.-T.H.)
- Department of Orthopedics, Shinnhomei Clinic, Chiayi 600, Taiwan
| | - Wan-Ting Hsu
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 621, Taiwan; (H.-J.C.); (W.-T.H.)
| | - Cheng-Nan Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi 600, Taiwan;
| | - Chin Li
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi 621, Taiwan; (H.-J.C.); (W.-T.H.)
- Correspondence: ; Tel.: 886-5-272-0411; Fax: 886-5-272-2871
| |
Collapse
|
418
|
Wright T, Hope V, Ciccarone D, Lewer D, Scott J, Harris M. Prevalence and severity of abscesses and cellulitis, and their associations with other health outcomes, in a community-based study of people who inject drugs in London, UK. PLoS One 2020; 15:e0235350. [PMID: 32663203 PMCID: PMC7360031 DOI: 10.1371/journal.pone.0235350] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/12/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Skin and soft tissue infections (SSTI) are a common but preventable cause of morbidity and mortality among people who inject drugs (PWID). They can be severe, and hospitalisations of PWID with SSTI are rising. The most common SSTI presentations are abscesses and cellulitis. METHODS We used data from Care & Prevent, a cross-sectional community survey of PWID in London. We reported the lifetime prevalence of SSTI, severity of infections, key risk factors, and associated sequelae. Pictorial questions were used to assess SSTI severity. RESULTS We recruited 455 PWID. SSTI lifetime prevalence was high: 64% reported an abscess and/or cellulitis. Over one-third (37%) reported a severe infection, 137 (47%) reported hospitalisation. SSTIrisk factors were: aged 35+ years, injecting once or more times a day, subcutaneous or intra-muscular injections, and making four or more attempts to achieve an injection. Those who reported having other health conditions were at higher odds of having an abscess or cellulitis, with risk tending to increase with number of reported conditions. Half (46%) employed self-care for their worst SSTI, and 43% waited for ten or more days before seeking medical care or not seeking medical care at all. CONCLUSIONS Abscess and cellulitis are very common among PWID in London. We corroborate findings indicating SSTIs are associated with risks, e.g. venous access problems, as well as other co-morbid conditions: septicaemia, endocarditis, DVT, and kidney disease. These co-morbidities may impact SSTIs severity and outcomes. Delayed healthcare seeking potentially exacerbates infection severity, which in turn increases poorer health outcomes and complications.
Collapse
Affiliation(s)
- Talen Wright
- Department of Public Health, Environments & Society, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Vivian Hope
- Public Health Institute, Liverpool John Moores University, Liverpool, United Kingdom
| | - Daniel Ciccarone
- Department of Family and Community Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| | - Dan Lewer
- Institute of Epidemiology and Healthcare, University College London, London, United Kingdom
| | - Jenny Scott
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, United Kingdom
| | - Magdalena Harris
- Department of Public Health, Environments & Society, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
419
|
Kim YG, Choi J, Kim K. Mesenchymal Stem Cell‐Derived Exosomes for Effective Cartilage Tissue Repair and Treatment of Osteoarthritis. Biotechnol J 2020; 15:e2000082. [DOI: 10.1002/biot.202000082] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/25/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Young Guk Kim
- Department of Chemical and Biochemical Engineering Dongguk University 30 Pildong‐ro 1‐gil Seoul 04620 Republic of Korea
| | - Jonghoon Choi
- School of Integrative Engineering Chung‐Ang University 47 Heukseok‐ro Seoul 06911 Republic of Korea
| | - Kyobum Kim
- Department of Chemical and Biochemical Engineering Dongguk University 30 Pildong‐ro 1‐gil Seoul 04620 Republic of Korea
| |
Collapse
|
420
|
Lord MS, Melrose J, Day AJ, Whitelock JM. The Inter-α-Trypsin Inhibitor Family: Versatile Molecules in Biology and Pathology. J Histochem Cytochem 2020; 68:907-927. [PMID: 32639183 DOI: 10.1369/0022155420940067] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Inter-α-trypsin inhibitor (IαI) family members are ancient and unique molecules that have evolved over several hundred million years of vertebrate evolution. IαI is a complex containing the proteoglycan bikunin to which heavy chain proteins are covalently attached to the chondroitin sulfate chain. Besides its matrix protective activity through protease inhibitory action, IαI family members interact with extracellular matrix molecules and most notably hyaluronan, inhibit complement, and provide cell regulatory functions. Recent evidence for the diverse roles of the IαI family in both biology and pathology is reviewed and gives insight into their pivotal roles in tissue homeostasis. In addition, the clinical uses of these molecules are explored, such as in the treatment of inflammatory conditions including sepsis and Kawasaki disease, which has recently been associated with severe acute respiratory syndrome coronavirus 2 infection in children.
Collapse
Affiliation(s)
- Megan S Lord
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia
| | - James Melrose
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia.,Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St. Leonards, NSW, Australia.,Sydney Medical School, Northern, Sydney University, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research and Lydia Becker Institute of Immunology and Inflammation, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - John M Whitelock
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia.,Stem Cell Extracellular Matrix & Glycobiology, Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, Faculty of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
421
|
Synovial cell cross-talk with cartilage plays a major role in the pathogenesis of osteoarthritis. Sci Rep 2020; 10:10868. [PMID: 32616761 PMCID: PMC7331607 DOI: 10.1038/s41598-020-67730-y] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 06/12/2020] [Indexed: 01/13/2023] Open
Abstract
We elucidated the molecular cross-talk between cartilage and synovium in osteoarthritis, the most widespread arthritis in the world, using the powerful tool of single-cell RNA-sequencing. Multiple cell types were identified based on profiling of 10,640 synoviocytes and 26,192 chondrocytes: 12 distinct synovial cell types and 7 distinct articular chondrocyte phenotypes from matched tissues. Intact cartilage was enriched for homeostatic and hypertrophic chondrocytes, while damaged cartilage was enriched for prefibro- and fibro-, regulatory, reparative and prehypertrophic chondrocytes. A total of 61 cytokines and growth factors were predicted to regulate the 7 chondrocyte cell phenotypes. Based on production by > 1% of cells, 55% of the cytokines were produced by synovial cells (39% exclusive to synoviocytes and not expressed by chondrocytes) and their presence in osteoarthritic synovial fluid confirmed. The synoviocytes producing IL-1beta (a classic pathogenic cytokine in osteoarthritis), mainly inflammatory macrophages and dendritic cells, were characterized by co-expression of surface proteins corresponding to HLA-DQA1, HLA-DQA2, OLR1 or TLR2. Strategies to deplete these pathogenic intra-articular cell subpopulations could be a therapeutic option for human osteoarthritis.
Collapse
|
422
|
Kouroupis D, Bowles AC, Best TM, Kaplan LD, Correa D. CD10/Neprilysin Enrichment in Infrapatellar Fat Pad-Derived Mesenchymal Stem Cells Under Regulatory-Compliant Conditions: Implications for Efficient Synovitis and Fat Pad Fibrosis Reversal. Am J Sports Med 2020; 48:2013-2027. [PMID: 32427493 DOI: 10.1177/0363546520917699] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Synovitis and infrapatellar fat pad (IFP) fibrosis participate in various conditions of the knee. Substance P (SP), a neurotransmitter secreted within those structures and historically associated with nociception, also modulates local neurogenic inflammatory and fibrotic responses. Exposure of IFP mesenchymal stem cells (IFP-MSCs) to a proinflammatory/profibrotic environment (ex vivo priming with TNFα, IFNγ, and CTGF) induces their expression of CD10/neprilysin, effectively degrading SP in vitro and in vivo. PURPOSE/HYPOTHESIS The purpose was to test the therapeutic effects of IFP-MSCs processed under regulatory-compliant protocols, comparing them side-by-side with standard fetal bovine serum (FBS)-grown cells. The hypothesis was that when processed under such protocols, IFP-MSCs do not require ex vivo priming to acquire a CD10-rich phenotype efficiently degrading SP and reversing synovitis and IFP fibrosis. STUDY DESIGN Controlled laboratory study. METHODS Human IFP-MSCs were processed in FBS or either of 2 alternative conditions-regulatory-compliant pooled human platelet lysate (hPL) and chemically reinforced medium (Ch-R)-and then subjected to proinflammatory/profibrotic priming with TNFα, IFNγ, and CTGF. Cells were assessed for in vitro proliferation, stemness, immunophenotype, differentiation potential, transcriptional and secretory profiles, and SP degradation. Based on a rat model of acute synovitis and IFP fibrosis, the in vivo efficacy of cells degrading SP plus reversing structural signs of inflammation and fibrosis was assessed. RESULTS When compared with FBS, IFP-MSCs processed with either hPL or Ch-R exhibited a CD10High phenotype and showed enhanced proliferation, differentiation, and immunomodulatory transcriptional and secretory profiles (amplified by priming). Both methods recapitulated and augmented the secretion of growth factors seen with FBS plus priming, with some differences between them. Functionally, in vitro SP degradation was more efficient in hPL and Ch-R, confirmed upon intra-articular injection in vivo where CD10-rich IFP-MSCs also dramatically reversed signs of synovitis and IFP fibrosis even without priming or at significantly lower cell doses. CONCLUSION hPL and Ch-R formulations can effectively replace FBS plus priming to induce specific therapeutic attributes in IFP-MSCs. The resulting fine-tuned, regulatory-compliant, cell-based product has potential future utilization as a novel minimally invasive cell therapy for the treatment of synovitis and IFP fibrosis. CLINICAL RELEVANCE The therapeutic enhancement of IFP-MSCs manufactured under regulatory-compliant conditions suggests that such a strategy could accelerate the time from preclinical to clinical phases. The therapeutic efficacy obtained at lower MSC numbers than currently needed and the avoidance of cell priming for efficient results could have a significant effect on the design of clinical protocols to potentially treat conditions involving synovitis and IFP fibrosis.
