1
|
Geraghty T, Ishihara S, Obeidat AM, Adamczyk NS, Hunter RS, Li J, Wang L, Lee H, Ko FC, Malfait AM, Miller RE. Acute systemic macrophage depletion in osteoarthritic mice alleviates pain-related behaviors and does not affect joint damage. Arthritis Res Ther 2024; 26:224. [PMID: 39707543 PMCID: PMC11660666 DOI: 10.1186/s13075-024-03457-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 12/08/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a painful degenerative joint disease and a leading source of years lived with disability globally due to inadequate treatment options. Neuroimmune interactions reportedly contribute to OA pain pathogenesis. Notably, in rodents, macrophages in the DRG are associated with onset of persistent OA pain. Our objective was to determine the effects of acute systemic macrophage depletion on pain-related behaviors and joint damage using surgical mouse models in both sexes. METHODS We depleted CSF1R + macrophages by treating male macrophage Fas-induced apoptosis (MaFIA) transgenic mice 8- or 16-weeks post destabilization of the medial meniscus (DMM) with AP20187 or vehicle control (10 mg/kg i.p., 1x/day for 5 days), or treating female MaFIA mice 12 weeks post partial meniscectomy (PMX) with AP20187 or vehicle control. We measured pain-related behaviors 1-3 days before and after depletion, and, 3-4 days after the last injection we examined joint histopathology and performed flow cytometry of the dorsal root ganglia (DRGs). In a separate cohort of male 8-week DMM mice or age-matched naïve vehicle controls, we conducted DRG bulk RNA-sequencing analyses after the 5-day vehicle or AP20187 treatment. RESULTS Eight- and 16-weeks post DMM in male mice, AP20187-induced macrophage depletion resulted in attenuated mechanical allodynia and knee hyperalgesia. Female mice showed alleviation of mechanical allodynia, knee hyperalgesia, and weight bearing deficits after macrophage depletion at 12 weeks post PMX. Macrophage depletion did not affect the degree of cartilage degeneration, osteophyte width, or synovitis in either sex. Flow cytometry of the DRG revealed that macrophages and neutrophils were reduced after AP20187 treatment. In addition, in the DRG, only MHCII + M1-like macrophages were significantly decreased, while CD163 + MHCII- M2-like macrophages were not affected in both sexes. DRG bulk RNA-seq revealed that Cxcl10 and Il1b were upregulated with DMM surgery compared to naïve mice, and downregulated in DMM after acute macrophage depletion. CONCLUSIONS Acute systemic macrophage depletion reduced the levels of pro-inflammatory macrophages in the DRG and alleviated pain-related behaviors in established surgically induced OA in mice of both sexes, without affecting joint damage. Overall, these studies provide insight into immune cell regulation in the DRG during OA.
Collapse
Affiliation(s)
- Terese Geraghty
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Natalie S Adamczyk
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Rahel S Hunter
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Jun Li
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Lai Wang
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Hoomin Lee
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Frank C Ko
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA.
- Chicago Center on Musculoskeletal Pain, Chicago, IL, USA.
| |
Collapse
|
2
|
McClure JJ, McIlroy GD, Symons RA, Clark SM, Cunningham I, Han W, Kania K, Colella F, Rochford JJ, De Bari C, Roelofs AJ. Disentangling the detrimental effects of local from systemic adipose tissue dysfunction on articular cartilage in the knee. Osteoarthritis Cartilage 2024; 32:1552-1565. [PMID: 39103079 DOI: 10.1016/j.joca.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 08/07/2024]
Abstract
OBJECTIVE Obesity increases osteoarthritis (OA) risk due to adipose tissue dysfunction with associated metabolic syndrome and excess weight. Lipodystrophy syndromes exhibit systemic metabolic and inflammatory abnormalities similar to obesity without biomechanical overloading. Here, we used lipodystrophy mouse models to investigate the effects of systemic versus intra-articular adipose tissue dysfunction on the knee. METHODS Intra-articular adipose tissue development was studied using reporter mice. Mice with selective lipodystrophy of intra-articular adipose tissue were generated by conditional knockout (cKO) of Bscl2 in Gdf5-lineage cells, and compared with whole-body Bscl2 knockout (KO) mice with generalised lipodystrophy and associated systemic metabolic dysfunction. OA was induced by surgically destabilising the medial meniscus (DMM) and obesity by high-fat diet (HFD). Gene expression was analysed by quantitative RT-PCR and tissues were analysed histologically. RESULTS The infrapatellar fat pad (IFP), in contrast to overlying subcutaneous adipose tissue, developed from a template established from the Gdf5-expressing joint interzone during late embryogenesis, and was populated shortly after birth by adipocytes stochastically arising from Pdgfrα-expressing Gdf5-lineage progenitors. While female Bscl2 KO mice with generalised lipodystrophy developed spontaneous knee cartilage damage, Bscl2 cKO mice with intra-articular lipodystrophy did not, despite the presence of synovial hyperplasia and inflammation of the residual IFP. Furthermore, male Bscl2 cKO mice showed no worse cartilage damage after DMM. However, female Bscl2 cKO mice showed increased susceptibility to the cartilage-damaging effects of HFD-induced obesity. CONCLUSION Our findings emphasise the prevalent role of systemic metabolic and inflammatory effects in impairing cartilage homeostasis, with a modulatory role for intra-articular adipose tissue.
Collapse
Affiliation(s)
- Jessica J McClure
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - George D McIlroy
- The Rowett Institute and Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Rebecca A Symons
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Susan M Clark
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Iain Cunningham
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Weiping Han
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A⁎STAR), Singapore
| | - Karolina Kania
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Fabio Colella
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Justin J Rochford
- The Rowett Institute and Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Cosimo De Bari
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Anke J Roelofs
- Arthritis & Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
3
|
Oláh T, Cucchiarini M, Madry H. Temporal progression of subchondral bone alterations in OA models involving induction of compromised meniscus integrity in mice and rats: A scoping review. Osteoarthritis Cartilage 2024; 32:1220-1234. [PMID: 38876436 DOI: 10.1016/j.joca.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/17/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024]
Abstract
OBJECTIVE To categorize the temporal progression of subchondral bone alterations induced by compromising meniscus integrity in mouse and rat models of knee osteoarthritis (OA). METHOD Scoping review of investigations reporting subchondral bone changes with appropriate negative controls in the different mouse and rat models of OA induced by compromising meniscus integrity. RESULTS The available literature provides appropriate temporal detail on subchondral changes in these models, covering the entire spectrum of OA with an emphasis on early and mid-term time points. Microstructural changes of the subarticular spongiosa are comprehensively described; those of the subchondral bone plate are not. In mouse models, global subchondral bone alterations are unidirectional, involving an advancing sclerosis of the trabecular structure over time. In rats, biphasic subchondral bone alterations begin with an osteopenic degeneration and loss of subchondral trabeculae, progressing to a late sclerosis of the entire subchondral bone. Rat models, independently from the applied technique, relatively faithfully mirror the early bone loss detected in larger animals, and the late subchondral bone sclerosis observed in human advanced OA. CONCLUSION Mice and rats allow us to study the microstructural consequences of compromising meniscus integrity at high temporal detail. Thickening of the subchondral bone plate, an early loss of thinner subarticular trabecular elements, followed by a subsequent sclerosis of the entire subchondral bone are all important and reliable hallmarks that occur in parallel with the advancing articular cartilage degeneration. Thoughtful decisions on the study design, laterality, selection of controls and volumes of interest are crucial to obtain meaningful data.
Collapse
Affiliation(s)
- Tamás Oláh
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany; Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany.
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany.
| |
Collapse
|
4
|
Obeidat AM, Kim SY, Burt KG, Hu B, Li J, Ishihara S, Xiao R, Miller RE, Little C, Malfait AM, Scanzello CR. A standardized approach to evaluation and reporting of synovial histopathology in two surgically induced murine models of osteoarthritis. Osteoarthritis Cartilage 2024; 32:1273-1282. [PMID: 38823432 PMCID: PMC11408105 DOI: 10.1016/j.joca.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/03/2024]
Abstract
OBJECTIVE Synovial pathology has been linked to osteoarthritis (OA) pain in patients. Microscopic grading systems for synovial changes in human OA have been described, but a standardized approach for murine models of OA is needed. We sought to develop a reproducible approach and set of minimum recommendations for reporting of synovial histopathology in mouse models of OA. METHODS Coronal and sagittal sections from male mouse knee joints subjected to destabilization of medial meniscus (DMM) or partial meniscectomy (PMX) were collected as part of other studies. Stains included Hematoxylin and Eosin (H&E), Toluidine Blue (T-Blue), and Safranin O/Fast Green (Saf-O). Four blinded readers graded pathological features (hyperplasia, cellularity, and fibrosis) at specific anatomic locations. Inter-reader agreement of each feature score was determined. RESULTS There was acceptable to very good agreement when using 3-4 individual readers. After DMM and PMX, expected medial predominant changes in hyperplasia and cellularity were observed, with fibrosis noted at 12 weeks post-PMX. Synovial changes were consistent from section to section in the mid-joint area. When comparing stains, H&E and T-blue resulted in better agreement compared to Saf-O stain. CONCLUSIONS To account for the pathologic and anatomic variability in synovial pathology and allow for a more standardized evaluation that can be compared across studies, we recommend evaluating a minimum set of 3 pathological features at standardized anatomic areas. Further, we suggest reporting individual feature scores separately before relying on a single summed "synovitis" score. H&E or T-blue are preferred, inter-reader agreement for each feature should be considered.
Collapse
Affiliation(s)
- Alia M Obeidat
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago, IL, United States.
| | - Sung Yeon Kim
- University of Pennsylvania School of Engineering and Applied Sciences, Philadelphia, PA 19104, United States.
| | - Kevin G Burt
- Translational Musculoskeletal Research Center, Corp. Michael J Crescenz VA Medical Center, Philadelphia, PA 19104, United States; Department of Orthopaedic Surgery, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Baofeng Hu
- Translational Musculoskeletal Research Center, Corp. Michael J Crescenz VA Medical Center, Philadelphia, PA 19104, United States; Division of Rheumatology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Jun Li
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago, IL, United States.
| | - Shingo Ishihara
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago, IL, United States.
| | - Rui Xiao
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Department of Pediatrics Division of Biostatistics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States.
| | - Rachel E Miller
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago, IL, United States.
| | - Christopher Little
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney, NSW 2065, Australia.
| | - Anne-Marie Malfait
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago, IL, United States.
| | - Carla R Scanzello
- Translational Musculoskeletal Research Center, Corp. Michael J Crescenz VA Medical Center, Philadelphia, PA 19104, United States; Division of Rheumatology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
5
|
Bergman RF, Lammlin L, Junginger L, Farrell E, Goldman S, Darcy R, Rasner C, Obeidat AM, Malfait AM, Miller RE, Maerz T. Sexual dimorphism of the synovial transcriptome underpins greater PTOA disease severity in male mice following joint injury. Osteoarthritis Cartilage 2024; 32:1060-1073. [PMID: 37716404 DOI: 10.1016/j.joca.2023.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/20/2023] [Accepted: 07/26/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a disease with sex-dependent prevalence and severity in both human and animal models. We sought to elucidate sex differences in synovitis, mechanical sensitization, structural damage, bone remodeling, and the synovial transcriptome in the anterior cruciate ligament rupture (ACLR) mouse model of post-traumatic OA (PTOA). DESIGN Male and female 12-week-old C57/BL6J mice were randomized to Sham or noninvasive ACLR with harvests at 7d or 28d post-ACLR (n = 9 per sex in each group - Sham, 7d ACLR, 28d ACLR). Knee hyperalgesia, mechanical allodynia, and intra-articular matrix metalloproteinase (MMP) activity (via intravital imaging) were measured longitudinally. Trabecular and subchondral bone (SCB) remodeling and osteophyte formation were assessed by µCT. Histological scoring of PTOA, synovitis, and anti-MMP13 immunostaining were performed. NaV1.8-Cre;tdTomato mice were used to document localization and sprouting of nociceptors. Bulk RNA-seq of synovium in Sham, 7d, and 28d post-ACLR, and contralateral joints (n = 6 per group per sex) assessed injury-induced and sex-dependent gene expression. RESULTS Male mice exhibited more severe joint damage at 7d and 28d and more severe synovitis at 28d, accompanied by 19% greater MMP activity, 8% lower knee hyperalgesia threshold, and 43% lower hindpaw withdrawal threshold in injured limbs compared to female injured limbs. Females had injury-induced catabolic responses in trabecular and SCB, whereas males exhibited 133% greater normalized osteophyte volume relative to females and sclerotic remodeling of trabecular and SCB. NaV1.8+ nociceptor sprouting in SCB and medial synovium was induced by injury and comparable between sexes. RNA-seq of synovium demonstrated similar injury-induced transcriptomic programs between the sexes at 7d, but only female mice exhibited a transcriptomic signature indicative of synovial inflammatory resolution by 28d, whereas males had persistent pro-inflammatory, pro-fibrotic, pro-neurogenic, and pro-angiogenic gene expression. CONCLUSION Male mice exhibited more severe overall joint damage and pain behavior after ACLR, which was associated with persistent activation of synovial inflammatory, fibrotic, and neuroangiogenic processes, implicating persistent synovitis in driving sex differences in murine PTOA.
Collapse
Affiliation(s)
- Rachel F Bergman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Lindsey Lammlin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Lucas Junginger
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Easton Farrell
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Sam Goldman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Rose Darcy
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Cody Rasner
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Alia M Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University, Chicago, IL, United States
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University, Chicago, IL, United States
| | - Rachel E Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University, Chicago, IL, United States
| | - Tristan Maerz
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
6
|
Zhang C, Pathrikar TV, Baby HM, Li J, Zhang H, Selvadoss A, Ovchinnikova A, Ionescu A, Chubinskaya S, Miller RE, Bajpayee AG. Charge-Reversed Exosomes for Targeted Gene Delivery to Cartilage for Osteoarthritis Treatment. SMALL METHODS 2024; 8:e2301443. [PMID: 38607953 PMCID: PMC11470115 DOI: 10.1002/smtd.202301443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/18/2024] [Indexed: 04/14/2024]
Abstract
Gene therapy has the potential to facilitate targeted expression of therapeutic proteins to promote cartilage regeneration in osteoarthritis (OA). The dense, avascular, aggrecan-glycosaminoglycan (GAG) rich negatively charged cartilage, however, hinders their transport to reach chondrocytes in effective doses. While viral vector mediated gene delivery has shown promise, concerns over immunogenicity and tumorigenic side-effects persist. To address these issues, this study develops surface-modified cartilage-targeting exosomes as non-viral carriers for gene therapy. Charge-reversed cationic exosomes are engineered for mRNA delivery by anchoring cartilage targeting optimally charged arginine-rich cationic motifs into the anionic exosome bilayer by using buffer pH as a charge-reversal switch. Cationic exosomes penetrated through the full-thickness of early-stage arthritic human cartilage owing to weak-reversible ionic binding with GAGs and efficiently delivered the encapsulated eGFP mRNA to chondrocytes residing in tissue deep layers, while unmodified anionic exosomes do not. When intra-articularly injected into destabilized medial meniscus mice knees with early-stage OA, mRNA loaded charge-reversed exosomes overcame joint clearance and rapidly penetrated into cartilage, creating an intra-tissue depot and efficiently expressing eGFP; native exosomes remained unsuccessful. Cationic exosomes thus hold strong translational potential as a platform technology for cartilage-targeted non-viral delivery of any relevant mRNA targets for OA treatment.