Collapse
Affiliation(s)
- Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Annie C Bowles
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Department of Biomedical Engineering, College of Engineering, University of Miami, Miami, Florida, USA
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Lee D Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
423
|
Hip Joint Effusion-Synovitis Is Associated With Hip Pain and Sports/Recreation Function in Female Professional Ballet Dancers. Clin J Sport Med 2020; 30:341-347. [PMID: 32639441 DOI: 10.1097/jsm.0000000000000595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To compare hip joint effusion-synovitis prevalence in professional ballet dancers with nondancing athletes and to evaluate the relationship between effusion-synovitis and clinical measures and cartilage defects. DESIGN Case-control study. SETTING Elite ballet and sport. PARTICIPANTS Forty-nine professional ballet dancers and 49 age-matched and sex-matched athletes. INDEPENDENT VARIABLES Group (dancers/athletes), sex, age, years of training, Copenhagen Hip and Groin Outcome Scores (HAGOSs), hip rotation range of motion (ROM), generalized joint hypermobility (GJH), and hip cartilage defect scores. MAIN OUTCOME MEASURES Hip joint effusion-synovitis (absent, grade 1 = 2-4 mm, grade 2 = >4 mm) scored with 3-Tesla magnetic resonance imaging. RESULTS Hip joint effusion-synovitis was found in 22 (45%) dancers and 13 (26.5%) athletes (P = 0.06). Grade 2 effusion-synovitis was only found in dancers (n = 8, r = 0.31, P = 0.009). The prevalence of effusion-synovitis was similar in men (n = 11, 26%) and women (n = 24, 43%, P = 0.09). Female dancers with effusion-synovitis had lower HAGOS pain (r = 0.63, P = 0.001) and sports/recreation scores (r = 0.66, P = 0.001) compared with those without effusion-synovitis. The HAGOS scores were not related to effusion-synovitis in male dancers or female and male athletes (P > 0.01 for all). Effusion-synovitis was not related to hip ROM, GJH, or cartilage defect scores (P > 0.05 for all). CONCLUSIONS Hip joint effusion-synovitis was related to higher levels of pain and lower sports/recreation function in female ballet dancers. Effusion-synovitis was not related to hip rotation ROM, GJH or cartilage defects. Larger sized joint effusion-synovitis was exclusively found in dancers.
Collapse
|
424
|
Differential patterns of pathology in and interaction between joint tissues in long-term osteoarthritis with different initiating causes: phenotype matters. Osteoarthritis Cartilage 2020; 28:953-965. [PMID: 32360537 DOI: 10.1016/j.joca.2020.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/23/2020] [Accepted: 04/15/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine if osteoarthritis (OA) progression and joint tissue-pathology associations link specific animal models to different human OA phenotypes. DESIGN Male 11-week-old C57BL6 mice had unilateral medial-meniscal-destabilization (DMM) or antigen-induced-arthritis (AIA). Joint tissue histopathology was scored day-3 to week-16. Tissue-pathology associations (corrected for time and at week-16) were determined by partial correlation coefficients, and odds ratios (OR) calculated for likelihood of cartilage damage and joint inflammation by ordinal-logistic-regression. RESULTS Despite distinct temporal patterns of progression, by week-16 joint-wide OA pathology in DMM and AIA was equivalent. Significant pathology associations common to both models included: osteophyte size and maturity (r > 0.4); subchondral bone (SCB) sclerosis and osteophyte maturity (r > 0.25); cartilage erosion and chondrocyte hypertrophy/apoptosis (r > 0.4), SCB sclerosis (r > 0.26), osteophyte size (r > 0.3), and maturity (r > 0.32). DMM-specific associations were between cartilage proteoglycan loss and structural damage (r = 0.56), osteophyte maturity (r = 0.49), size (r = 0.45), and SCB sclerosis (r = 0.28). AIA-specific associations were between SCB sclerosis and chondrocyte hypertrophy/apoptosis (r = 0.40) and osteophyte size (r = 0.37); and synovitis with cartilage structural damage (r = 0.18). No tissue-pathology associations were common to both models at week-16. Increased likelihood of cartilage structural damage was associated with: chondrocyte hypertrophy/apoptosis (OR>1.7), and osteophyte size (OR>2.3) in both models; SCB sclerosis (OR = 2.0) and proteoglycan loss (OR = 2.4) in DMM; and synovitis (OR = 1.2) in AIA. Joint inflammation was associated positively with cartilage proteoglycan loss (OR = 1.4) and inversely with osteophyte size (OR = 0.21) in AIA only. CONCLUSION This study highlights the importance of defining OA-models by initiating mechanisms and progression, not just end-stage joint-tissue pathology.
Collapse
|
425
|
Maleitzke T, Elazaly H, Festbaum C, Eder C, Karczewski D, Perka C, Duda GN, Winkler T. Mesenchymal Stromal Cell-Based Therapy-An Alternative to Arthroplasty for the Treatment of Osteoarthritis? A State of the Art Review of Clinical Trials. J Clin Med 2020; 9:jcm9072062. [PMID: 32630066 PMCID: PMC7409016 DOI: 10.3390/jcm9072062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 01/06/2023] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disorder worldwide and to date no regenerative treatment has been established in clinical practice. This review evaluates the current literature on the clinical translation of mesenchymal stromal cell (MSC)-based therapy in OA management with a focus on safety, outcomes and procedural specifics. PubMed, Cochrane Library and clinicaltrials.gov were searched for clinical studies using MSCs for OA treatment. 290 articles were initially identified and 42 articles of interest, including a total of 1325 patients, remained for further examination. Most of the included studies used adipose tissue-derived MSCs or bone-marrow-derived MSCs to treat patients suffering from knee OA. MSC-based therapy for knee OA appears to be safe and presumably effective in selected parameters. Yet, a direct comparison between studies was difficult due to a pronounced variance regarding methodology, assessed outcomes and evidence levels. Intensive scientific engagement is needed to identify the most effective source and dosage of MSCs for OA treatment in the future. Consent on outcome measures has to be reached and eventually patient sub-populations need to be identified that will profit most from MSC-based treatment for OA.
Collapse
Affiliation(s)
- Tazio Maleitzke
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Hisham Elazaly
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Christian Festbaum
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
| | - Christian Eder
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
| | - Daniel Karczewski
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
| | - Carsten Perka
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Tobias Winkler
- Center for Musculoskeletal Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (T.M.); (C.F.); (C.E.); (D.K.); (C.P.)
- Julius Wolff Institute, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; (H.E.); (G.N.D.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-559084
| |
Collapse
|
426
|
Frischholz S, Berberich O, Böck T, Meffert RH, Blunk T. Resveratrol counteracts IL-1β-mediated impairment of extracellular matrix deposition in 3D articular chondrocyte constructs. J Tissue Eng Regen Med 2020; 14:897-908. [PMID: 32181981 DOI: 10.1002/term.3031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/05/2020] [Accepted: 03/06/2020] [Indexed: 11/09/2022]
Abstract
When aiming at cell-based therapies in osteoarthritis (OA), proinflammatory conditions mediated by cytokines such as IL-1β need to be considered. In recent studies, the phytoalexin resveratrol (RSV) has exhibited potent anti-inflammatory properties. However, long-term effects on 3D cartilaginous constructs under inflammatory conditions with regard to tissue quality, especially extracellular matrix (ECM) composition, have remained unexplored. Therefore, we employed long-term model cultures for cell-based therapies in an in vitro OA environment and evaluated effects of RSV. Pellet constructs made from expanded porcine articular chondrocytes were cultured with either IL-1β (1-10 ng/ml) or RSV (50 μM) alone, or a cotreatment with both agents. Treatments were applied for 14 days, either directly after pellet formation or after a preculture period of 7 days. Culture with IL-1β (10 ng/ml) decreased pellet size and DNA amount and severely compromised glycosaminoglycan (GAG) and collagen content. Cotreatment with RSV distinctly counteracted the proinflammatory catabolism and led to partial rescue of the ECM composition in both culture systems, with especially strong effects on GAG. Marked MMP13 expression was detected in IL-1β-treated pellets, but none upon RSV cotreatment. Expression of collagen type I was increased upon IL-1β treatment and still observed when adding RSV, whereas collagen type X, indicating hypertrophy, was detected exclusively in pellets treated with RSV alone. In conclusion, RSV can counteract IL-1β-mediated degradation and distinctly improve cartilaginous ECM deposition in 3D long-term inflammatory cultures. Nevertheless, potential hypertrophic effects should be taken into account when considering RSV as cotreatment for articular cartilage repair techniques.