Collapse
Affiliation(s)
- Chenzhen Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Tanvi V. Pathrikar
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Helna M. Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Jun Li
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | | | - Andreia Ionescu
- Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Susan Chubinskaya
- Department of Pediatrics, Rush University Medical College, Chicago, IL 60612, USA
| | - Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ambika G. Bajpayee
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
7
|
Geraghty T, Ishihara S, Obeidat AM, Adamczyk NS, Hunter RS, Li J, Wang L, Lee H, Ko FC, Malfait AM, Miller RE. Acute systemic macrophage depletion in osteoarthritic mice alleviates pain-related behaviors and does not affect joint damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.608301. [PMID: 39229102 PMCID: PMC11370380 DOI: 10.1101/2024.08.16.608301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background Osteoarthritis (OA) is a painful degenerative joint disease and a leading source of years lived with disability globally due to inadequate treatment options. Neuroimmune interactions reportedly contribute to OA pain pathogenesis. Notably, in rodents, macrophages in the DRG are associated with onset of persistent OA pain. Our objective was to determine the effects of acute systemic macrophage depletion on pain-related behaviors and joint damage using surgical mouse models in both sexes. Methods We depleted CSF1R+ macrophages by treating male macrophage Fas-induced apoptosis (MaFIA) transgenic mice 8- or 16-weeks post destabilization of the medial meniscus (DMM) with AP20187 or vehicle control (10 mg/kg i.p., 1x/day for 5 days), or treating female MaFIA mice 12 weeks post partial meniscectomy (PMX) with AP20187 or vehicle control. We measured pain-related behaviors 1-3 days before and after depletion, and, 3-4 days after the last injection we examined joint histopathology and performed flow cytometry of the dorsal root ganglia (DRGs). In a separate cohort of male 8-week DMM mice or age-matched naïve vehicle controls, we conducted DRG bulk RNA-sequencing analyses after the 5-day vehicle or AP20187 treatment. Results Eight- and 16-weeks post DMM in male mice, AP20187-induced macrophage depletion resulted in attenuated mechanical allodynia and knee hyperalgesia. Female mice showed alleviation of mechanical allodynia, knee hyperalgesia, and weight bearing deficits after macrophage depletion at 12 weeks post PMX. Macrophage depletion did not affect the degree of cartilage degeneration, osteophyte width, or synovitis in either sex. Flow cytometry of the DRG revealed that macrophages and neutrophils were reduced after AP20187 treatment. In addition, in the DRG, only MHCII+ M1-like macrophages were significantly decreased, while CD163+MHCII- M2-like macrophages were not affected in both sexes. DRG bulk RNA-seq revealed that Cxcl10 and Il1b were upregulated with DMM surgery compared to naïve mice, and downregulated in DMM after acute macrophage depletion. Conclusions Acute systemic macrophage depletion reduced the levels of pro-inflammatory macrophages in the DRG and alleviated pain-related behaviors in established surgically induced OA in mice of both sexes, without affecting joint damage. Overall, these studies provide insight into immune cell regulation in the DRG during OA.
Collapse
Affiliation(s)
- Terese Geraghty
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Shingo Ishihara
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Alia M Obeidat
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Natalie S Adamczyk
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Rahel S Hunter
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Jun Li
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Lai Wang
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Hoomin Lee
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Frank C Ko
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Anne-Marie Malfait
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| | - Rachel E Miller
- Rush University Medical Center, Department of Internal Medicine, Division of Rheumatology, Chicago, IL USA
- Rush University Medical Center, Department of Anatomy & Cell Biology, Chicago, IL USA
| |
Collapse
|
8
|
Tong MJ, Song MX, Liu Z, Yu W, Wang CZ, Cai CD, Zhang YK, Zhang YQ, Wang LP, Zhu ZZ, Yin XF, Yan ZQ. A Bionic Thermosensitive Sustainable Delivery System for Reversing the Progression of Osteoarthritis by Remodeling the Joint Homeostasis. Adv Healthc Mater 2024; 13:e2303792. [PMID: 38394066 DOI: 10.1002/adhm.202303792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Although the pathogenesis of osteoarthritis (OA) is unclear, inflammatory cytokines are related to its occurrence. However, few studies focused on the therapeutic strategies of regulating joint homeostasis by simultaneously remodeling the anti-inflammatory and immunomodulatory microenvironments. Fibroblast growth factor 18 (FGF18) is the only disease-modifying OA drug (DMOAD) with a potent ability and high efficiency in maintaining the phenotype of chondrocytes within cell culture models. However, its potential role in the immune microenvironment remains unknown. Besides, information on an optimal carrier, whose interface and chondral-biomimetic microenvironment mimic the native articular tissue, is still lacking, which substantially limits the clinical efficacy of FGF18. Herein, to simulate the cartilage matrix, chondroitin sulfate (ChS)-based nanoparticles (NPs) are integrated into poly(D, L-lactide)-poly(ethylene glycol)-poly(D, L-lactide) (PLEL) hydrogels to develop a bionic thermosensitive sustainable delivery system. Electrostatically self-assembled ChS and ε-poly-l-lysine (EPL) NPs are prepared for the bioencapsulation of FGF18. This bionic delivery system suppressed the inflammatory response in interleukin-1β (IL-1β)-mediated chondrocytes, promoted macrophage M2 polarization, and inhibited M1 polarization, thereby ameliorating cartilage degeneration and synovitis in OA. Thus, the ChS-based hydrogel system offers a potential strategy to regulate the chondrocyte-macrophage crosstalk, thus re-establishing the anti-inflammatory and immunomodulatory microenvironment for OA therapy.
Collapse
Affiliation(s)
- Min-Ji Tong
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Meng-Xiong Song
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Zhe Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Wei Yu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chen-Zhong Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chuan-Dong Cai
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ying-Kai Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yue-Qi Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li-Peng Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhen-Zhong Zhu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xiao-Fan Yin
- Department of Orthopedic Surgery, Minhang Hospital, Fudan University, Shanghai, 201100, China
| | - Zuo-Qin Yan
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
9
|
Xie W, Jiang S, Donat A, Knapstein PR, Albertsen LC, Kokot JL, Erdmann C, Rolvien T, Frosch KH, Baranowsky A, Keller J. Tranexamic Acid Attenuates the Progression of Posttraumatic Osteoarthritis in Mice. Am J Sports Med 2024; 52:766-778. [PMID: 38305280 PMCID: PMC10905980 DOI: 10.1177/03635465231220855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/25/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND Posttraumatic osteoarthritis (OA) is a common disorder associated with a high socioeconomic burden, particularly in young, physically active, and working patients. Tranexamic acid (TXA) is commonly used in orthopaedic trauma surgery as an antifibrinolytic agent to control excessive bleeding. Previous studies have reported that TXA modulates inflammation and bone cell function, both of which are dysregulated during posttraumatic OA disease progression. PURPOSE To evaluate the therapeutic effects of systemic and topical TXA treatment on the progression of posttraumatic OA in the knee of mice. STUDY DESIGN Controlled laboratory study. METHODS OA was induced via anterior cruciate ligament (ACL) transection on the right knee of female mice. Mice were treated with TXA or vehicle intraperitoneally daily or intra-articularly weekly for 4 weeks, starting on the day of surgery. Articular cartilage degeneration, synovitis, bone erosion, and osteophyte formation were scored histologically. Micro-computed tomography evaluation was conducted to measure the subchondral bone microstructure and osteophyte volume. Cartilage thickness and bone remodeling were assessed histomorphometrically. RESULTS Both systemic and topical TXA treatment significantly reduced cartilage degeneration, synovitis, and bone erosion scores and increased the ratio of hyaline to calcified cartilage thickness in posttraumatic OA. Systemic TXA reversed ACL transection-induced subchondral bone loss and osteophyte formation, whereas topical treatment had no effect. Systemic TXA decreased the number and surface area of osteoclasts, whereas those of osteoblasts were not affected. No effect of topical TXA on osteoblast or osteoclast parameters was observed. CONCLUSION Both systemic and topical TXA exerted protective effects on the progression of posttraumatic OA. Drug repurposing of TXA may, therefore, be useful for the prevention or treatment of posttraumatic OA, particularly after ACL surgery. CLINICAL RELEVANCE TXA might be beneficial in patients with posttraumatic OA of the knee.
Collapse
Affiliation(s)
- Weixin Xie
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Shan Jiang
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Donat
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Richard Knapstein
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lilly-Charlotte Albertsen
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Judith Luisa Kokot
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cordula Erdmann
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Rolvien
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karl-Heinz Frosch
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Baranowsky
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Keller
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
10
|
Patnaik R, Riaz S, Sivani BM, Faisal S, Naidoo N, Rizzo M, Banerjee Y. Evaluating the potential of Vitamin D and curcumin to alleviate inflammation and mitigate the progression of osteoarthritis through their effects on human chondrocytes: A proof-of-concept investigation. PLoS One 2023; 18:e0290739. [PMID: 38157375 PMCID: PMC10756552 DOI: 10.1371/journal.pone.0290739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 08/14/2023] [Indexed: 01/03/2024] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disorder primarily affecting the elderly, characterized by a prominent inflammatory component. The long-term side effects associated with current therapeutic approaches necessitate the development of safer and more efficacious alternatives. Nutraceuticals, such as Vitamin D and curcumin, present promising therapeutic potentials due to their safety, efficacy, and cost-effectiveness. In this study, we utilized a proinflammatory human chondrocyte model of OA to assess the anti-inflammatory properties of Vitamin D and curcumin, with a particular focus on the Protease-Activated Receptor-2 (PAR-2) mediated inflammatory pathway. Employing a robust siRNA approach, we effectively modulated the expression of PAR-2 to understand its role in the inflammatory process. Our results reveal that both Vitamin D and curcumin attenuate the expression of PAR-2, leading to a reduction in the downstream proinflammatory cytokines, such as Tumor Necrosis Factor-alpha (TNF-α), Interleukin 6 (IL-6), and Interleukin 8 (IL-8), implicated in the OA pathogenesis. Concurrently, these compounds suppressed the expression of Receptor Activator of Nuclear Factor kappa-Β Ligand (RANKL) and its receptor RANK, which are associated with PAR-2 mediated TNF-α stimulation. Additionally, Vitamin D and curcumin downregulated the expression of Interferon gamma (IFN-γ), known to elevate RANKL levels, underscoring their potential therapeutic implications in OA. This study, for the first time, provides evidence of the mitigating effect of Vitamin D and curcumin on PAR-2 mediated inflammation, employing an siRNA approach in OA. Thus, our findings pave the way for future research and the development of novel, safer, and more effective therapeutic strategies for managing OA.
Collapse
Affiliation(s)
- Rajashree Patnaik
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Sumbal Riaz
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Bala Mohan Sivani
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Shemima Faisal
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Nerissa Naidoo
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine, and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Yajnavalka Banerjee
- College of Medicine and Health Sciences, Mohammed Bin Rashid University of Medicine, and Health Sciences (MBRU), Dubai, United Arab Emirates
- Centre for Medical Education, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
11
|
Haubruck P, Heller R, Blaker CL, Clarke EC, Smith SM, Burkhardt D, Liu Y, Stoner S, Zaki S, Shu CC, Little CB. Streamlining quantitative joint-wide medial femoro-tibial histopathological scoring of mouse post-traumatic knee osteoarthritis models. Osteoarthritis Cartilage 2023; 31:1602-1611. [PMID: 37716405 DOI: 10.1016/j.joca.2023.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 07/03/2023] [Accepted: 07/26/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVES Histological scoring remains the gold-standard for quantifying post-traumatic osteoarthritis (ptOA) in animal models, allowing concurrent evaluation of numerous joint tissues. Available systems require scoring multiple sections/joint making analysis laborious and expensive. We investigated if a single section allowed equivalent quantitation of pathology in different joint tissues and disease stages, in three ptOA models. METHOD Male 10-12-week-old C57BL/6 mice underwent surgical medial-meniscal-destabilization, anterior-cruciate-ligament (ACL) transection, non-invasive-ACL-rupture, or served as sham-surgical, non-invasive-ACL-strain, or naïve/non-operated controls. Mice (n = 12/group) were harvested 1-, 4-, 8-, and 16-week post-intervention. Serial sagittal toluidine-blue/fast-green stained sections of the medial-femoro-tibial joint (n = 7/joint, 84 µm apart) underwent blinded scoring of 40 histology-outcomes. We evaluated agreement between single-slide versus entire slide-set maximum or median scores (weighted-kappa), and sensitivity/specificity of single-slide versus median/maximum to detect OA pathology. RESULTS A single optimal mid-sagittal section showed excellent agreement with median (weighted-kappa 0.960) and maximum (weighted-kappa 0.926) scores. Agreement for individual histology-outcomes was high with only 19/240 median and 15/240 maximum scores having a weighted-kappa ≤0.4, the majority of these (16/19 and 11/15) in control groups. Statistically-significant histology-outcome differences between ptOA models and their controls detected with the entire slide-set were reliably reproduced using a single slide (sensitivity >93.15%, specificity >93.10%). The majority of false-negatives with single-slide scoring were meniscal and subchondral bone histology-outcomes (89%) and occurred in weeks 1-4 post-injury (84%). CONCLUSION A single mid-sagittal slide reduced the time needed to score diverse histopathological changes by 87% without compromising the sensitivity or specificity of the analysis, across a variety of ptOA models and time-points.
Collapse
Affiliation(s)
- Patrick Haubruck
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, D-69118 Heidelberg, Germany; Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Raban Heller
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, D-69118 Heidelberg, Germany; Institute for Experimental Endocrinology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353 Berlin, Germany; Bundeswehr Hospital Berlin, Clinic of Traumatology and Orthopaedics, D-10115 Berlin, Germany
| | - Carina L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia; Murray Maxwell Biomechanics Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Elizabeth C Clarke
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Susan M Smith
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Daniel Burkhardt
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Yolanda Liu
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Shihani Stoner
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Sanaa Zaki
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia; Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia
| | - Cindy C Shu
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, New South Wales 2065, Australia.
| |
Collapse
|
12
|
Donnenfield JI, Fleming BC, Proffen BL, Podury A, Murray MM. Microscopic and transcriptomic changes in porcine synovium one year following disruption of the anterior cruciate ligament. Osteoarthritis Cartilage 2023; 31:1554-1566. [PMID: 37742942 PMCID: PMC10841386 DOI: 10.1016/j.joca.2023.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 09/26/2023]
Abstract
OBJECTIVE There is no disease-modifying treatment for posttraumatic osteoarthritis (PTOA). This may be partly due to an incomplete understanding of synovitis, which has been causally linked to PTOA progression. The microscopic and transcriptomic changes in synovium seen in early- to mid-stage PTOA were evaluated to better characterize this knowledge gap. METHODS Seventy-two Yucatan minipigs underwent transection of the anterior cruciate ligament (ACL). Subjects were randomized to no further intervention, ligament reconstruction, or ligament repair, followed by microscopic synovium evaluation and RNA-sequencing at 1, 4, and 52 weeks. Six additional subjects received no ligament transection and served as 1- and 4-week controls and 12 contralateral knees served as 52-week controls. RESULTS Synovial lining thickness, stromal cellularity, and overall microscopic synovitis reached their highest levels in the first few weeks following injury. Inflammatory infiltration continued to increase over the course of a year. Leaving the ACL transected, reconstructing the ligament, or repairing the ligament did not modulate synovitis development at 1, 4, or 52 weeks. Differential gene expression analysis of PTOA-affected synovium compared to control synovium revealed increased cell proliferation, angiogenesis, collagen breakdown, and diminished lipid metabolism at 1 and 4 weeks, and increased axonogenesis and focal adhesion with reduced immune activation at 52 weeks. CONCLUSIONS Synovitis was present one year after ACL injury and was not alleviated by surgical intervention. Gene expression in early synovitis was characterized by cell proliferation, angiogenesis, proteolysis, and reduced lipolysis, which was followed by nerve growth and cellular adhesion with less immune activation at 52 weeks.
Collapse
Affiliation(s)
- Jonah I Donnenfield
- Division of Sports Medicine, Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Braden C Fleming
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA.
| | - Benedikt L Proffen
- Division of Sports Medicine, Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Archana Podury
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA.
| | - Martha M Murray
- Division of Sports Medicine, Department of Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Obeidat AM, Kim SY, Burt KG, Hu B, Li J, Ishihara S, Xiao R, Miller RE, Little C, Malfait AM, Scanzello CR. Recommendations For a Standardized Approach to Histopathologic Evaluation of Synovial Membrane in Murine Models of Experimental Osteoarthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.14.562259. [PMID: 37904981 PMCID: PMC10614774 DOI: 10.1101/2023.10.14.562259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Background Synovial pathology has been linked to osteoarthritis (OA) pain in patients. Microscopic grading systems for synovial changes in human OA have been described, but a standardized approach for murine models of OA is needed. We sought to develop a reproducible approach and set of minimum recommendations for synovial histopathology in mouse models of OA. Methods Coronal and sagittal sections from male mouse knee joints subjected to destabilization of medial meniscus (DMM) or partial meniscectomy (PMX) were collected as part of other studies. Stains included Hematoxylin and Eosin (H&E), Toluidine Blue (T-Blue) and Safranin O/Fast Green (Saf-O). Four blinded readers graded pathological features (hyperplasia, cellularity, and fibrosis) at specific anatomic locations in the medial and lateral compartments. Inter-reader reliability of each feature was determined. Results There was acceptable to very good agreement between raters. After DMM, increased hyperplasia and cellularity and a trend towards increased fibrosis were observed 6 weeks after DMM in the medial locations, and persisted up to 16 weeks. In the PMX model, cellularity and hyperplasia were evident in both medial and lateral compartments while fibrotic changes were largely seen on the medial side. Synovial changes were consistent from section to section in the mid-joint area mice. H&E, T-blue, and Saf-O stains resulted in comparable reliability. Conclusions To allow for a standard evaluation that can be implemented and compared across labs and studies, we recommend using 3 readers to evaluate a minimum set of 3 pathological features at standardized anatomic areas. Pre-defining areas to be scored, and reliability for each pathologic feature should be considered.
Collapse
Affiliation(s)
- Alia M Obeidat
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago IL
| | - Sung Yeon Kim
- University of Pennsylvania School of Engineering and Applied Sciences, Philadelphia PA 19104
| | - Kevin G Burt
- Translational Musculoskeletal Research Center, Corp. Michael J Crescenz VA Medical Center, Philadelphia PA 19104
- Department of Orthopaedic Surgery, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104
| | - Baofeng Hu
- Translational Musculoskeletal Research Center, Corp. Michael J Crescenz VA Medical Center, Philadelphia PA 19104
- Division of Rheumatology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104
| | - Jun Li
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago IL
| | - Shingo Ishihara
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago IL
| | - Rui Xiao
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Department of Pediatrics Division of Biostatistics, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Rachel E Miller
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago IL
| | - Christopher Little
- Kolling Institute, Faculty of Medicine and Health, The University of Sydney, NSW, 2065, Australia
| | - Anne-Marie Malfait
- Division of Rheumatology, Department of Internal Medicine, Rush University Medical College, Chicago IL
| | - Carla R Scanzello
- Translational Musculoskeletal Research Center, Corp. Michael J Crescenz VA Medical Center, Philadelphia PA 19104
- Division of Rheumatology, Perelman School of Medicine University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
14
|
Julovi SM, Dao A, Trinh K, O'Donohue AK, Shu C, Smith S, Shingde M, Schindeler A, Rogers NM, Little CB. Disease-modifying interactions between chronic kidney disease and osteoarthritis: a new comorbid mouse model. RMD Open 2023; 9:e003109. [PMID: 37562858 PMCID: PMC10423836 DOI: 10.1136/rmdopen-2023-003109] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE The prevalence of comorbid chronic kidney disease (CKD) and osteoarthritis (OA) is increasing globally. While sharing common risk factors, the mechanism and consequences of concurrent CKD-OA are unclear. The aims of the study were to develop a preclinical comorbid model, and to investigate the disease-modifying interactions. METHODS Seventy (70) male 8-10 week-old C57BL/6 mice were subjected to 5/6 nephrectomy (5/6Nx)±destabilisation of medial meniscus (DMM) or sham surgery. OA pathology and CKD were assessed 12 weeks postinduction by blinded histology scoring, micro-CT, immunohistochemistry for osteoclast and matrix metalloproteinase (MMP)-13 activity, and serum analysis of bone metabolic markers. RESULTS The 5/6Nx model recapitulated characteristic features of CKD, with renal fibrosis and deranged serum alkaline phosphatase, calcium and phosphate. There was no histological evidence of cartilage pathology induced by 5/6Nx alone, however, synovial MMP-13 expression and subchondral bone osteoclastic activity were increased (p<0.05), with accompanying reductions (p<0.05) in subchondral trabecular bone, bone volume and mineral density. DMM significantly (p<0.05) increased tibiofemoral cartilage damage, subchondral bone sclerosis, marginal osteophytes and synovitis, in association with increased cartilage and synovial MMP-13. DMM alone induced (p<0.05) renal fibrosis, proteinuria and increased (p<0.05) 5/6Nx-induced serum urea. However, DMM in 5/6Nx-mice resulted in significantly reduced (p<0.05) cartilage pathology and marginal osteophyte development, in association with reduced subchondral bone volume and density, and inhibition of 5/6Nx-induced subchondral bone osteoclast activation. CONCLUSION This study assessed a world-first preclinical comorbid CKD-OA model. Our findings demonstrate significant bidirectional disease-modifying interaction between CKD and OA.
Collapse
Affiliation(s)
- Sohel M Julovi
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- The Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Aiken Dao
- The Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Bioengineering & Molecular Medicine (BAMM) Laboratory, the Children's Hospital at Westmead and the Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Katie Trinh
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- The Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alexandra K O'Donohue
- The Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Bioengineering & Molecular Medicine (BAMM) Laboratory, the Children's Hospital at Westmead and the Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Cindy Shu
- The Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Raymond Purves Bone and Joint Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Susan Smith
- Raymond Purves Bone and Joint Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Meena Shingde
- Department of Tissue Pathology and Diagnostic Oncology, Institute of Clinical Pathology and Medical Research, Wentworthville, New South Wales, Australia
| | - Aaron Schindeler
- The Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Bioengineering & Molecular Medicine (BAMM) Laboratory, the Children's Hospital at Westmead and the Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Natasha M Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- The Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Christopher B Little
- The Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Raymond Purves Bone and Joint Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
15
|
Grote CW, Mackay MJ, Lu Q, Liu X, Meyer AR, Wang J. A whole-joint histopathologic grading system for murine knee osteoarthritis. J Orthop Res 2023; 41:1407-1418. [PMID: 36370134 PMCID: PMC10175513 DOI: 10.1002/jor.25482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/27/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
This study aims to develop a comprehensive and easily executable histopathologic grading scheme for murine knee osteoarthritis (OA) using specific scoring criteria for both cartilage and periarticular changes, which may overcome important limitations of the existing grading systems. The new grading scheme was developed based on mouse knee OA models with observation periods up to 24 months of age (spontaneous OA) or 24-week post-injury (posttraumatic OA). Semi-quantitative assessments of the histopathologic OA changes were applied to all four quadrants per femorotibial joint for 50 joints (200 quadrants) using specific scoring criteria rather than mild to severe grades. Scoring elements per quadrant were as follows: cartilage lesion (0-7), osteophyte (0-3), subchondral bone change (0-3), synovitis (0-3), and ectopic periarticular soft-tissue chondrogenesis and ossification (0-3). The new histopathologic grading scheme had high intra- and interobserver reproducibility (correlation coefficients r > 0.95) across experienced and novice observers. Sensitivity and reliability analyses confirmed the ability of the new scheme to detect minimal but significant OA progression (p < 0.01) within a 2-week interval and to accurately identify tissue- and quadrant-specific OA severity within the joints. In conclusion, this study presents the first whole-joint histopathologic grading scheme for murine knee OA that covers all-stage osteoarthritic changes in all major joint tissues, including periarticular soft-tissue ossification that is not included in any of the existing OA grading systems. This reproducible scheme is easy to execute and sensitive to minimal OA progression without using computer software, suitable for quick OA severity assessments of the entire femorotibial joint.
Collapse
Affiliation(s)
- Caleb W. Grote
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Matthew J. Mackay
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Qinghua Lu
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Xiangliang Liu
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
| | - Anders R. Meyer
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jinxi Wang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
16
|
Fu Y, Batushansky A, Kinter M, Huebner JL, Kraus VB, Griffin TM. Effects of Leptin and Body Weight on Inflammation and Knee Osteoarthritis Phenotypes in Female Rats. JBMR Plus 2023; 7:e10754. [PMID: 37457883 PMCID: PMC10339097 DOI: 10.1002/jbm4.10754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/22/2023] [Accepted: 04/13/2023] [Indexed: 07/18/2023] Open
Abstract
Leptin is a proinflammatory adipokine that contributes to obesity-associated osteoarthritis (OA), especially in women. However, the extent to which leptin causes knee OA separate from the effect of increased body weight is not clear. We hypothesized that leptin is necessary to induce knee OA in obese female rats but not sufficient to induce knee OA in lean rats lacking systemic metabolic inflammation. The effect of obesity without leptin signaling was modeled by comparing female lean Zucker rats to pair fed obese Zucker rats, which possess mutant fa alleles of the leptin receptor gene. The effect of leptin without obesity was modeled in female F344BN F1 hybrid rats by systemically administering recombinant rat leptin versus saline for 23 weeks via osmotic pumps. Primary OA outcomes included cartilage histopathology and subchondral bone micro-computed tomography. Secondary outcomes included targeted cartilage proteomics, serum inflammation, and synovial fluid inflammation following an acute intra-articular challenge with interleukin-1β (IL-1β). Compared to lean Zucker rats, obese Zucker rats developed more severe tibial osteophytes and focal cartilage lesions in the medial tibial plateau, with modest changes in proximal tibial epiphysis trabecular bone structure. In contrast, exogenous leptin treatment, which increased plasma leptin sixfold without altering body weight, caused mild generalized cartilage fibrillation and reduced Safranin O staining compared to vehicle-treated animals. Leptin also significantly increased subchondral and trabecular bone volume and bone mineral density in the proximal tibia. Cartilage metabolic and antioxidant enzyme protein levels were substantially elevated with leptin deficiency and minimally suppressed with leptin treatment. In contrast, leptin treatment induced greater changes in systemic and local inflammatory mediators compared to leptin receptor deficiency, including reduced serum IL-6 and increased synovial fluid IL-1β. In conclusion, rat models that separately elevate leptin or body weight develop distinct OA-associated phenotypes, revealing how obesity increases OA pathology through both leptin-dependent and independent pathways. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Yao Fu
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Albert Batushansky
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
| | - Michael Kinter
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Oklahoma Center for GeroscienceUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Janet L. Huebner
- Duke Molecular Physiology InstituteDuke University, School of Medicine, Duke UniversityDurhamNorth CarolinaUSA
| | - Virginia B. Kraus
- Duke Molecular Physiology InstituteDuke University, School of Medicine, Duke UniversityDurhamNorth CarolinaUSA
- Division of Rheumatology, Department of MedicineDuke University, School of Medicine, Duke UniversityDurhamNorth CarolinaUSA
| | - Timothy M. Griffin
- Aging and Metabolism Research ProgramOklahoma Medical Research FoundationOklahoma CityOklahomaUSA
- Oklahoma Center for GeroscienceUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Department of Biochemistry and Molecular BiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Veterans Affairs Medical CenterOklahoma CityOklahomaUSA
| |
Collapse
|
17
|
Zhou K, Yang C, Shi K, Liu Y, Hu D, He X, Yang Y, Chu B, Peng J, Zhou Z, Qian Z. Activated macrophage membrane-coated nanoparticles relieve osteoarthritis-induced synovitis and joint damage. Biomaterials 2023; 295:122036. [PMID: 36804660 DOI: 10.1016/j.biomaterials.2023.122036] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/27/2023] [Accepted: 02/03/2023] [Indexed: 02/13/2023]
Abstract
Osteoarthritis (OA) is a common joint condition that is a leading cause of disability worldwide. There are currently no disease-modifying treatments for osteoarthritis, which is associated with multiple kinds of inflammatory cytokines produced by M1 macrophages in the synovium of the joint. Despite recent therapeutic advancements with anti-cytokine biologics, the OA therapy response rate continues to be inadequate. To treat OA, the pro-inflammatory and anti-inflammatory responses of synoviocytes and macrophages must be controlled simultaneously. Therefore, the immune regulation capabilities of an ideal nano-drug should not only minimize pro-inflammatory responses but also effectively boost anti-inflammatory responses. In this paper, an M2H@RPK nanotherapeutic system was developed, KAFAK and shRNA-LEPR were condensed with polyethylenimine (PEI) to form a complex, which was then modified with hyaluronic acid (HA) to negatively charge to cover the M2 membrane. It was discovered that the repolarization of macrophages from the M1 to the M2 phenotype lowered pro-inflammatory responses while enhancing anti-inflammatory responses in macrophages and synoviocytes. In vitro and in vivo studies demonstrate that M2H@RPK dramatically decreases proinflammatory cytokines, controls synovial inflammation, and provides significant therapeutic efficacy by reducing joint damage. Overall, it has been demonstrated that M2H@RPK provides inflammation-targeted therapy by macrophage repolarization, and it represents a promising OA therapeutic strategy.
Collapse
Affiliation(s)
- Kai Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China; Department of Orthopedics, Orthopedic Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Chengli Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China; Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yue Liu
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Danrong Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Xinlong He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yun Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Bingyang Chu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Jinrong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Zongke Zhou
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China.
| |
Collapse
|
18
|
Fleischer MM, Hartner SE, Newton MD, Baker KC, Maerz T. Early patellofemoral cartilage and bone pathology in a rat model of noninvasive anterior cruciate ligament rupture. Connect Tissue Res 2023; 64:175-185. [PMID: 36318110 DOI: 10.1080/03008207.2022.2136571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Anterior cruciate ligament rupture (ACLR) is a risk factor for the development of post-traumatic osteoarthritis (PTOA). While PTOA in the tibiofemoral joint compartment is well-characterized, very little is known about pathology in the patellofemoral compartment after ACL injury. Here, we evaluated the extent to which ACLR induces early patellofemoral joint damage in a rat model. METHODS Adult female Lewis rats were randomized to noninvasive ACLR or Sham. Two weeks post-injury, contrast-enhanced micro-computed tomography (µCT) of femoral and patellar cartilage was performed using 20% v/v ioxaglate. Morphometric parameters of femoral trochlear and patellar cartilage, subchondral bone, and trabecular bone were derived from µCT. Sagittal Safranin-O/Fast-Green-stained histologic sections were graded using the OARSI score in a blinded fashion. RESULTS Cartilage and bone remodelling consistent with an early PTOA phenotype were observed in both femoral trochleas and patellae. ACLR caused osteophyte formation of the patella and pathology in the superficial zone of articular cartilage, including surface fibrillation, fissures, increased cellularity, and abnormal chondrocyte clustering. There were significant increases in thickness of patellar and trochlear cartilage. Loss of subchondral bone thickness, bone volume fraction, and tissue mineral density, as well as changes to patellar and trochlear trabecular microarchitecture, were indicative of catabolic bone remodelling. Several injury-induced changes, including increased cartilage thickness and trabecular spacing and decreased trabecular number were more severe in the patella compared to the trochlea. CONCLUSION The patellofemoral joint develops mild but evident pathology in the early period following ACL rupture, extending the utility of this model to the study of patellofemoral PTOA.