Collapse
Affiliation(s)
- Sebastian Frischholz
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| | - Oliver Berberich
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| | - Thomas Böck
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| | - Rainer H Meffert
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University of Würzburg, Würzburg, Germany
| |
Collapse
|
427
|
Kruppel-like factor 4 upregulates matrix metalloproteinase 13 expression in chondrocytes via mRNA stabilization. Cell Tissue Res 2020; 382:307-319. [PMID: 32556726 DOI: 10.1007/s00441-020-03228-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 04/29/2020] [Indexed: 10/24/2022]
Abstract
Matrix metalloproteinase 13 (MMP13) is indispensable for normal skeletal development and is also a principal proteinase responsible for articular joint pathologies. MMP13 mRNA level needs to be tightly regulated in both positive and negative manners to achieve normal development and also to prevent joint destruction. We showed previously that Kruppel-like factor 4 (KLF4) strongly induces the expression of members of the MMP family of genes including that for MMP13 in cultured chondrocytes. Through expression-based screening of approximately 400 compounds, we identified several that efficiently downregulated MMP13 gene expression induced by KLF4. Compounds grouped as topoisomerase inhibitors (transcriptional inhibitors) downregulated MMP13 expression levels, which proved the validity of our screening method. In this screening, trichostatin A (TSA) was identified as one of the most potent repressors. Mechanistically, increased MMP13 mRNA levels induced by KLF4 were not mainly caused by increased rates of RNA polymerase II-mediated MMP13 transcription, but arose from escaping mRNA decay. TSA treatment almost completely blunted the effect of KLF4. Importantly, KLF4 was detected in chondrocytes at the joint destruction sites in a rodent model of osteoarthritis. Our results partially explain how KLF4 regulates numerous proteinase gene expressions simultaneously in chondrocytes. Also, these observations suggest that modulation of KLF4 activity or expression could be a novel therapeutic target for osteoarthritis.
Collapse
|
428
|
Alahdal M, Duan L, Ouyang H, Wang D. The role of indoleamine 2,3 dioxygenase 1 in the osteoarthritis. Am J Transl Res 2020; 12:2322-2343. [PMID: 32655775 PMCID: PMC7344072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/05/2020] [Indexed: 06/11/2023]
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease and a leading cause of disability. It involves articular cartilage destruction and a whole joint inflammation. In spite of OA pathogenesis is still unclear, new studies on the OA pathophysiological aetiology and immunomodulation therapy continuously achieve significant advances with new concepts. Here, we focus on the indoleamine-2,3-dioxygenase1 (IDO1) activity in the osteoarthritis (OA), which is one of the noticeable enzymes in the synovial fluid of arthritis patients. It was recognized as an essential mediator of autoreactive B and T cell responses in rheumatoid arthritis (RA) and an interesting therapeutic target against RA. However, the role IDO1 plays in the OA pathogenesis hasn't been discussed. The new OA experimental analysis evidenced IDO1 overexpression in the synovial fluid of OA patients, and recent studies reported that IDO1 metabolites were found higher in the OA synovial fluid than RA and spondyloarthropathies (SpA) patients. Moreover, the positive relation of IDO1 metabolites with OA pain and joint stiffness has been confirmed. Thus, the IDO1 plays a pivotal role in the pathogenesis of OA. In this review, the role IDO1 plays in the OA pathogenesis has been deeply discussed. It could be a promising target in the immunotherapy of OA disease.
Collapse
Affiliation(s)
- Murad Alahdal
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen 518035, P. R. China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of MedicineHangzhou, P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen 518035, P. R. China
| | - Li Duan
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen 518035, P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen 518035, P. R. China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of MedicineHangzhou, P. R. China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Shenzhen Second People’s Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center)Shenzhen 518035, P. R. China
- Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic TechnologyShenzhen 518035, P. R. China
| |
Collapse
|
429
|
Bariguian Revel F, Fayet M, Hagen M. Topical Diclofenac, an Efficacious Treatment for Osteoarthritis: A Narrative Review. Rheumatol Ther 2020; 7:217-236. [PMID: 32086778 PMCID: PMC7211216 DOI: 10.1007/s40744-020-00196-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Indexed: 12/26/2022] Open
Abstract
Multiple head-to-head trials have demonstrated that topical nonsteroidal anti-inflammatory drugs (NSAIDs), including topical diclofenac, provide at least equivalent analgesia, improvement in physical function, and reduction of stiffness compared with oral NSAIDs in osteoarthritis and have fewer systemic adverse events. While efficacy of topical diclofenac in osteoarthritis is well established, understanding of the time to onset of action, duration of effect, and the minimum effective concentration is limited. Factors likely to influence these parameters include drug penetration and localization. Diclofenac concentrations in the joint tissues are likely to be more relevant than plasma concentrations. However, although diclofenac penetrates and is retained in these "effect compartments" at the site of inflammation and drug activity, no specific minimum effective concentration of diclofenac in plasma or synovial tissue has been identified. Recent evidence suggests that a reduction in inflammatory markers may be a better predictor of efficacy than plasma concentrations. This narrative review explores existing evidence in these areas and identifies the gaps where further research is needed. Based on our findings, topical NSAIDs such as diclofenac should be considered as a guideline-supported, generally well-tolerated, and effective first-line treatment option for knee and hand OA, especially for older patients and those who have comorbid conditions and/or risk factors for various systemic (gastrointestinal, hepatic, renal, or cardiovascular) adverse events associated with oral NSAIDs, particularly at high doses and with long-term use.
Collapse
Affiliation(s)
| | - Marina Fayet
- GSK Consumer Healthcare S.A, Route de l'Etraz 2, 1260, Nyon, Switzerland
| | - Martina Hagen
- GSK Consumer Healthcare S.A, Route de l'Etraz 2, 1260, Nyon, Switzerland.
| |
Collapse
|
430
|
Hedderich J, El Bagdadi K, Angele P, Grässel S, Meurer A, Straub RH, Zaucke F, Jenei-Lanzl Z. Norepinephrine Inhibits the Proliferation of Human Bone Marrow-Derived Mesenchymal Stem Cells via β2-Adrenoceptor-Mediated ERK1/2 and PKA Phosphorylation. Int J Mol Sci 2020; 21:ijms21113924. [PMID: 32486305 PMCID: PMC7312191 DOI: 10.3390/ijms21113924] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/14/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) represent an alternative to chondrocytes to support cartilage regeneration in osteoarthritis (OA). The sympathetic neurotransmitter norepinephrine (NE) has been shown to inhibit their chondrogenic potential; however, their proliferation capacity under NE influence has not been studied yet. Therefore, we used BMSCs obtained from trauma and OA donors and compared the expression of adrenergic receptors (AR). Then, BMSCs from both donor groups were treated with NE, as well as with combinations of NE and α1-, α2- or β1/2-AR antagonists (doxazosin, yohimbine or propranolol). Activation of AR-coupled signaling was investigated by analyzing ERK1/2 and protein kinase A (PKA) phosphorylation. A similar but not identical subset of ARs was expressed in trauma (α2B-, α2C- and β2-AR) and OA BMSCs (α2A-, α2B-, and β2-AR). NE in high concentrations inhibited the proliferation of both trauma and OA BMCSs significantly. NE in low concentrations did not influence proliferation. ERK1/2 as well as PKA were activated after NE treatment in both BMSC types. These effects were abolished only by propranolol. Our results demonstrate that NE inhibits the proliferation and accordingly lowers the regenerative capacity of human BMSCs likely via β2-AR-mediated ERK1/2 and PKA phosphorylation. Therefore, targeting β2-AR-signaling might provide novel OA therapeutic options.
Collapse
Affiliation(s)
- Jessica Hedderich
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, 60528 Frankfurt/Main, Germany; (J.H.); (K.E.B.); (A.M.); (F.Z.)
| | - Karima El Bagdadi
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, 60528 Frankfurt/Main, Germany; (J.H.); (K.E.B.); (A.M.); (F.Z.)
| | - Peter Angele
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Susanne Grässel
- Department of Orthopedic Surgery, Experimental Orthopedics, Centre for Medical Biotechnology (ZMB), University of Regensburg, 93053 Regensburg, Germany;
| | - Andrea Meurer
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, 60528 Frankfurt/Main, Germany; (J.H.); (K.E.B.); (A.M.); (F.Z.)
| | - Rainer H. Straub
- Laboratory of Experimental Rheumatology and Neuroendocrine Immunology, Department of Internal Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany;
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, 60528 Frankfurt/Main, Germany; (J.H.); (K.E.B.); (A.M.); (F.Z.)
| | - Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, 60528 Frankfurt/Main, Germany; (J.H.); (K.E.B.); (A.M.); (F.Z.)
- Correspondence: ; Tel.: +49-69-6705-408
| |
Collapse
|
431
|
Zhao T, Ma C, Xie B, Zhao B, Wang W, Liu J. Evaluation of Common Variants in the AKNA Gene and Susceptibility to Knee Osteoarthritis Among the Han Chinese. Genet Test Mol Biomarkers 2020; 24:425-430. [PMID: 32460535 DOI: 10.1089/gtmb.2020.0014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: Osteoarthritis (OA) is a complex degenerative joint disease that is associated with both genetic and environmental factors. The AKNA gene, located at 9q32, has recently been identified as being associated with knee osteoarthritis (KOA) in the Mexican population. Our aim was to investigate the relationship of common variants in this gene with the risk of KOA in a large Han Chinese population. Methods: A total of 2,500 Han Chinese subjects were recruited, consisting of 824 KOA patients and 1,676 controls. Eight tag single nucleotide polymorphisms (SNPs) located within the ANKA gene were selected for genotyping. Single marker-based association analyses were conducted using multiple modes of inheritance, including genotypic, allelic, dominant, and recessive. Haplotype-based association analyses were also performed. Plink was utilized for genetic association analyses. In addition, we examined the GTEx database to test the expression quantitative loci effects of the significant SNPs within the AKNA gene. Results: Among these eight SNPs evaluated we identified one, rs10817595, as being significantly associated with the risk of KOA. Compared to the CC genotype at this locus, the odds ratio (95% confidence interval) for KOA with the AA genotype was 1.58 (1.23-2.01)-fold greater. A linkage disequilibrium block that included this SNP was also determined to be significantly associated with the risk of KOA (χ2 = 25.08, p = 3.58 × 10-6). In general, the minor allele A of SNP rs10817595 was associated with an increased risk of KOA. Conclusion: This study is the first to present evidence for a potential link between the risk of KOA and an AKNA gene polymorphism among persons with a Han Chinese ancestry. Future functional analyses based on animal models and sequencing-based population studies are needed to elucidate the biological plausibility and genetic architecture of AKNA for KOA susceptibility.