Collapse
Affiliation(s)
| | | | - Michael D Newton
- Department of Orthopaedic Surgery, Beaumont Health, Royal Oak, MI, USA
| | - Kevin C Baker
- Department of Orthopaedic Surgery, Beaumont Health, Royal Oak, MI, USA
- Bone & Joint Center, Department of Orthopaedic Surgery, Henry Ford Health System, Detroit, MI, USA
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Huang L, Jin M, Gu R, Xiao K, Lu M, Huo X, Sun M, Yang Z, Wang Z, Zhang W, Zhi L, Meng Z, Ma J, Ma J, Zhang R. miR-199a-5p Reduces Chondrocyte Hypertrophy and Attenuates Osteoarthritis Progression via the Indian Hedgehog Signal Pathway. J Clin Med 2023; 12:jcm12041313. [PMID: 36835852 PMCID: PMC9959662 DOI: 10.3390/jcm12041313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Osteoarthritis (OA), the most common type of arthritis, is an age-associated disease, characterized by the progressive degradation of articular cartilage, synovial inflammation, and degeneration of subchondral bone. Chondrocyte proliferation is regulated by the Indian hedgehog (IHH in humans, Ihh in animals) signaling molecule, which regulates hypertrophy and endochondral ossification in the development of the skeletal system. microRNAs (miRNAs, miRs) are a family of about 22-nucleotide endogenous non-coding RNAs, which negatively regulate gene expression. In this study, the expression level of IHH was upregulated in the damaged articular cartilage tissues among OA patients and OA cell cultures, while that of miR-199a-5p was the opposite. Further investigations demonstrated that miR-199a-5p could directly regulate IHH expression and reduce chondrocyte hypertrophy and matrix degradation via the IHH signal pathway in the primary human chondrocytes. The intra-articular injection of synthetic miR-199a-5p agomir attenuated OA symptoms in rats, including the alleviation of articular cartilage destruction, subchondral bone degradation, and synovial inflammation. The miR-199a-5p agomir could also inhibit the Ihh signaling pathway in vivo. This study might help in understanding the role of miR-199a-5p in the pathophysiology and molecular mechanisms of OA and indicate a potential novel therapeutic strategy for OA patients.
Collapse
Affiliation(s)
- Lei Huang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Meng Jin
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Ruiying Gu
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Kunlin Xiao
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Mengnan Lu
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Xinyu Huo
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Mengyao Sun
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Zhi Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Zhiyuan Wang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Weijie Zhang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Liqiang Zhi
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Ziang Meng
- Department of Mathematics and Computing Science, Simon Fraser University, Vancouver, BC V6B 5K3, Canada
| | - Jie Ma
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Jianbing Ma
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Correspondence: (J.M.); (R.Z.)
| | - Rui Zhang
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Correspondence: (J.M.); (R.Z.)
| |
Collapse
|
20
|
Knights AJ, Farrell EC, Ellis OM, Lammlin L, Junginger LM, Rzeczycki PM, Bergman RF, Pervez R, Cruz M, Knight E, Farmer D, Samani AA, Wu CL, Hankenson KD, Maerz T. Synovial fibroblasts assume distinct functional identities and secrete R-spondin 2 in osteoarthritis. Ann Rheum Dis 2023; 82:272-282. [PMID: 36175067 PMCID: PMC9972892 DOI: 10.1136/ard-2022-222773] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/22/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Synovium is acutely affected following joint trauma and contributes to post-traumatic osteoarthritis (PTOA) progression. Little is known about discrete cell types and molecular mechanisms in PTOA synovium. We aimed to describe synovial cell populations and their dynamics in PTOA, with a focus on fibroblasts. We also sought to define mechanisms of synovial Wnt/β-catenin signalling, given its emerging importance in arthritis. METHODS We subjected mice to non-invasive anterior cruciate ligament rupture as a model of human joint injury. We performed single-cell RNA-sequencing to assess synovial cell populations, subjected Wnt-GFP reporter mice to joint injury to study Wnt-active cells, and performed intra-articular injections of the Wnt agonist R-spondin 2 (Rspo2) to assess whether gain of function induced pathologies characteristic of PTOA. Lastly, we used cultured fibroblasts, macrophages and chondrocytes to study how Rspo2 orchestrates crosstalk between joint cell types. RESULTS We uncovered seven distinct functional subsets of synovial fibroblasts in healthy and injured synovium, and defined their temporal dynamics in early and established PTOA. Wnt/β-catenin signalling was overactive in PTOA synovium, and Rspo2 was strongly induced after injury and secreted exclusively by Prg4hi lining fibroblasts. Trajectory analyses predicted that Prg4hi lining fibroblasts arise from a pool of Dpp4+ mesenchymal progenitors in synovium, with SOX5 identified as a potential regulator of this emergence. We also showed that Rspo2 orchestrated pathological crosstalk between synovial fibroblasts, macrophages and chondrocytes. CONCLUSIONS Synovial fibroblasts assume distinct functional identities during PTOA in mice, and Prg4hi lining fibroblasts secrete Rspo2 that may drive pathological joint crosstalk after injury.
Collapse
Affiliation(s)
- Alexander J. Knights
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Easton C. Farrell
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Olivia M. Ellis
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lindsey Lammlin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lucas M. Junginger
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Phillip M. Rzeczycki
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Rachel F. Bergman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Rida Pervez
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Monique Cruz
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Eleanor Knight
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Dennis Farmer
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Alexa A. Samani
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Chia-Lung Wu
- Department of Orthopaedic Surgery and Rehabilitation, Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| | - Kurt D. Hankenson
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Tristan Maerz
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan, USA .,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
21
|
Chandrabalan A, Firth A, Litchfield RB, Appleton CT, Getgood A, Ramachandran R. Human osteoarthritis knee joint synovial fluids cleave and activate Proteinase-Activated Receptor (PAR) mediated signaling. Sci Rep 2023; 13:1124. [PMID: 36670151 PMCID: PMC9859807 DOI: 10.1038/s41598-023-28068-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disorder with increasing worldwide incidence. Mechanistic insights into OA pathophysiology are evolving and there are currently no disease-modifying OA drugs. An increase in protease activity is linked to progressive degradation of the cartilage in OA. Proteases also trigger inflammation through a family of G protein-coupled receptors (GPCRs) called the Proteinase-Activated Receptors (PARs). PAR signaling can trigger pro-inflammatory responses and targeting PARs is proposed as a therapeutic approach in OA. Several enzymes can cleave the PAR N-terminus, but the endogenous protease activators of PARs in OA remain unclear. Here we characterized PAR activating enzymes in knee joint synovial fluids from OA patients and healthy donors using genetically encoded PAR biosensor expressing cells. Calcium signaling assays were performed to examine receptor activation. The class and type of enzymes cleaving the PARs was further characterized using protease inhibitors and fluorogenic substrates. We find that PAR1, PAR2 and PAR4 activating enzymes are present in knee joint synovial fluids from healthy controls and OA patients. Compared to healthy controls, PAR1 activating enzymes are elevated in OA synovial fluids while PAR4 activating enzyme levels are decreased. Using enzyme class and type selective inhibitors and fluorogenic substrates we find that multiple PAR activating enzymes are present in OA joint fluids and identify serine proteinases (thrombin and trypsin-like) and matrix metalloproteinases as the major classes of PAR activating enzymes in the OA synovial fluids. Synovial fluid driven increase in calcium signaling was significantly reduced in cells treated with PAR1 and PAR2 antagonists, but not in PAR4 antagonist treated cells. OA associated elevation of PAR1 cleavage suggests that targeting this receptor may be beneficial in the treatment of OA.
Collapse
Affiliation(s)
- Arundhasa Chandrabalan
- Department of Physiology and Pharmacology, Bone and Joint Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Andrew Firth
- Division of Orthopedic Surgery, Bone and Joint Institute, Fowler Kennedy Sport Medicine Clinic, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Robert B Litchfield
- Division of Orthopedic Surgery, Bone and Joint Institute, Fowler Kennedy Sport Medicine Clinic, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - C Thomas Appleton
- Department of Physiology and Pharmacology, Bone and Joint Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.,Department of Medicine, Bone and Joint Institute, Schulich School of Medicine and Dentistry, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, London, ON, Canada
| | - Alan Getgood
- Division of Orthopedic Surgery, Bone and Joint Institute, Fowler Kennedy Sport Medicine Clinic, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Bone and Joint Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
| |
Collapse
|
22
|
Sophocleous A, Azfer A, Huesa C, Stylianou E, Ralston SH. Probiotics Inhibit Cartilage Damage and Progression of Osteoarthritis in Mice. Calcif Tissue Int 2023; 112:66-73. [PMID: 36261653 PMCID: PMC9813193 DOI: 10.1007/s00223-022-01030-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/19/2022] [Indexed: 01/09/2023]
Abstract
Increasing interest has focussed on the possible role of alterations in the microbiome in the pathogenesis of metabolic disease, inflammatory disease, and osteoporosis. Here we examined the role of the microbiome in a preclinical model of osteoarthritis in mice subjected to destabilisation of medical meniscus (DMM). The intestinal microbiome was depleted by broad-spectrum antibiotics from 1 week before birth until the age of 6 weeks when mice were subjected reconstitution of the microbiome with faecal microbial transplant (FMT) followed by the administration of a mixture of probiotic strains Lacticaseibacillus paracasei 8700:2, Lactiplantibacillus plantarum HEAL9 and L. plantarum HEAL19 or vehicle. All mice were subjected to DMM at the age of 8 weeks. The severity of osteoarthritis was evaluated by histological analysis and effects on subchondral bone were investigated by microCT analyses. The combination of FMT and probiotics significantly inhibited cartilage damage at the medial femoral condyle such that the OARSI score was 4.64 ± 0.32 (mean ± sem) in the FMT and probiotic group compared with 6.48 ± 0.53 in the FMT and vehicle group (p = 0.007). MicroCT analysis of epiphyseal bone from the femoral condyle showed that the probiotic group had higher BV/TV, increased Tb.Th, and moderately thicker subchondral bone plates than the control group. There was no difference between groups in joint inflammation or in serum concentrations of inflammatory cytokines and chemokines. We conclude that treatment with probiotics following FMT in mice where the microbiome has been depleted inhibits DMM-induced cartilage damage and impacts on the structure of subchondral bone particularly at the femoral condyle. While further studies are required to elucidate the mechanism of action, our research suggests that these probiotics may represent a novel intervention for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Antonia Sophocleous
- Department of Life Sciences, School of Sciences, European University of Cyprus, Nicosia, Cyprus
- Rheumatology and Bone Diseases Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh, UK
| | - Asim Azfer
- Rheumatology and Bone Diseases Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh, UK
| | - Carmen Huesa
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Eleni Stylianou
- Department of Life Sciences, School of Sciences, European University of Cyprus, Nicosia, Cyprus
| | - Stuart H Ralston
- Rheumatology and Bone Diseases Unit, Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
23
|
Ozkan H, Di Francesco M, Willcockson H, Valdés-Fernández J, Di Francesco V, Granero-Moltó F, Prósper F, Decuzzi P, Longobardi L. Sustained inhibition of CC-chemokine receptor-2 via intraarticular deposition of polymeric microplates in post-traumatic osteoarthritis. Drug Deliv Transl Res 2023; 13:689-701. [PMID: 36109442 PMCID: PMC9794532 DOI: 10.1007/s13346-022-01235-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2022] [Indexed: 12/31/2022]
Abstract
Posttraumatic osteoarthritis (PTOA) is mostly treated via corticosteroid administration, and total joint arthroplasty continues to be the sole effective intervention in severe conditions. To assess the therapeutic potential of CCR2 targeting in PTOA, we used biodegradable microplates (µPLs) to achieve a slow and sustained intraarticular release of the CCR2 inhibitor RS504393 into injured knees and followed joint damage during disease progression. RS504393-loaded µPLs (RS-µPLs) were fabricated via a template-replica molding technique. A mixture of poly(lactic-co-glycolic acid) (PLGA) and RS504393 was deposited into 20 × 10 μm (length × height) wells in a polyvinyl alcohol (PVA) square-patterned template. After physicochemical and toxicological characterizations, the RS504393 release profile from µPL was assessed in PBS buffer. C57BL/6 J male mice were subjected to destabilization of the medial meniscus (DMM)/sham surgery, and RS-µPLs (1 mg/kg) were administered intraarticularly 1 week postsurgery. Administrations were repeated at 4 and 7 weeks post-DMM. Drug free-µPLs (DF-µPLs) and saline injections were performed as controls. Mice were euthanized at 4 and 10 weeks post-DMM, corresponding to the early and severe PTOA stages, respectively. Knees were evaluated for cartilage structure score (ACS, H&E), matrix loss (safranin O score), osteophyte formation and maturation from cartilage to bone (cartilage quantification), and subchondral plate thickness. The RS-µPL architecture ensured the sustained release of CCR2 inhibitors over several weeks, with ~ 20% of RS504393 still available at 21 days. This prolonged release improved cartilage structure and reduced bone damage and synovial hyperplasia at both PTOA stages. Extracellular matrix loss was also attenuated, although with less efficacy. The results indicate that local sustained delivery is needed to optimize CCR2-targeted therapies.
Collapse
Affiliation(s)
- Huseyin Ozkan
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-, Chapel Hill, 3300 Thurston Bowels Bldg, Campus, Box 7280, Chapel Hill, NC 27599 USA
| | - Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano Di Tecnologia, Genoa, Italy
| | - Helen Willcockson
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-, Chapel Hill, 3300 Thurston Bowels Bldg, Campus, Box 7280, Chapel Hill, NC 27599 USA
| | - José Valdés-Fernández
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-, Chapel Hill, 3300 Thurston Bowels Bldg, Campus, Box 7280, Chapel Hill, NC 27599 USA ,Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain
| | - Valentina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano Di Tecnologia, Genoa, Italy
| | - Froilán Granero-Moltó
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain ,Department of Orthopedic Surgery and Traumatology, Clínica Universidad de Navarra, Pamplona, Spain ,Program of Regenerative Medicine, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain ,Instituto de Investigacion Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Felipe Prósper
- Cell Therapy Area, Clínica Universidad de Navarra, Pamplona, Spain ,Program of Regenerative Medicine, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain ,Instituto de Investigacion Sanitaria de Navarra (IdiSNA), Pamplona, Spain ,Department of Hematology, Clínica Universidad de Navarra, Pamplona, Spain ,Program of Hemato-Oncology, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano Di Tecnologia, Genoa, Italy
| | - Lara Longobardi
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-, Chapel Hill, 3300 Thurston Bowels Bldg, Campus, Box 7280, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
24
|
Huesa C, Dunning L, MacDougall K, Fegen M, Ortiz A, McCulloch K, McGrath S, Litherland GJ, Crilly A, Van ‘T Hof RJ, Ferrell WR, Goodyear CS, Lockhart JC. Moderate exercise protects against joint disease in a murine model of osteoarthritis. Front Physiol 2022; 13:1065278. [PMID: 36545287 PMCID: PMC9760924 DOI: 10.3389/fphys.2022.1065278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/16/2022] [Indexed: 12/07/2022] Open
Abstract
Exercise is recommended as a non-pharmacological therapy for osteoarthritis (OA). Various exercise regimes, with differing intensities and duration, have been used in a range of OA rodent models. These studies show gentle or moderate exercise reduces the severity of OA parameters while high intensity load bearing exercise is detrimental. However, these studies were largely conducted in rats or in mouse models induced by severe injury, age or obesity, whilst destabilization of the medial meniscus (DMM) in mice has become a widely accepted model due to its lower variability, moderate progression and timescale. The present study was undertaken to provide insight into the effect of moderate exercise on early joint pathology in the DMM mouse model. Exercise was induced a week after induction by forced wheel walking for three or 7 weeks. Joints were analyzed by microcomputed tomography and histology. Assessment of skeletal parameters revealed that exercise offered protection against cartilage damage after 7 weeks of exercise, and a temporary protection against osteosclerosis was displayed after 3 weeks of exercise. Furthermore, exercise modified the metaphyseal trabecular microarchitecture of the osteoarthritic leg in both time points examined. Collectively, our findings corroborate previous studies showing that exercise has an important effect on bone in OA, which subsequently, at 8 weeks post-induction, translates into less cartilage damage. Thus, providing an exercise protocol in a surgical mouse model of OA, which can be used in the future to further dissect the mechanisms by which moderate exercise ameliorates OA.
Collapse
Affiliation(s)
- C. Huesa
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom,School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom
| | - L. Dunning
- School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom
| | - K. MacDougall
- School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom
| | - M. Fegen
- School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom
| | - A. Ortiz
- School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom
| | - K. McCulloch
- School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom
| | - S. McGrath
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - G. J. Litherland
- School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom
| | - A. Crilly
- School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom
| | - R. J. Van ‘T Hof
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - W. R. Ferrell
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - C. S. Goodyear
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom,*Correspondence: C. S. Goodyear, ; J. C. Lockhart,
| | - J. C. Lockhart
- School of Health and Life Sciences, University of the West of Scotland, Paisley, United Kingdom,*Correspondence: C. S. Goodyear, ; J. C. Lockhart,
| |
Collapse
|
25
|
Willcockson H, Ozkan H, Arbeeva L, Mucahit E, Musawwir L, Longobardi L. Early ablation of Ccr2 in aggrecan-expressing cells following knee injury ameliorates joint damage and pain during post-traumatic osteoarthritis. Osteoarthritis Cartilage 2022; 30:1616-1630. [PMID: 36075514 PMCID: PMC9671864 DOI: 10.1016/j.joca.2022.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 08/10/2022] [Accepted: 08/26/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate whether Ccr2 inactivation in aggrecan-expressing cells induced before post-traumatic OA (PTOA) onset or during progression, improves joint structures, synovial thickness and pain. DESIGN We induced a Ccr2 deletion in aggrecan-expressing cells (CCR2-AggKO) in skeletally mature mice using a tamoxifen-inducible Ccr2 inactivation. We stimulated PTOA changes (destabilization of medial meniscus, DMM) in CCR2-AggKO and CCR2+/+ mice, inducing recombination before DMM or 4 wks after DMM (early-vs late-inactivation). Joint damage was evaluated 2, 4, 8, 12 wks post-DMM using multiple scores: articular-cartilage structure (ACS), Safranin-O, histomorphometry, osteophyte size/maturity, subchondral bone thickness and synovial hyperplasia. Spontaneous (incapacitance meter) and evoked pain (von-Frey filaments) were assessed up to 20 wks. RESULTS Early aggrecan-Ccr2 inactivation in CCR2-AggKO mice (N=8) resulted in improved ACS score (8-12wk, P=0.002), AC area (4-12wk, P<0.05) and Saf-O score (2wks P=0.004, 4wks P=0.02, 8-12wks P=0.002) compared to CCR2+/+. Increased subchondral bone thickness was delayed only at 2 wks and exclusively following early recombination. Osteophyte size was not affected, but osteophyte maturation (cartilage-to-bone) was delayed (4wks P=0.04; 8 wks P=0.03). Although late aggrecan-Ccr2 deletion led to some cartilage improvement, most data did not reach statistical significance; osteophyte maturity was delayed at 12wks. Early aggrecan-Ccr2 deletion led to improved pain measures of weight bearing compared to CCR2+/+ mice (N = 9, 12wks diff 0.13 [0.01, 0.26], 16wks diff 0.15 [0.05, 0.26], 20wks diff 0.23 [0.14, 0.31]). Improved mechanosensitivity in evoked pain, although less noticeable, was detected. CONCLUSIONS We demonstrated that deletion of Ccr2 in aggrecan expressing cells reduces the initiation but not progression of OA.
Collapse
Affiliation(s)
- H Willcockson
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - H Ozkan
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - L Arbeeva
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - E Mucahit
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - L Musawwir
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - L Longobardi
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| |
Collapse
|
26
|
Little CB, Zaki S, Blaker CL, Clarke EC. Animal models of osteoarthritis. Bone Joint Res 2022; 11:514-517. [PMID: 35909339 PMCID: PMC9396918 DOI: 10.1302/2046-3758.118.bjr-2022-0217.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Cite this article: Bone Joint Res 2022;11(8):514–517.
Collapse
Affiliation(s)
- Christopher B. Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
| | - Sanaa Zaki
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Sydney, Australia
| | - Carina L. Blaker
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
| | - Elizabeth C. Clarke
- Murray Maxwell Biomechanics Laboratory, Kolling Institute, Faculty of Medicine and Health, University of Sydney and Northern Sydney Local Health District, Sydney, Australia
| |
Collapse
|
27
|
Zhuo X, Wu Y, Fu X, Liang X, Xiang Y, Li J, Mao C, Jiang Y. The Yin‐Yang roles of protease‐activated receptors in inflammatory signalling and diseases. FEBS J 2022; 289:4000-4020. [DOI: 10.1111/febs.16406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 01/26/2022] [Accepted: 02/15/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Xin Zhuo
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Yue Wu
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Xiujuan Fu
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Xiaoyu Liang
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Yuxin Xiang
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Jianbin Li
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Canquan Mao
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| | - Yuhong Jiang
- School of Life Science and Engineering Southwest Jiaotong University Chengdu China
| |
Collapse
|
28
|
Development and characterization of a humanized mouse model of osteoarthritis. Osteoarthritis Cartilage 2022; 30:875-885. [PMID: 35307533 DOI: 10.1016/j.joca.2022.02.620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 02/01/2022] [Accepted: 02/05/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE In light of the role of immune cells in OA pathogenesis, the development of sophisticated animal models closely mimicking the immune dysregulation during the disease development and progression could be instrumental for the preclinical evaluation of novel treatments. Among these models, immunologically humanized mice may represent a relevant system, particularly for testing immune-interacting DMOADs or cell therapies before their transfer to the clinic. Our objective, therefore, was to develop an experimental model of OA by destabilization of the medial meniscus (DMM) in humanized mice. METHOD Irradiated 5-week-old NOD/LtSz-scid IL2Rγnull (NSG) mice were humanized by intravenous injection of CD34+ human hematopoietic stem cells. The engraftment efficiency was evaluated by flow cytometry 17 weeks after the humanization procedure. Humanized and non-humanized NSG mice underwent DMM or sham surgery and OA development was assessed 1, 6, and 12 weeks after the surgery. RESULTS 120 days after the humanization, human T and B lymphocytes, macrophages and NK cells, were present in the blood and spleen of the humanized NSG mice. The DMM surgery induced articular cartilage and meniscal alterations associated with an increase in OA and the meniscal score. Moreover, the surgery triggered an inflammatory response that was sustained at a low grade in the DMM group. CONCLUSIONS Our study shows for the first time the feasibility of inducing OA by DMM in humanized mice. This novel OA model could constitute a useful tool to bridge the gap between the preclinical and clinical evaluation of immune interacting DMOADs and cell-based therapies.
Collapse
|
29
|
Wesdorp MA, Capar S, Bastiaansen-Jenniskens YM, Kops N, Creemers LB, Verhaar JA, Van Osch GJ, Wei W. Intra-articular Administration of Triamcinolone Acetonide in a Murine Cartilage Defect Model Reduces Inflammation but Inhibits Endogenous Cartilage Repair. Am J Sports Med 2022; 50:1668-1678. [PMID: 35315287 PMCID: PMC9069659 DOI: 10.1177/03635465221083693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Cartilage defects result in joint inflammation. The presence of proinflammatory factors has been described to negatively affect cartilage formation. PURPOSE To evaluate the effect and timing of administration of triamcinolone acetonide (TAA), an anti-inflammatory drug, on cartilage repair using a mouse model. STUDY DESIGN Controlled laboratory study. METHODS A full-thickness cartilage defect was created in the trochlear groove of 10-week-old male DBA/1 mice (N = 80). Mice received an intra-articular injection of TAA or saline on day 1 or 7 after induction of the defect. Mice were euthanized on days 10 and 28 for histological evaluation of cartilage defect repair, synovial inflammation, and synovial membrane thickness. RESULTS Mice injected with TAA had significantly less synovial inflammation at day 10 than saline-injected mice independent of the time of administration. At day 28, the levels of synovitis dropped toward healthy levels; nevertheless, the synovial membrane was thinner in TAA- than in saline-injected mice, reaching statistical significance in animals injected on day 1 (70.1 ± 31.9 µm vs 111.9 ± 30.9 µm, respectively; P = .01) but not in animals injected on day 7 (68.2 ± 21.86 µm vs 90.2 ± 21.29 µm, respectively; P = .26). A thinner synovial membrane was moderately associated with less filling of the defect after 10 and 28 days (r = 0.42, P = .02; r = 0.47, P = .01, respectively). Whereas 10 days after surgery there was no difference in the area of the defect filled and the cell density in the defect area between saline- and TAA-injected knees, filling of the defect at day 28 was lower in TAA- than in saline-injected knees for both injection time points (day 1 injection, P = .04; day 7 injection, P = .01). Moreover, there was less collagen type 2 staining in the filled defect area in TAA- than in saline-injected knees after 28 days, reaching statistical significance in day 1-injected knees (2.6% vs 18.5%, respectively; P = .01) but not in day 7-injected knees (7.4% vs 15.8%, respectively; P = .27). CONCLUSION Intra-articular injection of TAA reduced synovial inflammation but negatively affected cartilage repair. This implies that inhibition of inflammation may inhibit cartilage repair or that TAA has a direct negative effect on cartilage formation. CLINICAL RELEVANCE Our findings show that TAA can inhibit cartilage defect repair. Therefore, we suggest not using TAA to reduce inflammation in a cartilage repair setting.
Collapse
Affiliation(s)
- Marinus A. Wesdorp
- Department of Orthopaedic Surgery and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Serdar Capar
- Department of Orthopaedic Surgery and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | | | - Nicole Kops
- Department of Orthopaedic Surgery and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Laura B. Creemers
- Department of Orthopedic Surgery, UMC Utrecht, University Medical Center, Utrecht, the Netherlands
| | - Jan A.N. Verhaar
- Department of Orthopaedic Surgery and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Gerjo J.V.M. Van Osch
- Department of Orthopaedic Surgery and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Delft, the Netherlands,Gerjo J.V.M. Van Osch, PhD, Department of Orthopaedic Surgery and Sports Medicine and Department of Otorhinolaryngology, Erasmus MC, University Medical Center, Room Ee16.55c, Dr Molewaterplein 40, Rotterdam, 3015 GD, the Netherlands ()
| | - Wu Wei
- Department of Orthopaedic Surgery and Sports Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands,Department of Orthopedic Surgery, Elisabeth-Tweesteden Ziekenhuis, Tilburg, the Netherlands
| |
Collapse
|
30
|
Clement-Lacroix P, Little CB, Smith MM, Cottereaux C, Merciris D, Meurisse S, Mollat P, Touitou R, Brebion F, Gosmini R, De Ceuninck F, Botez I, Lepescheux L, van der Aar E, Christophe T, Vandervoort N, Blanqué R, Comas D, Deprez P, Amantini D. Pharmacological characterization of GLPG1972/S201086, a potent and selective small-molecule inhibitor of ADAMTS5. Osteoarthritis Cartilage 2022; 30:291-301. [PMID: 34626798 DOI: 10.1016/j.joca.2021.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 07/29/2021] [Accepted: 08/09/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) is a key enzyme in degradation of cartilage in osteoarthritis (OA). We report the pharmacological characterization of GLPG1972/S201086, a new, potent and selective small-molecule ADAMTS5 inhibitor. METHODS Potency and selectivity of GLPG1972/S201086 for ADAMTS5 were determined using fluorescently labeled peptide substrates. Inhibitory effects of GLPG1972/S201086 on interleukin-1α-stimulated glycosaminoglycan release in mouse femoral head cartilage explants and on interleukin-1β-stimulated release of an ADAMTS5-derived aggrecan neoepitope (quantified with ELISA) in human articular cartilage explants were determined. In the destabilization of the medial meniscus (DMM) mouse and menisectomized (MNX) rat models, effects of oral GLPG1972/S201086 on relevant OA histological and histomorphometric parameters were evaluated. RESULTS GLPG1972/S201086 inhibited human and rat ADAMTS5 (IC50 ± SD: 19 ± 2 nM and <23 ± 1 nM, respectively), with 8-fold selectivity over ADAMTS4, and 60->5,000-fold selectivity over other related proteases in humans. GLPG1972/S201086 dose-dependently inhibited cytokine-stimulated aggrenolysis in mouse and human cartilage explants (100% at 20 μM and 10 μM, respectively). In DMM mice, GLPG1972/S201086 (30-120 mg/kg b.i.d) vs vehicle reduced femorotibial cartilage proteoglycan loss (23-37%), cartilage structural damage (23-39%) and subchondral bone sclerosis (21-36%). In MNX rats, GLPG1972/S201086 (10-50 mg/kg b.i.d) vs vehicle reduced cartilage damage (OARSI score reduction, 6-23%), and decreased proteoglycan loss (∼27%) and subchondral bone sclerosis (77-110%). CONCLUSIONS GLPG1972/S201086 is a potent, selective and orally available ADAMTS5 inhibitor, demonstrating significant protective efficacy on both cartilage and subchondral bone in two relevant in vivo preclinical OA models.
Collapse
Affiliation(s)
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratories, University of Sydney, Kolling Institute, Northern Sydney Local Health District, Royal North Shore Hospital, St Leonards, NSW, Australia.
| | - M M Smith
- Raymond Purves Bone and Joint Research Laboratories, University of Sydney, Kolling Institute, Northern Sydney Local Health District, Royal North Shore Hospital, St Leonards, NSW, Australia.
| | | | | | | | - P Mollat
- Galapagos SASU, Romainville, France.
| | - R Touitou
- Galapagos SASU, Romainville, France.
| | - F Brebion
- Galapagos SASU, Romainville, France.
| | - R Gosmini
- Galapagos SASU, Romainville, France.
| | | | - I Botez
- Institut de Recherches Servier, France.
| | | | | | | | | | - R Blanqué
- Galapagos SASU, Romainville, France.
| | - D Comas
- Galapagos SASU, Romainville, France.
| | - P Deprez
- Galapagos SASU, Romainville, France.
| | | |
Collapse
|
31
|
Wang F, Liu M, Wang N, Luo J. G Protein-Coupled Receptors in Osteoarthritis. Front Endocrinol (Lausanne) 2022; 12:808835. [PMID: 35154008 PMCID: PMC8831737 DOI: 10.3389/fendo.2021.808835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is the most common chronic joint disease characterized, for which there are no available therapies being able to modify the progression of OA and prevent long-term disability. Critical roles of G-protein coupled receptors (GPCRs) have been established in OA cartilage degeneration, subchondral bone sclerosis and chronic pain. In this review, we describe the pathophysiological processes targeted by GPCRs in OA, along with related preclinical model and/or clinical trial data. We review examples of GPCRs which may offer attractive therapeutic strategies for OA, including receptors for cannabinoids, hormones, prostaglandins, fatty acids, adenosines, chemokines, and discuss the main challenges for developing these therapies.