Collapse
Affiliation(s)
- Tianyun Zhao
- Department of Orthopedics and The First Hospital of Tianshui City, Tianshui, China.,Department of Sports Medicine, The First Hospital of Tianshui City, Tianshui, China
| | - Chi Ma
- Department of Orthopedics and The First Hospital of Tianshui City, Tianshui, China
| | - Baopin Xie
- Department of Sports Medicine, The First Hospital of Tianshui City, Tianshui, China
| | - Bin Zhao
- Department of Sports Medicine, The First Hospital of Tianshui City, Tianshui, China
| | - Wei Wang
- Department of Sports Medicine, The First Hospital of Tianshui City, Tianshui, China
| | - Jibin Liu
- Department of Oncology Research, The Affiliated Oncology Hospital of Nantong University, Nantong, China
| |
Collapse
|
432
|
Bellini M, Pest MA, Miranda-Rodrigues M, Qin L, Jeong JW, Beier F. Overexpression of MIG-6 in the cartilage induces an osteoarthritis-like phenotype in mice. Arthritis Res Ther 2020; 22:119. [PMID: 32430054 PMCID: PMC7236969 DOI: 10.1186/s13075-020-02213-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most common form of arthritis and characterized by degeneration of the articular cartilage. Mitogen-inducible gene 6 (Mig-6) has been identified as a negative regulator of the epidermal growth factor receptor (EGFR). Cartilage-specific Mig-6 knockout (KO) mice display increased EGFR signaling, an anabolic buildup of the articular cartilage, and formation of chondro-osseous nodules. Since our understanding of the EGFR/Mig-6 network in the cartilage remains incomplete, we characterized mice with cartilage-specific overexpression of Mig-6 in this study. METHODS Utilizing knee joints from cartilage-specific Mig-6-overexpressing (Mig-6over/over) mice (at multiple time points), we evaluated the articular cartilage using histology, immunohistochemical staining, and semi-quantitative histopathological scoring (OARSI) at multiple ages. MicroCT analysis was employed to examine skeletal morphometry, body composition, and bone mineral density. RESULTS Our data show that cartilage-specific Mig-6 overexpression did not cause any major developmental abnormalities in the articular cartilage, although Mig-6over/over mice have slightly shorter long bones compared to the control group. Moreover, there was no significant difference in bone mineral density and body composition in any of the groups. However, our results indicate that Mig-6over/over male mice show accelerated cartilage degeneration at 12 and 18 months of age. Immunohistochemistry for SOX9 demonstrated that the number of positively stained cells in Mig-6over/over mice was decreased relative to controls. Immunostaining for MMP13 appeared increased in areas of cartilage degeneration in Mig-6over/over mice. Moreover, staining for phospho-EGFR (Tyr-1173) and lubricin (PRG4) was decreased in the articular cartilage of Mig-6over/over mice. CONCLUSION Overexpression of Mig-6 in the articular cartilage causes no major developmental phenotype; however, these mice develop earlier OA during aging. These data demonstrate that Mig-6/EGFR pathways are critical for joint homeostasis and might present a promising therapeutic target for OA.
Collapse
Affiliation(s)
- Melina Bellini
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Western University Bone and Joint Institute, London, ON, Canada
| | - Michael A Pest
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Western University Bone and Joint Institute, London, ON, Canada
| | - Manuela Miranda-Rodrigues
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
- Western University Bone and Joint Institute, London, ON, Canada
- Children's Health Research Institute, London, ON, Canada
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Frank Beier
- Department of Physiology and Pharmacology, Western University, London, ON, Canada.
- Western University Bone and Joint Institute, London, ON, Canada.
- Children's Health Research Institute, London, ON, Canada.
| |
Collapse
|
433
|
Gun Bilgic D, Hatipoglu OF, Cigdem S, Bilgic A, Cora T. NF-ĸβ upregulates ADAMTS5 expression by direct binding after TNF-α treatment in OUMS-27 chondrosarcoma cell line. Mol Biol Rep 2020; 47:4215-4223. [PMID: 32415504 DOI: 10.1007/s11033-020-05514-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
Inflammation caused-aggrecan degradation is a critical event in the pathogenesis of osteoarthritis (OA). The aggrecanases like a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) are assumed to be key players in the aggrecan destruction. To develop the comprehensive therapy method for OA, it is essential to elucidate the activation mechanism of ADAMTS5 gene after stimulation of inflammatory cytokines like tumor necrosis factor-α (TNF-α). The cell lines of human chondrosarcoma (OUMS-27) and embryonic kidney (HEK293T) were incubated with tumor necrosis factor-α (TNF-α) for certain time periods, and the expression level of ADAMTS5 was measured in both mRNA and protein levels. Tissue-specific ADAMTS5 activation was founded to be induced after TNF-α treatment. Then, the constructs for the promoter region of ADAMTS5 were prepared and luciferase assay was conducted to understand the involvement mechanism of nuclear factor-kappa beta (NF-ĸβ) in ADAMTS5 activation. It was demonstrated that NF-ĸβ induces the ADAMTS5 expression level by directly binding the promoter region of ADAMTS5. Although the TNF-α blocker is used for OA treatment, the development of a more comprehensive treatment strategy is an urgent need. Our experimental data contributes in terms of selecting NF-ĸβ as a target molecule. Up to date, NF-ĸβ has been proven to involve in the ADAMTS5 up-regulation after several pro-inflammatory cytokines stimulation. In conclusion, our findings make important contributions to the knowledge about the roles of NF-ĸβ in ADAMTS5 activation under inflammatory conditions. So, NF-ĸβ could be considered to be a potential target for OA treatment.
Collapse
Affiliation(s)
- Dilek Gun Bilgic
- Department of Medical Genetics, Manisa Celal Bayar University Medical Faculty, Manisa, Turkey.
| | - Omer Faruk Hatipoglu
- Department of Pharmacology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Sadık Cigdem
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Abdulkadir Bilgic
- Department of Orthopedics and Traumatology, Manisa City Hospital, Manisa, Turkey
| | - Tulin Cora
- Department of Medical Genetics, Selcuk University Medical Faculty, Konya, Turkey
| |
Collapse
|
434
|
Hamasaki M, Terkawi MA, Onodera T, Tian Y, Ebata T, Matsumae G, Alhasan H, Takahashi D, Iwasaki N. Transcriptional profiling of murine macrophages stimulated with cartilage fragments revealed a strategy for treatment of progressive osteoarthritis. Sci Rep 2020; 10:7558. [PMID: 32371954 PMCID: PMC7200748 DOI: 10.1038/s41598-020-64515-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/14/2020] [Indexed: 01/15/2023] Open
Abstract
Accumulating evidence suggests that synovitis is associated with osteoarthritic process. Macrophages play principal role in development of synovitis. Our earlier study suggests that interaction between cartilage fragments and macrophages exacerbates osteoarthritic process. However, molecular mechanisms by which cartilage fragments trigger cellular responses remain to be investigated. Therefore, the current study aims at analyzing molecular response of macrophages to cartilage fragments. To this end, we analyzed the transcriptional profiling of murine macrophages exposed to cartilage fragments by RNA sequencing. A total 153 genes were differentially upregulated, and 105 genes were down-regulated in response to cartilage fragments. Bioinformatic analysis revealed that the most significantly enriched terms of the upregulated genes included scavenger receptor activity, integrin binding activity, TNF signaling, and toll-like receptor signaling. To further confirm our results, immunohistochemical staining was performed to detected regulated molecules in synovial tissues of OA patients. In consistence with RNA-seq results, MARCO, TLR2 and ITGα5 were mainly detected in the intima lining layer of synovial tissues. Moreover, blockade of TLR2 or ITGα5 but not Marco using specific antibody significantly reduced production of TNF-α in stimulated macrophages by cartilage fragments. Our data suggested that blocking TLR2 or ITGα5 might be promising therapeutic strategy for treating progressive osteoarthritis.
Collapse
Affiliation(s)
- Masanari Hamasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Mohamad Alaa Terkawi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan.
- Global Institution for Collaborative Research and Education (GI-CoRE), Frontier Research Center for Advanced Material and Life Science Bldg No 2, Hokkaido University, Sapporo, Japan.
| | - Tomohiro Onodera
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan.