Collapse
Affiliation(s)
- Fanhua Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Ning Wang
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, United Kingdom
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
32
|
Rzeczycki P, Rasner C, Lammlin L, Junginger L, Goldman S, Bergman R, Redding S, Knights AJ, Elliott M, Maerz T. Cannabinoid receptor type 2 is upregulated in synovium following joint injury and mediates anti-inflammatory effects in synovial fibroblasts and macrophages. Osteoarthritis Cartilage 2021; 29:1720-1731. [PMID: 34537380 PMCID: PMC8883578 DOI: 10.1016/j.joca.2021.09.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 08/09/2021] [Accepted: 09/06/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Joint injury-induced perturbations to the endocannabinoid system (ECS), a regulator of both inflammation and nociception, remain largely uncharacterized. We employed a mouse model of ACL rupture to assess alterations to nociception, inflammation, and the ECS while using in vitro models to determine whether CB2 agonism can mitigate inflammatory signaling in macrophages and fibroblast-like synoviocytes (FLS). DESIGN Mice underwent noninvasive ACL rupture (ACLR) via tibial compression-based loading. Nociception was measured longitudinally using mechanical allodynia and knee hyperalgesia testing. Synovitis was assessed using histological scoring and histomorphometry. Gene and protein markers of inflammation were characterized in whole joints and synovium. Immunohistochemistry assessed injury-induced alterations to CB1+, CB2+, and F4/80+ cells in synovium. To assess whether CB2 agonism can inhibit pro-inflammatory macrophage polarization, murine bone marrow-derived macrophages (mBMDM) were stimulated with IL-1β or conditioned medium from IL-1β-treated FLS and treated with vehicle (DMSO), the CB2 agonist HU308, or cannabidiol (CBD). Macrophage polarization was assessed as the ratio of M1-associated (IL1b, MMP1b, and IL6) to M2-associated (IL10, IL4, and CD206) gene expression. Human FLS (hFLS) isolated from synovial tissue of OA patients were treated with vehicle (DMSO) or HU308 following TNF-α or IL-1β stimulation to assess inhibition of catabolic/inflammatory gene expression. RESULTS ACLR induces synovitis, progressively-worsening PTOA severity, and an immediate and sustained increase in both mechanical allodynia and knee hyperalgesia, which persist beyond the resolution of molecular inflammation. Enrichment of CB2, but not CB1, was observed in ACLR synovium at 3d, 14d, and 28d, and CB2 was found to be associated with F4/80 (+) cells, which are increased in number in ACLR synovium at all time points. The CB2 agonist HU308 strongly inhibited mBMDM M1-type polarization following stimulation with either IL-1β or conditioned medium from IL-1β-treated mFLS, which was characterized by reductions in Il1b, Mmp1b, and Il6 and increases in Cd206 gene expression. Cannabidiol similarly inhibited IL-1β-induced mBMDM M1 polarization via a reduction in Il1b and an increase in Cd206 and Il4 gene expression. Lastly, in OA hFLS, HU308 treatment inhibited IL-1β-induced CCL2, MMP1, MMP3, and IL6 expression and further inhibited TNF-α-induced CCL2, MMP1, and GMCSF expression, demonstrating human OA-relevant anti-inflammatory effects by targeting CB2. CONCLUSIONS Joint injury perturbs the intra-articular ECS, characterized by an increase in synovial F4/80(+) cells, which express CB2, but not CB1. Targeting CB2 in murine macrophages and human FLS induced potent anti-inflammatory and anti-catabolic effects, which indicates that the CB2 receptor plays a key role in regulating inflammatory signaling in the two primary effector cells in the synovium. The intraarticular ECS is therefore a potential therapeutic target for blocking pathological inflammation in future disease-modifying PTOA treatments.
Collapse
Affiliation(s)
- P Rzeczycki
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - C Rasner
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - L Lammlin
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - L Junginger
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - S Goldman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - R Bergman
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - S Redding
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - A J Knights
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - M Elliott
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - T Maerz
- Orthopaedic Research Laboratories, Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
33
|
Haubruck P, Pinto MM, Moradi B, Little CB, Gentek R. Monocytes, Macrophages, and Their Potential Niches in Synovial Joints - Therapeutic Targets in Post-Traumatic Osteoarthritis? Front Immunol 2021; 12:763702. [PMID: 34804052 PMCID: PMC8600114 DOI: 10.3389/fimmu.2021.763702] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Synovial joints are complex structures that enable normal locomotion. Following injury, they undergo a series of changes, including a prevalent inflammatory response. This increases the risk for development of osteoarthritis (OA), the most common joint disorder. In healthy joints, macrophages are the predominant immune cells. They regulate bone turnover, constantly scavenge debris from the joint cavity and, together with synovial fibroblasts, form a protective barrier. Macrophages thus work in concert with the non-hematopoietic stroma. In turn, the stroma provides a scaffold as well as molecular signals for macrophage survival and functional imprinting: “a macrophage niche”. These intricate cellular interactions are susceptible to perturbations like those induced by joint injury. With this review, we explore how the concepts of local tissue niches apply to synovial joints. We introduce the joint micro-anatomy and cellular players, and discuss their potential interactions in healthy joints, with an emphasis on molecular cues underlying their crosstalk and relevance to joint functionality. We then consider how these interactions are perturbed by joint injury and how they may contribute to OA pathogenesis. We conclude by discussing how understanding these changes might help identify novel therapeutic avenues with the potential of restoring joint function and reducing post-traumatic OA risk.
Collapse
Affiliation(s)
- Patrick Haubruck
- Centre for Orthopaedics, Trauma Surgery and Spinal Cord Injury, Trauma and Reconstructive Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Marlene Magalhaes Pinto
- Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Babak Moradi
- Clinic of Orthopaedics and Trauma Surgery, University Clinic of Schleswig-Holstein, Kiel, Germany
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Institute of Bone and Joint Research, Faculty of Medicine and Health University of Sydney, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Rebecca Gentek
- Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
34
|
Yang T, Sun K, Wang C, Swarnkar G, Quan S, Kress D, Xiao J, Alippe Y, Zheng H, Brophy RH, Hao D, McAlinden A, Abu-Amer Y, Shen J, Mbalaviele G. Gasdermin D deficiency attenuates arthritis induced by traumatic injury but not autoantibody-assembled immune complexes. Arthritis Res Ther 2021; 23:286. [PMID: 34784954 PMCID: PMC8594229 DOI: 10.1186/s13075-021-02668-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/31/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gasdermin D (GSDMD) is cleaved by several proteases including by caspase-1, a component of intracellular protein complexes called inflammasomes. Caspase-1 also converts pro-interleukin-1β (pro-IL-1β) and pro-IL-18 into bioactive IL-1β and IL-18, respectively. GSDMD amino-terminal fragments form plasma membrane pores, which mediate the secretion of IL-1β and IL-18 and cause the inflammatory form of cell death pyroptosis. Here, we tested the hypothesis that GSDMD contributes to joint degeneration in the K/BxN serum transfer-induced arthritis (STIA) model in which autoantibodies against glucose-6-phosphate isomerase promote the formation of pathogenic immune complexes on the surface of myeloid cells, which highly express the inflammasomes. The unexpected outcomes with the STIA model prompted us to determine the role of GSDMD in the post-traumatic osteoarthritis (PTOA) model caused by meniscus ligamentous injury (MLI) based on the hypothesis that this pore-forming protein is activated by signals released from damaged joint tissues. METHODS Gsdmd +/+ and Gsdmd-/- mice were injected with K/BxN mouse serum or subjected to MLI to cause STIA or PTOA, respectively. Paw and ankle swelling and DXA scanning were used to assess the outcomes in the STIA model whereas histopathology and micro-computed tomography (μCT) were utilized to monitor joints in the PTOA model. Murine and human joint tissues were also examined for GSDMD, IL-1β, and IL-18 expression by qPCR, immunohistochemistry, or immunoblotting. RESULTS GSDMD levels were higher in serum-inoculated paws compared to PBS-injected paws. Unexpectedly, ablation of GSDMD failed to reduce joint swelling and osteolysis, suggesting that GSDMD was dispensable for the pathogenesis of STIA. GSDMD levels were also higher in MLI compared to sham-operated joints. Importantly, ablation of GSDMD attenuated MLI-associated cartilage degradation (p = 0.0097), synovitis (p = 0.014), subchondral bone sclerosis (p = 0.0006), and subchondral bone plate thickness (p = 0.0174) based on histopathological and μCT analyses. CONCLUSION GSDMD plays a key role in the pathogenesis of PTOA, but not STIA, suggesting that its actions in experimental arthropathy are tissue context-specific.
Collapse
Affiliation(s)
- Tong Yang
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Kai Sun
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Chun Wang
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Gaurav Swarnkar
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Songtao Quan
- Luoyang Orthopedic - Traumatological Hospital of Henan Province, Luoyang, Henan, China
| | - Dustin Kress
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Jianqiu Xiao
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Yael Alippe
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA
| | - Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert H Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Dingjun Hao
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospital for Children, St. Louis, MO, USA
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospital for Children, St. Louis, MO, USA
| | - Jie Shen
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8301, St. Louis, MO, 63110, USA.
| |
Collapse
|
35
|
Zaki S, Smith MM, Little CB. Pathology-pain relationships in different osteoarthritis animal model phenotypes: it matters what you measure, when you measure, and how you got there. Osteoarthritis Cartilage 2021; 29:1448-1461. [PMID: 34332049 DOI: 10.1016/j.joca.2021.03.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine whether osteoarthritis (OA) pain characteristics and mechanistic pathways in pre-clinical models are phenotype-specific. DESIGN Male 11-week-old C57BL6 mice had unilateral medial-meniscal-destabilization (DMM) or antigen-induced-arthritis (AIA), vs sham-surgery/immunised-controls (Sham/Im-CT). Pain behaviour (allodynia, mechanical- and thermal-hyperalgesia, hindlimb static weight-bearing, stride-length) and lumbar dorsal root ganglia (DRG) gene-expression were measured at baseline, day-3, week-1/-2/-4/-8/-16, and pain-behaviour:gene-expression:joint-pathology associations investigated. RESULTS DMM and AIA induced structural OA defined by progressively increasing cartilage erosion, subchondral bone sclerosis and osteophyte size and maturation. All pain-behaviours were modified, with model-specific differences in severity and temporal pattern. Tactile allodynia developed acutely in both models and persisted to week-16. During early-OA (wk4-8) there was; reduced right hindlimb weight-bearing in AIA; thermal-hyperalgesia and reduced stride-length in DMM. During chronic-OA (wk12-16); mechanical-hyperalgesia and reduced right hindlimb weight-bearing were observed in DMM only. There were no associations in either model between different pain-behaviour outcomes. A coordinated DRG-expression profile was observed in sham and Im-CT for all 11 genes tested, but not in AIA and DMM. At wk-16 despite equivalent joint pathology, changes in DRG-expression (Calca, Trpa1, Trpv1, Trpv4) were observed only in DMM. In AIA mechanical-hyperalgesia was associated with Trpv1 (r = -0.79) and Il1b (r = 0.53). In DMM stride-length was associated with Calca, Tac1, Trpv1, Trpv2, Trpv4 and Adamts5 (r = 0.4-0.57). DRG gene-expression change was correlated with subchondral-bone sclerosis in DMM, and cartilage damage in AIA. Positive pain-behaviour:joint-pathology associations were only present in AIA - for synovitis, subchondral-bone resorption, chondrocyte-hypertrophy and cartilage damage. CONCLUSION Pain and peripheral sensory neuronal responses are OA-phenotype-specific with distinct pathology:pain-outcome:molecular-mechanism relationships.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, at Royal North Shore Hospital, Australia.
| | - M M Smith
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, at Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, at Royal North Shore Hospital, Australia.
| |
Collapse
|
36
|
Lucena F, McDougall JJ. Protease Activated Receptors and Arthritis. Int J Mol Sci 2021; 22:9352. [PMID: 34502257 PMCID: PMC8430764 DOI: 10.3390/ijms22179352] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
The catabolic and destructive activity of serine proteases in arthritic joints is well known; however, these enzymes can also signal pain and inflammation in joints. For example, thrombin, trypsin, tryptase, and neutrophil elastase cleave the extracellular N-terminus of a family of G protein-coupled receptors and the remaining tethered ligand sequence then binds to the same receptor to initiate a series of molecular signalling processes. These protease activated receptors (PARs) pervade multiple tissues and cells throughout joints where they have the potential to regulate joint homeostasis. Overall, joint PARs contribute to pain, inflammation, and structural integrity by altering vascular reactivity, nociceptor sensitivity, and tissue remodelling. This review highlights the therapeutic potential of targeting PARs to alleviate the pain and destructive nature of elevated proteases in various arthritic conditions.
Collapse
Affiliation(s)
| | - Jason J. McDougall
- Departments of Pharmacology and Anesthesia, Pain Management & Perioperative Medicine, Dalhousie University, 5850 College Street, Halifax, NS B3H 4R2, Canada;
| |
Collapse
|
37
|
Perry J, Roelofs AJ, Mennan C, McCarthy HS, Richmond A, Clark SM, Riemen AHK, Wright K, De Bari C, Roberts S. Human Mesenchymal Stromal Cells Enhance Cartilage Healing in a Murine Joint Surface Injury Model. Cells 2021; 10:1999. [PMID: 34440768 PMCID: PMC8393840 DOI: 10.3390/cells10081999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 01/15/2023] Open
Abstract
Human umbilical cord (hUC)- or bone marrow (hBM)-derived mesenchymal stromal cells (MSCs) were evaluated as an allogeneic source of cells for cartilage repair. We aimed to determine if they could enhance healing of chondral defects with or without the recruitment of endogenous cells. hMSCs were applied into a focal joint surface injury in knees of adult mice expressing tdTomato fluorescent protein in cells descending from Gdf5-expressing embryonic joint interzone cells. Three experimental groups were used: (i) hUC-MSCs, (ii) hBM-MSCs and (iii) PBS (vehicle) without cells. Cartilage repair was assessed after 8 weeks and tdTomato-expressing cells were detected by immunostaining. Plasma levels of pro-inflammatory mediators and other markers were measured by electrochemiluminescence. Both hUC-MSC (n = 14, p = 0.009) and hBM-MSC (n = 13, p = 0.006) treatment groups had significantly improved cartilage repair compared to controls (n = 18). While hMSCs were not detectable in the repair tissue at 8 weeks post-implantation, increased endogenous Gdf5-lineage cells were detected in repair tissue of hUC-MSC-treated mice. This xenogeneic study indicates that hMSCs enhance intrinsic cartilage repair mechanisms in mice. Hence, hMSCs, particularly the more proliferative hUC-MSCs, could represent an attractive allogeneic cell population for treating patients with chondral defects and perhaps prevent the onset and progression of osteoarthritis.