- Global Institution for Collaborative Research and Education (GI-CoRE), Frontier Research Center for Advanced Material and Life Science Bldg No 2, Hokkaido University, Sapporo, Japan.
| | - Yuan Tian
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Taku Ebata
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Gen Matsumae
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Hend Alhasan
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Daisuke Takahashi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita-15, Nish-7, Kita-ku, Sapporo, 060-8638, Japan
- Global Institution for Collaborative Research and Education (GI-CoRE), Frontier Research Center for Advanced Material and Life Science Bldg No 2, Hokkaido University, Sapporo, Japan
| |
Collapse
|
435
|
Dawood AF, Alzamil N, Ebrahim HA, Abdel Kader DH, Kamar SS, Haidara MA, Al-Ani B. Metformin pretreatment suppresses alterations to the articular cartilage ultrastructure and knee joint tissue damage secondary to type 2 diabetes mellitus in rats. Ultrastruct Pathol 2020; 44:273-282. [PMID: 32404018 DOI: 10.1080/01913123.2020.1762815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) secondary to diabetes affects millions of people worldwide and can lead to disability. The protective effect of metformin pretreatment against alterations to the articular cartilage ultrastructure induced by type 2 diabetes mellitus (T2DM) associated with the inhibition of oxidative stress and inflammation has not been investigated before. Therefore, we induced T2DM in rats (the model group) using high carbohydrate and fat diet and a single injection of streptozotocin (50 mg/kg body weight). The protective group of rats started metformin (200 mg/kg body weight) treatment 14 days before diabetic induction and continued on metformin until the end of the experiment at week 12. Harvested tissues obtained from knee joints were prepared for staining with hematoxylin and eosin (H&E), safranin o staining, and electron microscopy. Histology images showed that OA was developed in the T2DM rats as demonstrated by a substantial damage to the articular cartilage and profound chondrocyte and territorial matrix ultrastructural alterations, which were partially protected by metformin. In addition, metformin significantly (p < .05) reduced hyperglycemia, glycated hemoglobin (HbA1 c), malondialdehyde (MDA), high sensitivity C-reactive protein (hs-CRP), and interleukin-6 blood levels induced by diabetes. Furthermore, a significant (p ≤ 0.015) correlation between either OA cartilage grade score or the thickness of the articular cartilage and the blood levels of HbA1 c, hs-CRP, MDA, superoxide dismutase (SOD) were observed. These findings demonstrate effective protection of the articular cartilage by metformin against damage induced secondary to T2DM in rats, possibly due to the inhibition of hyperglycemia and biomarkers of oxidative stress and inflammation.
Collapse
Affiliation(s)
- Amal F Dawood
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University , Riyadh, Saudi Arabia
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University , Cairo, Egypt
| | - Norah Alzamil
- Department of Clinical Science, Family Medicine, College of Medicine, Princess Nourah Bint Abdulrahman University , Riyadh, Saudi Arabia
| | - Hasnaa A Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University , Riyadh, Saudi Arabia
- Department of Anatomy, College of Medicine, Mansoura University , Mansoura, Egypt
| | - Dina H Abdel Kader
- Department of Medical Histology, Kasr Al-Aini Faculty of Medicine, Cairo University , Cairo, Egypt
| | - Samaa S Kamar
- Department of Medical Histology, Kasr Al-Aini Faculty of Medicine, Cairo University , Cairo, Egypt
| | - Mohamed A Haidara
- Department of Physiology, Kasr Al-Aini Faculty of Medicine, Cairo University , Cairo, Egypt
- Department of Physiology, College of Medicine, King Khalid University , Abha, Saudi Arabia
| | - Bahjat Al-Ani
- Department of Physiology, College of Medicine, King Khalid University , Abha, Saudi Arabia
| |
Collapse
|
436
|
Sharma N, Drobinski P, Kayed A, Chen Z, Kjelgaard-Petersen CF, Gantzel T, Karsdal MA, Michaelis M, Ladel C, Bay-Jensen AC, Lindemann S, Thudium CS. Inflammation and joint destruction may be linked to the generation of cartilage metabolites of ADAMTS-5 through activation of toll-like receptors. Osteoarthritis Cartilage 2020; 28:658-668. [PMID: 31734268 DOI: 10.1016/j.joca.2019.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/30/2019] [Accepted: 11/04/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Links between pain and joint degradation are poorly understood. We investigated the role of activation of Toll-like receptors (TLR) by cartilage metabolites in initiating and maintaining the inflammatory loop in OA causing joint destruction. METHODS Synovial membrane explants (SMEs) were prepared from OA patients' synovial biopsies. SMEs were cultured for 10 days under following conditions: culture medium alone, OSM + TNFα, TLR2 agonist - Pam2CSK4, Pam3CSK4 or synthetic aggrecan 32-mer, TLR4 agonist - Lipid A. Release of pro-inflammatory and degradation biomarkers (acMMP3 and C3M) were measured by ELISA in conditioned media along with IL-6. Additionally, human cartilage was digested with ADAMTS-5, with or without the ADAMTS-5 inhibiting nanobody - M6495. Digested cartilage solution (DCS) and synthetic 32-mer were tested for TLR activation in SEAP based TLR reporter assay. RESULTS Western blotting confirmed TLR2 and TLR4 in untreated OA synovial biopsies. TLR agonists showed an increase in release of biomarkers - acMMP3 and C3M in SME. Synthetic 32-mer showed no activation in the TLR reporter assay. ADAMTS-5 degraded cartilage fragments activated TLR2 in vitro. Adding M6495 - an anti-ADAMTS-5 inhibiting nanobody®, blocked ADAMTS-5-mediated DCS TLR2 activation. CONCLUSION TLR2 is expressed in synovium of OA patients and their activation by synthetic ligands causes increased tissue turnover. ADAMTS-5-mediated cartilage degradation leads to release of aggrecan fragments which activates the TLR2 receptor in vitro. M6495 suppressed cartilage degradation by ADAMTS-5, limiting the activation of TLR2. In conclusion, pain and joint destruction may be linked to generation of ADAMTS-5 cartilage metabolites.
Collapse
Affiliation(s)
- N Sharma
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.
| | - P Drobinski
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.
| | - A Kayed
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.
| | - Z Chen
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark; Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark.
| | | | - T Gantzel
- Orthopaedic Surgery Unit, Gentofte University Hospital, Gentofte, Denmark.
| | - M A Karsdal
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark.
| | - M Michaelis
- Head of Osteoarthritis Research and Early Clinical Development, Merck KGaA, Darmstadt, Germany.
| | - C Ladel
- Clinical Biomarker & Diagnostics Lead, Merck KGaA, Darmstadt, Germany.
| | - A C Bay-Jensen
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark.
| | - S Lindemann
- Head of Exploratory Osteoarthritis, Merck KGaA, Darmstadt, Germany.
| | - C S Thudium
- Rheumatology, Nordic Bioscience, Herlev Hovedgade 207, DK-2730, Herlev, Denmark.
| |
Collapse
|
437
|
Miller RJ, Malfait AM, Miller RE. The innate immune response as a mediator of osteoarthritis pain. Osteoarthritis Cartilage 2020; 28:562-571. [PMID: 31862470 PMCID: PMC6951330 DOI: 10.1016/j.joca.2019.11.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 11/20/2019] [Indexed: 02/08/2023]
Abstract
In this narrative review, we discuss the emerging role of innate immunity in osteoarthritis (OA) joint pain. First, we give a brief description of the pain pathway in the context of OA. Then we consider how neuro-immune signaling pathways may promote OA pain. First, activation of neuronal Pattern Recognition Receptors by mediators released in a damaged joint can result in direct excitation of nociceptors, as well as in production of chemokines and cytokines. Secondly, indirect neuro-immune signaling may occur when innate immune cells produce algogenic factors, including chemokines and cytokines, that act on the pain pathway. Neuro-immune crosstalk occurs at different levels of the pathway, starting in the joint but also in the innervating dorsal root ganglia and in the dorsal horn. Synovitis is characterized by recruitment of immune cells, including macrophages, mast cells, and CD4+ lymphocytes, which may contribute to nociceptor sensitization and OA pain through production of algogenic factors that amplify the activation of sensory neurons. We discuss examples where this scenario has been suggested by findings in human OA and in animal models. Overall, increasing evidence suggests that innate immune pathways play an initiating as well as facilitating role in pain, but information on how these pathways operate in OA remains limited. Since these innate pathways are eminently targetable, future studies in this area may provide fruitful leads towards a better management of symptomatic OA.
Collapse
|
438
|
Zhang H, Cai D, Bai X. Macrophages regulate the progression of osteoarthritis. Osteoarthritis Cartilage 2020; 28:555-561. [PMID: 31982565 DOI: 10.1016/j.joca.2020.01.007] [Citation(s) in RCA: 313] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 01/06/2020] [Accepted: 01/10/2020] [Indexed: 02/02/2023]
Abstract
OA is now well accepted as a low-grade inflammatory disease affecting the whole joint. In addition to mechanical loading, inflammation (particularly synovitis), contributes significantly to OA. Synovial macrophages act as immune cells and are of critical importance in the symptomology and structural progression of OA. Activated macrophages are regulated by mTOR, NF-κB, JNK, PI3K/Akt and other signaling pathways, and are polarized into either M1 or M2 subtypes in OA synovial tissues, synovial fluid, and peripheral blood. The activation state and the M1/M2 ratio is highly associated with OA severity. Aside from autocrine interactions, paracrine interactions between macrophages and chondrocytes play a vital role in the initiation and development of OA by secreting inflammatory cytokines, growth factors, matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs), which lead to subsequent cartilage degradation and destruction. Treatments targeting synovial macrophages relieve pain, and protect from synovitis, cartilage damage, and osteophyte formation during OA development. Macrophage reprogramming of transformation from the M1 to M2 subtype, more than a decrease in the quantity of activated macrophages, appears to be an effective treatment option for OA. This review provides a broad understanding of the contributions of polarized macrophages to joint health and disease. Multifunctional agents with immunomodulatory effects on macrophage reprogramming can skew the inflammatory microenvironment towards a pro-chondrogenic atmosphere, and are thus, potential therapeutic options for the treatment of OA and other immune diseases.