Collapse
Affiliation(s)
- Jade Perry
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| | - Anke J. Roelofs
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Claire Mennan
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| | - Helen S. McCarthy
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| | - Alison Richmond
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Susan M. Clark
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Anna H. K. Riemen
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Karina Wright
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| | - Cosimo De Bari
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
- Arthritis and Regenerative Medicine Laboratory, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK; (A.J.R.); (A.R.); (S.M.C.); (A.H.K.R.); (C.D.B.)
| | - Sally Roberts
- The Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, Shropshire SY10 7AG, UK; (C.M.); (H.S.M.); (K.W.); (S.R.)
- The School of Pharmacy & Bioengineering, Keele University, Staffordshire ST5 5BG, UK
- The Tissue Engineering & Regenerative Therapies Centre versus Arthritis, Cambridge CB2 2QQ, UK
| |
Collapse
|
38
|
Ariffin SMZ, Bennett D, Ferrell WR, Lockhart JC, Dunning L, Clements DN, Lascelles BDX, Ibrahim TAT, Johnston P. Protease activated receptor 2 and matriptase expression in the joints of cats with and without osteoarthritis. J Feline Med Surg 2021; 23:794-803. [PMID: 33284033 PMCID: PMC10812195 DOI: 10.1177/1098612x20977796] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES The aim of this study was to determine the presence of protease-activated receptor 2 (PAR2) and matriptase proteins and quantify PAR2 and matriptase mRNA expression in the articular cartilage and synovial membrane of cats with and without osteoarthritis (OA). METHODS A total of 28 articular cartilage samples from adult cats (14 OA and 14 normal), 10 synovial membranes from adult cats (five OA and five normal) and three cartilage samples from 9-week-old fetal cats were used. The presence of PAR2 and matriptase in the cartilage and synovial membrane of the adult samples was detected by immunohistochemical (IHC) staining, while real-time PCR was used for mRNA expression analyses in all samples. RESULTS PAR2 was detected in all OA and normal articular cartilage and synovial membrane samples but confined to only a few superficial chondrocytes in the normal samples. Matriptase was only detected in OA articular cartilage and synovial membrane samples. PAR2 and matriptase mRNA expression were, however, detected in all cartilage and synovial membrane samples. PAR2 and matriptase mRNA expression levels in OA articular cartilage were five (P <0.001) and 3.3 (P <0.001) times higher than that of the healthy group, respectively. There was no significant difference (P = 0.05) in the OA synovial membrane PAR2 and matriptase mRNA expression compared with the normal samples. CONCLUSIONS AND RELEVANCE Detection of PAR2 and matriptase proteins and gene expression in feline articular tissues is a novel and important finding, and supports the hypothesis that serine proteases are involved in the pathogenesis of feline OA. The consistent presence of PAR2 and matriptase protein in the cytoplasm of OA chondrocytes suggests a possible involvement of proteases in cartilage degradation. Further investigations into the PAR2 and matriptase pathobiology could enhance our understanding of the proteolytic cascades in feline OA, which might lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Siti M Zainal Ariffin
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan, Malaysia
| | - David Bennett
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - William R Ferrell
- Institute of Immunity, Infection and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - John C Lockhart
- Institute of Biomedical and Environmental Health Research, University of the West of Scotland, Paisley, UK
| | - Lynette Dunning
- Institute of Biomedical and Environmental Health Research, University of the West of Scotland, Paisley, UK
| | - Dylan N Clements
- Royal (Dick) School for Veterinary Studies and The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - B Duncan X Lascelles
- Translational Research in Pain, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
- Centre for Translational Pain Research, Department of Anaesthesiology, Duke University, Durham, NC, USA
| | - Tengku A Tengku Ibrahim
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor Darul Ehsan, Malaysia
| | - Pamela Johnston
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
39
|
Bone marrow derived mast cells injected into the osteoarthritic knee joints of mice induced by sodium monoiodoacetate enhanced spontaneous pain through activation of PAR2 and action of extracellular ATP. PLoS One 2021; 16:e0252590. [PMID: 34086763 PMCID: PMC8177436 DOI: 10.1371/journal.pone.0252590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
Conditions that resemble osteoarthritis (OA) were produced by injection of sodium monoiodoacetate (MIA) into the knee joints of mice. Bone marrow derived mast cells (BMMCs) injected into the OA knee joints enhanced spontaneous pain. Since no spontaneous pain was observed when BMMCs were injected into the knee joints of control mice that had not been treated with MIA, BMMCs should be activated within the OA knee joints and release some pain-inducible factors. Protease activated receptor-2 (PAR2) antagonist (FSLLRY-NH2) almost abolished the pain-enhancing effects of BMMCs injected into the OA knee joints, suggesting that tryptase, a mast cell protease that is capable of activating PAR2, should be released from the injected BMMCs and enhance pain through activation of PAR2. When PAR2 agonist (SLIGKV-NH2) instead of BMMCs was injected into the OA knee joints, it was also enhanced pain. Apyrase, an ATP degrading enzyme, injected into the OA knee joints before BMMCs suppressed the pain enhanced by BMMCs. We showed that purinoceptors (P2X4 and P2X7) were expressed in BMMCs and that extracellular ATP stimulated the release of tryptase from BMMCs. These observations suggest that ATP may stimulate degranulation of BMMCs and thereby enhanced pain. BMMCs injected into the OA knee joints stimulated expression of IL-1β, IL-6, TNF-α, CCL2, and MMP9 genes in the infrapatellar fat pads, and PAR2 antagonist suppressed the stimulatory effects of BMMCs. Our study suggests that intermittent pain frequently observed in OA knee joints may be due, at least partly, to mast cells through activation of PAR2 and action of ATP, and that intraarticular injection of BMMCs into the OA knee joints may provide a useful experimental system for investigating molecular mechanisms by which pain is induced in OA knee joints.
Collapse
|
40
|
Neuroimmune interactions and osteoarthritis pain: focus on macrophages. Pain Rep 2021; 6:e892. [PMID: 33981927 PMCID: PMC8108586 DOI: 10.1097/pr9.0000000000000892] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/01/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022] Open
Abstract
Bidirectional interactions between the immune system and the nervous system are increasingly appreciated as playing a pathogenic role in chronic pain. Unraveling the mechanisms by which inflammatory pain is mediated through communication between nerves and immune cells may lead to exciting new strategies for therapeutic intervention. In this narrative review, we focus on the role of macrophages in the pathogenesis of osteoarthritis (OA) pain. From regulating homeostasis to conducting phagocytosis, and from inducing inflammation to resolving it, macrophages are plastic cells that are highly adaptable to their environment. They rely on communicating with the environment through cytokines, growth factors, neuropeptides, and other signals to respond to inflammation or injury. The contribution of macrophages to OA joint damage has garnered much attention in recent years. Here, we discuss how macrophages may participate in the initiation and maintenance of pain in OA. We aim to summarize what is currently known about macrophages in OA pain and identify important gaps in the field to fuel future investigations.
Collapse
|
41
|
Abji F, Rasti M, Gómez-Aristizábal A, Muytjens C, Saifeddine M, Mihara K, Motahhari M, Gandhi R, Viswanathan S, Hollenberg MD, Oikonomopoulou K, Chandran V. Proteinase-Mediated Macrophage Signaling in Psoriatic Arthritis. Front Immunol 2021; 11:629726. [PMID: 33763056 PMCID: PMC7982406 DOI: 10.3389/fimmu.2020.629726] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 12/29/2020] [Indexed: 11/29/2022] Open
Abstract
Objective Multiple proteinases are present in the synovial fluid (SF) of an arthritic joint. We aimed to identify inflammatory cell populations present in psoriatic arthritis (PsA) SF compared to osteoarthritis (OA) and rheumatoid arthritis (RA), identify their proteinase-activated receptor 2 (PAR2) signaling function and characterize potentially active SF serine proteinases that may be PAR2 activators. Methods Flow cytometry was used to characterize SF cells from PsA, RA, OA patients; PsA SF cells were further characterized by single cell 3’-RNA-sequencing. Active serine proteinases were identified through cleavage of fluorogenic trypsin- and chymotrypsin-like substrates, activity-based probe analysis and proteomics. Fluo-4 AM was used to monitor intracellular calcium cell signaling. Cytokine expression was evaluated using a multiplex Luminex panel. Results PsA SF cells were dominated by monocytes/macrophages, which consisted of three populations representing classical, non-classical and intermediate cells. The classical monocytes/macrophages were reduced in PsA compared to OA/RA, whilst the intermediate population was increased. PAR2 was elevated in OA vs. PsA/RA SF monocytes/macrophages, particularly in the intermediate population. PAR2 expression and signaling in primary PsA monocytes/macrophages significantly impacted the production of monocyte chemoattractant protein-1 (MCP-1). Trypsin-like serine proteinase activity was elevated in PsA and RA SF compared to OA, while chymotrypsin-like activity was elevated in RA compared to PsA. Tryptase-6 was identified as an active serine proteinase in SF that could trigger calcium signaling partially via PAR2. Conclusion PAR2 and its activating proteinases, including tryptase-6, can be important mediators of inflammation in PsA. Components within this proteinase-receptor axis may represent novel therapeutic targets.
Collapse
Affiliation(s)
- Fatima Abji
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Mozhgan Rasti
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | | | - Carla Muytjens
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Mahmoud Saifeddine
- Department of Physiology & Pharmacology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Koichiro Mihara
- Department of Physiology & Pharmacology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Majid Motahhari
- Department of Physiology & Pharmacology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Rajiv Gandhi
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Division of Orthopaedic Surgery, Department of Surgery, Toronto Western Hospital, Toronto, ON, Canada
| | - Sowmya Viswanathan
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Division of Hematology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada.,Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Katerina Oikonomopoulou
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Vinod Chandran
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Division of Rheumatology, Department of Medicine, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Department of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
42
|
Blaker CL, Zaki S, Little CB, Clarke EC. Long-term Effect of a Single Subcritical Knee Injury: Increasing the Risk of Anterior Cruciate Ligament Rupture and Osteoarthritis. Am J Sports Med 2021; 49:391-403. [PMID: 33378213 DOI: 10.1177/0363546520977505] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Rupture of the anterior cruciate ligament (ACL) is a well-known risk factor for the development of posttraumatic osteoarthritis (PTOA), but patients with the "same injury" can have vastly different trajectories for the onset and progression of disease. Minor subcritical injuries preceding the critical injury event may drive this disparity through preexisting tissue pathologies and sensory changes. PURPOSE To investigate the role of subcritical injury on ACL rupture risk and PTOA through the evaluation of pain behaviors, joint mechanics, and tissue structural change in a mouse model of knee injury. STUDY DESIGN Controlled laboratory study. METHODS Ten-week-old male C57BL/6J mice were allocated to naïve control and subcritical knee injury groups. Injury was induced by a single mechanical compression to the right hindlimb, and mice were evaluated using joint histopathology, anteroposterior joint biomechanics, pain behaviors (mechanical allodynia and hindlimb weightbearing), and isolated ACL tensile testing to failure at 1, 2, 4, or 8 weeks after injury. RESULTS Subcritical knee injury produced focal osteochondral lesions in the patellofemoral and lateral tibiofemoral compartments with no resolution for the duration of the study (8 weeks). These lesions were characterized by focal loss of proteoglycan staining, cartilage structural change, chondrocyte pathology, microcracks, and osteocyte cell loss. Injury also resulted in the rapid onset of allodynia (at 1 week), which persisted over time and reduced ACL failure load (P = .006; mean ± SD, 7.91 ± 2.01 N vs 9.37 ± 1.01 N in naïve controls at 8 weeks after injury), accompanied by evidence of ACL remodeling at the femoral enthesis. CONCLUSION The present study in mice establishes a direct effect of a single subcritical knee injury on the development of specific joint tissue pathologies (osteochondral lesions and progressive weakening of the ACL) and allodynic sensitization. These findings demonstrate a predisposition for secondary critical injuries (eg, ACL rupture) and an increased risk of PTOA onset and progression (structurally and symptomatically). CLINICAL RELEVANCE Subcritical knee injuries are a common occurrence and, based on this study, can cause persistent sensory and structural change. These findings have important implications for the understanding of risk factors of ACL injury and subsequent PTOA, particularly with regard to prevention and management strategies following an often underreported event.
Collapse
Affiliation(s)
- Carina L Blaker
- Murray Maxwell Biomechanics Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, Northern Clinical School, University of Sydney, St Leonards, Australia.,Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, Northern Clinical School, University of Sydney, St Leonards, Australia
| | - Sanaa Zaki
- Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, Northern Clinical School, University of Sydney, St Leonards, Australia.,Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Camperdown, Australia
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratories, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, Northern Clinical School, University of Sydney, St Leonards, Australia
| | - Elizabeth C Clarke
- Murray Maxwell Biomechanics Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Faculty of Medicine and Health, Northern Clinical School, University of Sydney, St Leonards, Australia
| |
Collapse
|
43
|
Butterfield NC, Curry KF, Steinberg J, Dewhurst H, Komla-Ebri D, Mannan NS, Adoum AT, Leitch VD, Logan JG, Waung JA, Ghirardello E, Southam L, Youlten SE, Wilkinson JM, McAninch EA, Vancollie VE, Kussy F, White JK, Lelliott CJ, Adams DJ, Jacques R, Bianco AC, Boyde A, Zeggini E, Croucher PI, Williams GR, Bassett JHD. Accelerating functional gene discovery in osteoarthritis. Nat Commun 2021; 12:467. [PMID: 33473114 PMCID: PMC7817695 DOI: 10.1038/s41467-020-20761-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 12/14/2020] [Indexed: 01/29/2023] Open
Abstract
Osteoarthritis causes debilitating pain and disability, resulting in a considerable socioeconomic burden, yet no drugs are available that prevent disease onset or progression. Here, we develop, validate and use rapid-throughput imaging techniques to identify abnormal joint phenotypes in randomly selected mutant mice generated by the International Knockout Mouse Consortium. We identify 14 genes with functional involvement in osteoarthritis pathogenesis, including the homeobox gene Pitx1, and functionally characterize 6 candidate human osteoarthritis genes in mouse models. We demonstrate sensitivity of the methods by identifying age-related degenerative joint damage in wild-type mice. Finally, we phenotype previously generated mutant mice with an osteoarthritis-associated polymorphism in the Dio2 gene by CRISPR/Cas9 genome editing and demonstrate a protective role in disease onset with public health implications. We hope this expanding resource of mutant mice will accelerate functional gene discovery in osteoarthritis and offer drug discovery opportunities for this common, incapacitating chronic disease.
Collapse
Affiliation(s)
- Natalie C Butterfield
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Katherine F Curry
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Julia Steinberg
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
- Cancer Council NSW, Sydney, NSW, 2000, Australia
| | - Hannah Dewhurst
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Davide Komla-Ebri
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Naila S Mannan
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Anne-Tounsia Adoum
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Victoria D Leitch
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - John G Logan
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Julian A Waung
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Elena Ghirardello
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK
| | - Lorraine Southam
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Scott E Youlten
- The Garvan Institute of Medical Research and St. Vincent's Clinical School, University of New South Wales Medicine, Sydney, NSW, 2010, Australia
| | - J Mark Wilkinson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2RX, UK
- Centre for Integrated Research into Musculoskeletal Ageing and Sheffield Healthy Lifespan Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Elizabeth A McAninch
- Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, IL, 60612, USA
| | | | - Fiona Kussy
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Jacqueline K White
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | | | - David J Adams
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Richard Jacques
- School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, S1 4DA, UK
| | - Antonio C Bianco
- Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Alan Boyde
- Dental Physical Sciences, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764, Neuherberg, Germany
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Peter I Croucher
- The Garvan Institute of Medical Research and St. Vincent's Clinical School, University of New South Wales Medicine, Sydney, NSW, 2010, Australia
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK.