Collapse
Affiliation(s)
- H Zhang
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510280, China.
| | - D Cai
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510280, China.
| | - X Bai
- Department of Orthopedics, Orthopedic Hospital of Guangdong Province, Academy of Orthopedics·Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510280, China; Key Laboratory of Mental Health of the Ministry of Education, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005, Guangzhou, China.
| |
Collapse
|
439
|
Kimmerling KA, Gomoll AH, Farr J, Mowry KC. Amniotic Suspension Allograft Modulates Inflammation in a Rat Pain Model of Osteoarthritis. J Orthop Res 2020; 38:1141-1149. [PMID: 31814175 PMCID: PMC7187262 DOI: 10.1002/jor.24559] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 12/04/2019] [Indexed: 02/04/2023]
Abstract
Osteoarthritis (OA) affects over 301 million adults worldwide. Inflammation is a recognized component of the OA process; two potent pro-inflammatory cytokines involved in OA are interleukin-1β and tumor necrosis factor-α. Placental-derived tissues and fluids are known to contain anti-inflammatory and immunomodulatory cytokines and growth factors. The objective of this study was to evaluate the anti-inflammatory effects of amniotic suspension allograft (ASA) in an in vivo model of OA; we evaluated pain, function, and cytokine levels following ASA treatment in the rat monosodium iodoacetate (MIA) OA pain model. Rats were injected with 2 mg of MIA, which causes pain, cartilage degeneration, and inflammation, followed by treatment with saline, triamcinolone (positive control), or ASA 7 days following disease induction with MIA. Behavioral assays, including gait analysis, mechanical pain threshold, incapacitance, and swelling were evaluated, along with histology and serum and synovial fluid biomarkers. Treatment with ASA resulted in significant improvements in pain threshold, while weight bearing aversion and swelling were significantly decreased. There were no differences between groups in total joint score after histological grading. Serum biomarkers did not show differences, indicating a lack of systemic response; however, synovial fluid levels of IL-10 were significantly increased in animals treated with ASA. ASA treatment significantly reduced pain, weight-bearing aversion and swelling. This study provides mechanistic data regarding potential therapeutic effects of ASA in OA and preliminary evidence of the anti-inflammatory nature of ASA. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:1141-1149, 2020.
Collapse
Affiliation(s)
| | - Andreas H. Gomoll
- Department of Orthopaedic SurgeryHospital for Special SurgeryNew YorkNew York
| | - Jack Farr
- Knee Preservation and Cartilage Restoration Center, OrthoIndyIndianapolisIndiana
| | | |
Collapse
|
440
|
Sanpaolo ER, Corrado A, Cantatore FP. Mediterranean Diet and Osteoarticular Diseases. CURRENT NUTRITION & FOOD SCIENCE 2020. [DOI: 10.2174/1386207322666191203103530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Besides the well-known positive effect on the reduction of the risk of cardiovascular
diseases, in the latest decades growing evidence has accumulated on the beneficial effects
of MD on various aspects of health outcomes. Nevertheless, data concerning the existence of a direct
positive effect of MD, irrespective of its beneficial effect on body weight, in osteo-articular diseases,
are still controversial. In osteo-articular diseases, a pro-inflammatory environment is highlighted,
with an increased systemic levels of pro-inflammatory cytokines.
Objective:
Our objective is to investigate the effects of adherence to the Mediterranean diet and
osteo-articular diseases.
Conclusion:
Many foods included in MD, have anti-inflammatory properties, due to the presence of
nutrients, such as polyunsaturated (PUFA) and monounsaturated (MUFA) fats. The two types of polyunsaturated
fatty acids, omega-3 and omega-6, have opposing effects on the inflammatory process.
Omega-6 stimulates the production of pro-inflammatory cytokines, while omega-3 fatty acids exert
anti-inflammatory effects, including significant reductions in the release of pro-inflammatory cytokines.
Some studies have shown that the dietary pattern of MD consumption has an important role in
the prevention and development of inflammatory arthritis. Nevertheless, the possible influence of
MD on chronic osteo-articular diseases is very limited, this review updates the main experimental
and clinical evidences concerning the potential beneficial effects of the Mediterranean Diet on the
most diffuse osteo-articular chronic and degenerative joint disease.
Collapse
Affiliation(s)
- Eliana R. Sanpaolo
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Addolorata Corrado
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Francesco P. Cantatore
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
441
|
Tong W, Zhang X, Zhang Q, Fang J, Liu Y, Shao Z, Yang S, Wu D, Sheng X, Zhang Y, Tian H. Multiple umbilical cord-derived MSCs administrations attenuate rat osteoarthritis progression via preserving articular cartilage superficial layer cells and inhibiting synovitis. J Orthop Translat 2020; 23:21-28. [PMID: 32455113 PMCID: PMC7235619 DOI: 10.1016/j.jot.2020.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/26/2020] [Accepted: 03/11/2020] [Indexed: 01/08/2023] Open
Abstract
Background/objectives Articular cartilage erosion probably plays a substantial role in osteoarthritis (OA) initiation and development. Studies demonstrated that umbilical cord–derived mesenchymal stem cells (UCMSCs) could delay chondrocytes apoptosis and ameliorate OA progression in patients, but the detailed mechanisms are largely uncharacterised. In this study, we aimed to study the effects of UCMSCs on monosodium iodoacetate (MIA)–induced rat OA model, and explore the cellular mechanism of this effect. Methods Intra-articular injection of 0.3 mg MIA in 50 μL saline was performed on the left knee of the 200 g weight male Sprague-Dawley rat to induce rat knee OA. A single dose of 2.5 × 105 undifferentiated UCMSCs one day after MIA or three-time intra-articular injection of 2.5 × 105 UCMSCs on Days 1, 7 and 14 were given, respectively. Four weeks after MIA, joints were harvested and processed for paraffin sections. Safranine-O staining, haematoxylin and eosin staining and immunohistochemistry of MMP-13, ADAMTS-5, Col-2, CD68 and CD4 were performed to observe cartilage erosion and synovium. For in vitro studies, migration ability of cartilage superficial layer cells (SFCs) by UCMSCs were accessed by transwell assay. Furthermore, catabolism change of MIA-induced SFCs by UCMSCs was performed by real-rime polymerase chain reaction of Col-X and BCL-2 genes. CCK-8 assay was performed to check proliferation ability of SFCs by UCMSCs-conditioned media. Result In this study, we locally injected human UCMSCs, which is highly proliferative and noninvasively collectible, into MIA-induced rat knee OA. An important finding is on obviously ameliorated cartilage erosion and decreased OA Mankin score by repeated UCMSCs injection after MIA injection compared with single injection, both of which attenuated OA progression compared with vehicle. Interestingly, we observed significantly increased number of SFCs on the articular cartilage surface, probably related to elevated proliferation, mobilisation and inhibited catabolism marker: Col-X and BCL-2 gene expression of cultured SFCs by UCMSCs-conditioned media treatment in vitro. In addition to the change of unique SFCs, catabolism markers of ADAMTS-5 and MMP-13 were substantially upregulated in the whole cartilage layer chondrocytes as well. Strikingly, MIA-induced inflammatory cells infiltration, on both CD4+ Th cells and CD68+ macrophages, and hyperplasia of the synovium, which was alleviated by repeated UCMSCs injection. Conclusion Our study demonstrated a critical role of repeated UCMSCs dosing on preserving SFCs function, cartilage structure and inhibiting synovitis during OA progression, and thus provided mechanistic proof of evidence for the use of UCMSCs on OA patients in the future. The translational potential of this article UCMSCs are a relatively “young” stem cell, and noninvasively collectible. In our study, we clearly demonstrated that it could effectively delay OA progression, possibly through reserving SFCs function and inhibiting synovitis. Therefore, it could be a new promising therapeutic cell source for OA after further clinical trials.
Collapse
Affiliation(s)
- Wei Tong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Xiaoguang Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Quan Zhang
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, 430075, Hubei, China
| | - Jiarui Fang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yong Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Shuhua Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Dongcheng Wu
- Wuhan Hamilton Biotechnology Co., Ltd., Wuhan, 430075, Hubei, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430072, Hubei, China
| | - Xiaoming Sheng
- Applied Statistics, University of Utah College of Nursing, 10 South 2000 East, Salt Lake City, UT, 84112, USA
| | - Yingze Zhang
- The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Hongtao Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Corresponding author. Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
| |
Collapse
|
442
|
Effects of Balneotherapy in Jeju Magma-Seawater on Knee Osteoarthritis Model. Sci Rep 2020; 10:6620. [PMID: 32313003 PMCID: PMC7171195 DOI: 10.1038/s41598-020-62867-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/10/2020] [Indexed: 12/14/2022] Open
Abstract
Balneotherapy is a common non-pharmacological treatment for osteoarthritis (OA), however, the efficacy is controversial in knee OA. Jeju magma-seawater (JMS) has high contents of various minerals, which has anti-inflammatory and antioxidant properties via an oral route. Thus, we examined the effects of JMS bathing on knee OA and the combination effects with diclofenac sodium as an anti-inflammatory drug. Knee OA was induced by transection of the anterior cruciate ligament and the partial meniscectomy in rat. The rats were administered subcutaneously saline or diclofenac sodium in saline, followed by bathing in thermal distilled water or JMS for 8 weeks. The model represented the characteristic changes of the cartilage degradation, osteophyte formation and synovial inflammation, and the relevant symptoms of the joint swelling and stiffness. However, the JMS bathing reduced the joint thickness and improved the mobility. It also contributed to a well-preserved tissue supported by increases in bone mineral density of the joint and decreases in Mankin scores in the cartilages. The effects involved anti-inflammation, chondroprotection, anti-apoptosis, and chondrogenesis. Overall, the JMS bathing in combination with diclofenac sodium showed a similar trend associated with synergic effects. It suggests that JMS bathing can be promising for a clinical use in knee OA.
Collapse
|
443
|
Duan X, Cai L, Schmidt EJ, Shen J, Tycksen ED, O’Keefe R, Cheverud JM, Farooq Rai M. RNA-seq analysis of chondrocyte transcriptome reveals genetic heterogeneity in LG/J and SM/J murine strains. Osteoarthritis Cartilage 2020; 28:516-527. [PMID: 31945456 PMCID: PMC7108965 DOI: 10.1016/j.joca.2020.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the transcriptomic differences in chondrocytes obtained from LG/J (large, healer) and SM/J (small, non-healer) murine strains in an attempt to discern the molecular pathways implicated in cartilage regeneration and susceptibility to osteoarthritis (OA). DESIGN We performed RNA-sequencing on chondrocytes derived from LG/J (n = 16) and SM/J (n = 16) mice. We validated the expression of candidate genes and compared single nucleotide polymorphisms (SNPs) between the two mouse strains. We also examined gene expression of positional candidates for ear pinna regeneration and long bone length quantitative trait loci (QTLs) that display differences in cartilaginous expression. RESULTS We observed a distinct genetic heterogeneity between cells derived from LG/J and SM/J mouse strains. We found that gene ontologies representing cell development, cartilage condensation, and regulation of cell differentiation were enriched in LG/J chondrocytes. In contrast, gene ontologies enriched in the SM/J chondrocytes were mainly related to inflammation and degeneration. Moreover, SNP analysis revealed that multiple validated genes vary in sequence between LG/J and SM/J in coding and highly conserved noncoding regions. Finally, we showed that most QTLs have 20-30% of their positional candidates displaying differential expression between the two mouse strains. CONCLUSIONS While the enrichment of pathways related to cell differentiation, cartilage development and cartilage condensation infers superior healing potential of LG/J strain, the enrichment of pathways related to cytokine production, immune cell activation and inflammation entails greater susceptibility of SM/J strain to OA. These data provide novel insights into chondrocyte transcriptome and aid in identification of the quantitative trait genes and molecular differences underlying the phenotypic differences associated with individual QTLs.
Collapse
Affiliation(s)
- Xin Duan
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Lei Cai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Eric J. Schmidt
- School of Physician Assistant Medicine, College of Health Sciences, University of Lynchburg, Lynchburg, VA, United States
| | - Jie Shen
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - Eric D. Tycksen
- Genome Technology Access Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Regis O’Keefe
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States
| | - James M. Cheverud
- Department of Biology, Loyola University, Chicago, IL, United States
| | - Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, United States, Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
444
|
Total Knee Arthroplasty with a Ti6Al4V/PEEK Prosthesis on an Osteoarthritis Rat Model: Behavioral and Neurophysiological Analysis. Sci Rep 2020; 10:5277. [PMID: 32210280 PMCID: PMC7093407 DOI: 10.1038/s41598-020-62146-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 03/02/2020] [Indexed: 11/08/2022] Open
Abstract
Arthroplasty is a surgical procedure to restore the function of the joint of patient suffering from knee osteoarthritis. However, postoperative functional deficits are reported even after a rehabilitation program. In order to determine the origin of functional deficits of patient suffering from knee osteoarthritis and total knee arthroplasty, we developed a rodent model including a chemically-induced-osteoarthritis and designed a knee prosthesis (Ti6Al4V/PEEK) biomechanically and anatomically adapted to rat knee joint. Dynamic Weight-Bearing, gait kinematics, H-reflex from vastus medialis muscle and activities from metabosensitive III and IV afferent fibers in femoral nerve were assessed at 1 and 3 months post-surgery. Results indicate that knee osteoarthritis altered considerably the responses of afferent fibers to their known activators (i.e., lactic acid and potassium chloride) and consequently their ability to modulate the spinal sensorimotor loop, although, paradoxically, motor deficits seemed relatively light. On the contrary, results indicate that, after the total knee arthroplasty, the afferent responses and the sensorimotor function were slightly altered but that motor deficits were more severe. We conclude that neural changes attested by the recovery of the metabosensitive afferent activity and the sensorimotor loop were induced when a total knee replacement was performed and that these changes may disrupt or delay the locomotor recovery.
Collapse
|
445
|
The Role of Inflammation in the Pathogenesis of Osteoarthritis. Mediators Inflamm 2020; 2020:8293921. [PMID: 32189997 PMCID: PMC7072120 DOI: 10.1155/2020/8293921] [Citation(s) in RCA: 294] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 01/15/2023] Open
Abstract
A joint is the point of connection between two bones in our body. Inflammation of the joint leads to several diseases, including osteoarthritis, which is the concern of this review. Osteoarthritis is a common chronic debilitating joint disease mainly affecting the elderly. Several studies showed that inflammation triggered by factors like biomechanical stress is involved in the development of osteoarthritis. This stimulates the release of early-stage inflammatory cytokines like interleukin-1 beta (IL-1β), which in turn induces the activation of signaling pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphoinositide 3-kinase/protein kinase B (PI3K/AKT), and mitogen-activated protein kinase (MAPK). These events, in turn, generate more inflammatory molecules. Subsequently, collagenase like matrix metalloproteinases-13 (MMP-13) will degrade the extracellular matrix. As a result, anatomical and physiological functions of the joint are altered. This review is aimed at summarizing the previous studies highlighting the involvement of inflammation in the pathogenesis of osteoarthritis.
Collapse
|
446
|
Ter Heegde F, Luiz AP, Santana-Varela S, Magnúsdóttir R, Hopkinson M, Chang Y, Poulet B, Fowkes RC, Wood JN, Chenu C. Osteoarthritis-related nociceptive behaviour following mechanical joint loading correlates with cartilage damage. Osteoarthritis Cartilage 2020; 28:383-395. [PMID: 31911151 DOI: 10.1016/j.joca.2019.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/18/2019] [Accepted: 12/22/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In osteoarthritis (OA), the pain-structure relationship remains complex and poorly understood. Here, we used the mechanical joint loading (MJL) model of OA to investigate both knee pathology and nociceptive behaviour. DESIGN MJL was used to induce OA in the right knees of 12-week-old male C57BL/6 mice (40 cycles, 9N, 3x/week for 2 weeks). Mechanical sensitivity thresholds and weight-bearing ratios were measured before loading and at weeks one, three and six post-loading. At these time points, separate groups of loaded and non-loaded mice (n = 12/group) were sacrificed, joints collected, and fur corticosterone levels measured. μCT analyses of subchondral bone integrity was performed before joint sections were prepared for nerve quantification, cartilage or synovium grading (scoring system from 0 to 6). RESULTS Loaded mice showed increased mechanical hypersensitivity paired with altered weight-bearing. Initial ipsilateral cartilage lesions 1-week post-loading (1.8 ± 0.4) had worsened at weeks three (3.0 ± 0.6, CI = -1.8-0.6) and six (2.8 ± 0.4, CI = -1.6-0.4). This increase in lesion severity correlated with mechanical hypersensitivity development (correlation; 0.729, P = 0.0071). Loaded mice displayed increased synovitis (3.6 ± 0.5) compared to non-loaded mice (1.5 ± 0.5, CI = -2.2-0.3) 1-week post-loading which returned to normal by weeks three and six. Similarly, corticosterone levels were only increased at week one post-loading (0.21 ± 0.04 ng/mg) compared to non-loaded controls (0.14 ± 0.01 ng/mg, CI = -1.8-0.1). Subchondral bone integrity and nerve volume remained unchanged. CONCLUSIONS Our data indicates that although the loading induces an initial stress reaction and local inflammation, these processes are not directly responsible for the nociceptive phenotype observed. Instead, MJL-induced allodynia is mainly associated with OA-like progression of cartilage lesions.
Collapse
Affiliation(s)
- F Ter Heegde
- Skeletal Biology Group, Comparative Biomedical Science, Royal Veterinary College, London NW1 0TU, UK; Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK.
| | - A P Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK.
| | - S Santana-Varela
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK.
| | - R Magnúsdóttir
- Skeletal Biology Group, Comparative Biomedical Science, Royal Veterinary College, London NW1 0TU, UK.
| | - M Hopkinson
- Skeletal Biology Group, Comparative Biomedical Science, Royal Veterinary College, London NW1 0TU, UK.
| | - Y Chang
- Research Office, Royal Veterinary College, London NW1 0TU, UK.
| | - B Poulet
- Muscoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool L69 3BX, UK.
| | - R C Fowkes
- Endocrine Signalling Group, Comparative Biomedical Science, Royal Veterinary College, London NW1 0TU, UK.
| | - J N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, London WC1E 6BT, UK.
| | - C Chenu
- Skeletal Biology Group, Comparative Biomedical Science, Royal Veterinary College, London NW1 0TU, UK.
| |
Collapse
|
447
|
Luo Z, Hu Z, Bian Y, Su W, Li X, Li S, Wu J, Shi L, Song Y, Zheng G, Ni W, Xue J. Scutellarin Attenuates the IL-1β-Induced Inflammation in Mouse Chondrocytes and Prevents Osteoarthritic Progression. Front Pharmacol 2020; 11:107. [PMID: 32161544 PMCID: PMC7054241 DOI: 10.3389/fphar.2020.00107] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/28/2020] [Indexed: 12/24/2022] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease wherein the articular cartilage exhibits inflammation and degradation. Scutellarin (SCU) is a flavonoid glycoside with a range of pharmacological activities, as shown in previous studies demonstrating its anti-inflammatory activity. How SCU impacts the progression of OA, however, has not been explored to date. Herein, we assessed the impact of SCU on murine chondrocytes in an OA model system. In in vitro assays, we measured chondrocyte expression of key OA-associated factors such as matrix metalloproteinase 13 (MMP-13), a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS-5), cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) via qRT-PCR and Western blotting, the expression of interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), and prostaglandin E2 (PGE2) were detected by qRT-PCR. Our results showed that the downregulation of MMP-13, ADAMTS-5, COX-2, and iNOS expression by SCU and the overproduction of IL-6, TNF-α, and PGE2 induced by IL-1β were all inhibited by SCU in a concentration-dependent manner. Moreover, SCU was able to reverse aggrecan and collagen II degradation and nuclear factor-κB (NF-κB) and nuclear factor erythroid-derived 2-like 2 (Nrf2) signaling pathway activation both in vivo and in vitro. We further used a destabilization of the medial meniscus (DMM) murine model of OA to explore the therapeutic benefits of SCU in vivo. Together, our findings suggest SCU to be a potentially valuable therapeutic agent useful for treating OA.
Collapse
Affiliation(s)
- Zucheng Luo
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zhichao Hu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yujie Bian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Wenting Su
- Department of Dermatology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, China
| | - Xiaoyang Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Shi Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Jianbin Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Li Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Yonghuan Song
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China
| | - Gang Zheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Wenfei Ni
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jixin Xue
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopaedics, Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
448
|
Cui L, Han Y, Dong Z. miR-106a mimics the nuclear factor-κB signalling pathway by targeting DR6 in rats with osteoarthritis. Arch Med Sci 2020; 20:302-308. [PMID: 38414449 PMCID: PMC10895953 DOI: 10.5114/aoms.2020.92831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/04/2019] [Indexed: 02/29/2024] Open
Abstract
Introduction Osteoarthritis (OA) is a common inflammatory joint disease characterised by progressive cartilage destruction. Management of this condition remains a significant challenge, and new therapies are required. We investigated the protective effects of miR-106a mimics in a murine model of OA. Material and methods This study was performed using both in vitro and in vivo OA models. Primary chondrocytes were isolated from female rats, with inflammation induced via treatment with lipopolysaccharide (LPS). Then the effects of a miR-106a mimic were examined based on the level of inflammatory cytokine production and apoptotic signalling following LPS stimulation. An in vivo rat model of OA was generated by injecting LPS into the anterior cruciate ligament, followed by treatment with miR-106a mimics. Then, inflammatory and apoptotic protein expression was assessed in the cartilage tissue. Results Treatment with miR-106a mimic reduced the levels of inflammatory cytokines and apoptotic proteins in cartilage tissues following LPS-induced inflammation. Furthermore, the mimic ameliorated the expression of DR-6 mRNA and DR6, IκBα, and p65 proteins in chondrocytes. Similar effects were seen in the in vivo model, with the mimic attenuating expression of NF-κB, p65, IκBα, and DR6 proteins and improving histopathological outcomes in the chondrocytes of OA rats. Conclusions Treatment with miR-106a mimic ameliorates inflammation in cartilage tissues of OA subjects by activating death receptor 6 via the NF-κB signalling pathway.
Collapse
Affiliation(s)
- Luping Cui
- Department of Rheumatology and Immunology, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi Province, China
| | - Yongbin Han
- Department of Orthopedics, The First Hospital of Shan Xi Medical University, Taiyuan, Shanxi Province, China
| | - Zhijie Dong
- Department of Orthopedics, Hebei Province People’s Hospital, Shijiazhuang, Hebei Province, China
| |
Collapse
|
449
|
Sun X, Xiao L, Chen J, Chen X, Chen X, Yao S, Li H, Zhao G, Ma J. DNA methylation is involved in the pathogenesis of osteoarthritis by regulating CtBP expression and CtBP-mediated signaling. Int J Biol Sci 2020; 16:994-1009. [PMID: 32140068 PMCID: PMC7053340 DOI: 10.7150/ijbs.39945] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/26/2019] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis (OA) is a common type of arthritis. Chronic inflammation is an important contributor to the pathogenesis of OA. The maturation and secretion of proinflammatory cytokines are controlled by inflammasomes, especially NLRP1 (NLR Family Pyrin Domain Containing 1) and NLRP3. In this study, we identified a transactivation mechanism of NLRP3 mediated by CtBPs (C-terminal-binding proteins). We found that both the mRNA and protein levels of CtBPs were significantly increased in OA biopsies. Analyzing the profiles of differentially expressed genes in CtBP-knockdown and overexpression cells, we found that the expression of NLRP3 was dependent on CtBP levels. By the knockdown or overexpression of transcription factors that potentially bind to the promoter of NLRP3, we found that only AP1 could specifically regulate the expression of NLRP3. Using immunoprecipitation (IP) and Co-IP assays, we found that AP1 formed a transcriptional complex with a histone acetyltransferase p300 and CtBPs. The knockdown of any member of this transcriptional complex resulted in a decrease in the expression of NLRP3. To explore the underlying mechanism of CtBP overexpression, we analyzed their promoters and found that they were abundant in CpG islands. Treatment with the DNA methylation inhibitor 5-aza-2′-deoxycytidine (AZA) or knockdown of DNMTs (DNA methyltransferases) resulted in the overexpression of CtBPs, while overexpression of DNMTs caused the reverse effects on CtBP expression. Collectively, our results suggest that the decreased DNA methylation levels in the promoters of CtBPs upregulate their expression. Increased CtBPs associated with p300 and AP1 to form a transcriptional complex and activate the expression of NLRP3 and its downstream signaling, eventually aggravating the inflammatory response and leading to the pathogenesis of OA.
Collapse
Affiliation(s)
- Xiangxiang Sun
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Lin Xiao
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Juan Chen
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Xun Chen
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Xinlin Chen
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Shuxin Yao
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Hui Li
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Guanghui Zhao
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| | - Jianbing Ma
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi, China
| |
Collapse
|
450
|
Zharova TA, Kogan EA, Makarov VI, Smorchkov MM, Lychagin AV, Ivannikov SV, Zharkov NV, Loschenov VB. Correlation of synovial caspase-3 concentration and the photodynamic effectiveness in osteoarthritis treatment. Photodiagnosis Photodyn Ther 2020; 30:101669. [PMID: 31988026 DOI: 10.1016/j.pdpdt.2020.101669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/11/2019] [Accepted: 01/21/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND The present study focuses on investigation of Intra-articular PDT mechanisms for OA treatment. Also, a search for determination of the most effective dose of chlorin e6 (Ce6) for anti-inflammatory PDT of OA was carried out. METHODS The study was carried out on laboratory animals (11 Chinchilla rabbits, 1 year, 2.5 kg) with a gonarthritis model of post-traumatic OA. According to the instructions for using Photoditazin (Ce6 based PS) for PDT of human oncological and non-oncological diseases, the recommended dose is 0.7-1.2 mg/kg. For studies on rabbits, taking into account the conversion coefficient (3.2), the PS doses of 2.4, 3.2 and 6.4 mg/kg were selected. Fluorescence spectra were measured intra-articular before and after PDT using spectrometer with fiber-optic probe. The intrajoint PDT was carried out using a laser (662 ± 10 nm) and a fiber-optic catheter with a cylindrical diffuser inside a sapphire needle for a uniform distribution of the laser radiation. The immunohistochemical study was carried out by staining the samples with caspase-3. RESULTS Histological and immunohistochemical analysis showed that the best PS dose for intravenous administration for PDT of rabbit gonarthritis is 3.2 mg/kg. The PS concentration directly in the synovial tissue was 0.5 mg/kg, and this was enough to achieve the most positive results to reduce the caspase-3 level. CONCLUSION The caspase-3 level correlates well with other signs of inflammation in the synovial membrane (edema, etc.). Therefore, to assess the PDT effectiveness in the treatment of gonarthritis accompanied by synovitis, it is sufficient to analyze only for caspase-3. The efficacy of PDT with Ce6 showed that 3.2 mg/kg PS dose (1 mg/kg for a human) is the most effective.
Collapse
Affiliation(s)
- T A Zharova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Trubetskaya Str., 8-2, Russian Federation.
| | - E A Kogan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Trubetskaya Str., 8-2, Russian Federation.
| | - V I Makarov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991, Moscow, Vavilov Str., 38, Russian Federation.
| | - M M Smorchkov
- N.N. Priorov Central Research Institute of Traumatology and Orthopedics, 127299, Moscow, Priorov Str., 10, Russian Federation.
| | - A V Lychagin
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Trubetskaya Str., 8-2, Russian Federation.
| | - S V Ivannikov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Trubetskaya Str., 8-2, Russian Federation.
| | - N V Zharkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991, Moscow, Trubetskaya Str., 8-2, Russian Federation.
| | - V B Loschenov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 119991, Moscow, Vavilov Str., 38, Russian Federation; National Research Nuclear University MEPhI (Moscow Engineering Physics Institute), 115409, Moscow, Kashirskoe Shosse, 31, Russian Federation.
| |
Collapse
|