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK.
| |
Collapse
|
44
|
Saito M, Nishitani K, Ikeda HO, Yoshida S, Iwai S, Ji X, Nakahata A, Ito A, Nakamura S, Kuriyama S, Yoshitomi H, Murata K, Aoyama T, Ito H, Kuroki H, Kakizuka A, Matsuda S. A VCP modulator, KUS121, as a promising therapeutic agent for post-traumatic osteoarthritis. Sci Rep 2020; 10:20787. [PMID: 33247195 PMCID: PMC7695735 DOI: 10.1038/s41598-020-77735-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/13/2020] [Indexed: 12/20/2022] Open
Abstract
Post-traumatic osteoarthritis (PTOA) is a major cause which hinders patients from the recovery after intra-articular injuries or surgeries. Currently, no effective treatment is available. In this study, we showed that inhibition of the acute stage chondrocyte death is a promising strategy to mitigate the development of PTOA. Namely, we examined efficacies of Kyoto University Substance (KUS) 121, a valosin-containing protein modulator, for PTOA as well as its therapeutic mechanisms. In vivo, in a rat PTOA model by cyclic compressive loading, intra-articular treatments of KUS121 significantly improved the modified Mankin scores and reduced damaged-cartilage volumes, as compared to vehicle treatment. Moreover, KUS121 markedly reduced the numbers of TUNEL-, CHOP-, MMP-13-, and ADAMTS-5-positive chondrocytes in the damaged knees. In vitro, KUS121 rescued human articular chondrocytes from tunicamycin-induced cell death, in both monolayer culture and cartilage explants. It also significantly downregulated the protein or gene expression of ER stress markers, proinflammatory cytokines, and extracellular-matrix-degrading enzymes induced by tunicamycin or IL-1β. Collectively, these results demonstrated that KUS121 protected chondrocytes from cell death through the inhibition of excessive ER stress. Therefore, KUS121 would be a new, promising therapeutic agent with a protective effect on the progression of PTOA.
Collapse
Affiliation(s)
- Motoo Saito
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kohei Nishitani
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Hanako O Ikeda
- Department of Ophthalmology and Visual Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigeo Yoshida
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sachiko Iwai
- Department of Ophthalmology and Visual Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Xiang Ji
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihiro Nakahata
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Ito
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinichiro Nakamura
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinichi Kuriyama
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Yoshitomi
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichi Murata
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Advanced Medicine of Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiromu Ito
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Advanced Medicine of Rheumatic Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Kuroki
- Department of Physical Therapy, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akira Kakizuka
- Laboratory of Functional Biology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
45
|
Sogi Y, Yabe Y, Hagiwara Y, Tsuchiya M, Onoda Y, Sekiguchi T, Itaya N, Yoshida S, Yano T, Suzuki K, Onoki T, Itoi E. Joint hemorrhage accelerates cartilage degeneration in a rat immobilized knee model. BMC Musculoskelet Disord 2020; 21:761. [PMID: 33213419 PMCID: PMC7678279 DOI: 10.1186/s12891-020-03795-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 11/12/2020] [Indexed: 01/05/2023] Open
Abstract
Background Joint hemorrhage is caused by trauma, ligament reconstruction surgery, and bleeding disorders such as hemophilia. Recurrence of hemorrhage in the joint space induces hemosiderotic synovitis and oxidative stress, resulting in both articular cartilage degeneration and arthropathy. Joint immobilization is a common treatment option for articular fractures accompanied by joint hemorrhage. Although joint hemorrhage has negative effects on the articular cartilage, there is no consensus on whether a reduction in joint hemorrhage would effectively prevent articular cartilage degeneration. The purpose of this study was to investigate the effect of joint hemorrhage combined with joint immobilization on articular cartilage degeneration in a rat immobilized knee model. Methods The knee joints of adult male rats were immobilized at the flexion using an internal fixator from 3 days to 8 weeks. The rats were randomly divided into the following groups: immobilized blood injection (Im-B) and immobilized-normal saline injection (Im-NS) groups. The cartilage was evaluated in two areas (contact and non-contact areas). The cartilage was used to assess chondrocyte count, Modified Mankin score, and cartilage thickness. The total RNA was extracted from the cartilage in both areas, and the expression of metalloproteinase (MMP)-8, MMP-13, interleukin (IL)-1β, and tumor necrosis factor (TNF)-α was measured by quantitative real-time polymerase chain reaction. Results The number of chondrocytes in the Im-B group significantly decreased in both areas, compared with that in the Im-NS group. Modified Mankin score from 4 to 8 weeks of the Im-B group was significantly higher than that of the Im-NS group only in the contact area. The expression of MMP-8 and MMP-13 from 2 to 4 weeks and TNF-α from 2 to 8 weeks significantly increased in the Im-B group compared with those in the Im-NS group, but there was no significant difference in IL-1β expression. Conclusions The results showed that joint hemorrhage exacerbated immobilization-induced articular cartilage degeneration. Drainage of a joint hemorrhage or avoidance of loading may help prevent cartilage degeneration during joint immobilization with a hemorrhage. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-020-03795-0.
Collapse
Affiliation(s)
- Yasuhito Sogi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Yutaka Yabe
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Yoshihiro Hagiwara
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan.
| | - Masahiro Tsuchiya
- Department of Nursing, Faculty of Health Science, Tohoku Fukushi University, 1-8-1 Kunimi, Aoba-ku, Sendai, 981-8522, Japan
| | - Yoshito Onoda
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Takuya Sekiguchi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Nobuyuki Itaya
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Shinichiro Yoshida
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Toshihisa Yano
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Kazuaki Suzuki
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Takahiro Onoki
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| | - Eiji Itoi
- Department of Orthopaedic Surgery, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, 980-8574, Japan
| |
Collapse
|
46
|
Courties A, Do A, Leite S, Legris M, Sudre L, Pigenet A, Petit J, Nourissat G, Cambon-Binder A, Maskos U, Berenbaum F, Sellam J. The Role of the Non-neuronal Cholinergic System in Inflammation and Degradation Processes in Osteoarthritis. Arthritis Rheumatol 2020; 72:2072-2082. [PMID: 32638534 DOI: 10.1002/art.41429] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 06/27/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The non-neuronal cholinergic system represents non-neuronal cells that have the biochemical machinery to synthetize de novo and/or respond to acetylcholine (ACh). We undertook this study to investigate this biochemical machinery in chondrocytes and its involvement in osteoarthritis (OA). METHODS Expression of the biochemical machinery for ACh metabolism and nicotinic ACh receptors (nAChR), particularly α7-nAChR, in human OA and murine chondrocytes was determined by polymerase chain reaction and ligand-binding. We investigated the messenger RNA expression of the human duplicate α7-nACh subunit, called CHRFAM7A, which is responsible for truncated α7-nAChR. We assessed the effect of nAChR on chondrocytes activated by interleukin-1β (IL-1β) and the involvement of α7-nAChR using chondrocytes from wild-type (WT) and α7-deficient Chrna7-/- mice. The role of α7-nAChR in OA was explored after medial meniscectomy in WT and Chrna7-/- mice. RESULTS Human and murine chondrocytes express the biochemical partners of the non-neuronal cholinergic system and a functional α7-nAChR at their cell surface (n = 5 experiments with 5 samples each). The expression of CHRFAM7A in human OA chondrocytes (n = 23 samples) correlated positively with matrix metalloproteinase 3 (MMP-3) (r = 0.38, P < 0.05) and MMP-13 (r = 0.48, P < 0.05) expression. Nicotine decreased the IL-1β-induced IL-6 and MMP expression, in a dose-dependent manner, in WT chondrocytes but not in Chrna7-/- chondrocytes. Chrna7-/- mice that underwent meniscectomy (n = 7) displayed more severe OA cartilage damage (mean ± SD Osteoarthritis Research Society International [OARSI] score 4.46 ± 1.09) compared to WT mice that underwent meniscectomy (n = 9) (mean ± SD OARSI score 3.05 ± 0.9; P < 0.05). CONCLUSION The non-neuronal cholinergic system is functionally expressed in chondrocytes. Stimulation of nAChR induces antiinflammatory and anticatabolic activity through α7-nAChR, but the anticatabolic activity may be mitigated by truncated α7-nAChR in human chondrocytes. In vivo experiments strongly suggest that α7-nAChR has a protective role in OA.
Collapse
Affiliation(s)
- Alice Courties
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Ariane Do
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Sarah Leite
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Manon Legris
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Laure Sudre
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Audrey Pigenet
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Juliette Petit
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Geoffroy Nourissat
- 2INSERM UMR 938, Centre de Recherche Saint-Antoine, Clinique Maussins, Groupe Ramsay Générale de Santé, Paris, France
| | - Adeline Cambon-Binder
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Uwe Maskos
- Institut Pasteur, Neurobiologie Intégrative des Systèmes Cholinergiques, CNRS UMR 3571, Paris, France
| | - Francis Berenbaum
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Jérémie Sellam
- Sorbonne Université, INSERM UMR 938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, AP-HP, Paris, France
| |
Collapse
|
47
|
Vaamonde-García C, Burguera EF, Vela-Anero Á, Hermida-Gómez T, Filgueira-Fernández P, Fernández-Rodríguez JA, Meijide-Faílde R, Blanco FJ. Intraarticular Administration Effect of Hydrogen Sulfide on an In Vivo Rat Model of Osteoarthritis. Int J Mol Sci 2020; 21:ijms21197421. [PMID: 33050005 PMCID: PMC7582513 DOI: 10.3390/ijms21197421] [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: 09/14/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) is the most common articular chronic disease. However, its current treatment is limited and mostly symptomatic. Hydrogen sulfide (H2S) is an endogenous gas with recognized physiological activities. The purpose here was to evaluate the effects of the intraarticular administration of a slow-releasing H2S compound (GYY-4137) on an OA experimental model. OA was induced in Wistar rats by the transection of medial collateral ligament and the removal of the medial meniscus of the left joint. The animals were randomized into three groups: non-treated and intraarticularly injected with saline or GYY-4137. Joint destabilization induced articular thickening (≈5% increment), the loss of joint mobility and flexion (≈12-degree angle), and increased levels of pain (≈1.5 points on a scale of 0 to 3). Animals treated with GYY-4137 presented improved motor function of the joint, as well as lower pain levels (≈75% recovery). We also observed that cartilage deterioration was attenuated in the GYY-4137 group (≈30% compared with the saline group). Likewise, these animals showed a reduced presence of pro-inflammatory mediators (cyclooxygenase-2, inducible nitric oxide synthase, and metalloproteinase-13) and lower oxidative damage in the cartilage. The increment of the nuclear factor-erythroid 2-related factor 2 (Nrf-2) levels and Nrf-2-regulated gene expression (≈30%) in the GYY-4137 group seem to be underlying its chondroprotective effects. Our results suggest the beneficial impact of the intraarticular administration of H2S on experimental OA, showing a reduced cartilage destruction and oxidative damage, and supporting the use of slow H2S-producing molecules as a complementary treatment in OA.
Collapse
Affiliation(s)
- Carlos Vaamonde-García
- Grupo de Terapia Celular y Medicina Regenerativa, Universidad de A Coruña, Agrupación Estratégica CICA- INIBIC, Complexo Hospitalario Universitario A Coruña, Campus Oza, 15006 A Coruña, Spain; (C.V.-G.); (Á.V.-A.)
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
| | - Elena F. Burguera
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
- Centro de investigación biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Ángela Vela-Anero
- Grupo de Terapia Celular y Medicina Regenerativa, Universidad de A Coruña, Agrupación Estratégica CICA- INIBIC, Complexo Hospitalario Universitario A Coruña, Campus Oza, 15006 A Coruña, Spain; (C.V.-G.); (Á.V.-A.)
| | - Tamara Hermida-Gómez
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
- Centro de investigación biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Purificación Filgueira-Fernández
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
- Centro de investigación biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Jennifer A. Fernández-Rodríguez
- Grupo de Envejecimiento e Inflamación, Agrupación Estratégica CICA- INIBIC, Complexo Hospitalario Universitario A Coruña, Sergas, Universidad de A Coruña, As Xubias, 15006 A Coruña, Spain;
| | - Rosa Meijide-Faílde
- Grupo de Terapia Celular y Medicina Regenerativa, Universidad de A Coruña, Agrupación Estratégica CICA- INIBIC, Complexo Hospitalario Universitario A Coruña, Campus Oza, 15006 A Coruña, Spain; (C.V.-G.); (Á.V.-A.)
- Correspondence: (R.M.-F.); (F.J.B.); Tel.: +34-981167000 (ext. 5855) (R.M.-F.); +34-981176399 (F.J.B.)
| | - Francisco J. Blanco
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
- Grupo de Investigación de Reumatología y Salud (GIR), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Agrupación Estrategica CICA-INIBIC, Universidade da Coruña, Campus de Oza, 15006 A Coruña, Spain
- Correspondence: (R.M.-F.); (F.J.B.); Tel.: +34-981167000 (ext. 5855) (R.M.-F.); +34-981176399 (F.J.B.)
| |
Collapse
|
48
|
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
|
49
|
Perry J, McCarthy HS, Bou-Gharios G, van 't Hof R, Milner PI, Mennan C, Roberts S. Injected human umbilical cord-derived mesenchymal stromal cells do not appear to elicit an inflammatory response in a murine model of osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2020; 2:100044. [PMID: 32596691 PMCID: PMC7307639 DOI: 10.1016/j.ocarto.2020.100044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/07/2020] [Indexed: 01/08/2023] Open
Abstract
Objective This study investigated the effect of hUC-MSCs on osteoarthritis (OA) progression in a xenogeneic model. Design Male, 10 week-old C57BL/6 mice underwent sham surgery (n = 15) or partial medial meniscectomy (PMM; n = 76). 5x105 hUC-MSCs (from 3 donors: D1, D2 and D3) were phenotyped via RT-qPCR and immunoprofiling their response to inflammatory stimuli. They were injected into the mouse joints 3 and 6 weeks post-surgery, harvesting joints at 8 and 12 weeks post-surgery, respectively. A no cell ‘control’ group was also used (n = 29). All knee joints were assessed via micro-computed tomography (μCT) and histology and 10 plasma markers were analysed at 12 weeks. Results PMM resulted in cartilage loss and osteophyte formation resembling human OA at both time-points. Injection of one donor's hUC-MSCs into the joint significantly reduced the loss of joint space at 12 weeks post-operatively compared with the PMM control. This ‘effective’ population of MSCs up-regulated the genes, IDO and TSG6, when stimulated with inflammatory cytokines, more than those from the other two donors. No evidence of an inflammatory response to the injected cells in any animals, either histologically or with plasma biomarkers, arose. Conclusion Beneficial change in a PMM joint was seen with only one hUC-MSC population, perhaps indicating that cell therapy is not appropriate for severely osteoarthritic joints. However, none of the implanted cells appeared to elicit an inflammatory response at the time-points studied. The variability of UC donors suggests some populations may be more therapeutic than others and donor characterisation is essential in developing allogeneic cell therapies.
Collapse
Affiliation(s)
- J Perry
- Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, SY10 7AG, UK.,School of Pharmacy and Bioengineering (PhaB), Keele University, Keele, ST4 7QB, UK
| | - H S McCarthy
- Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, SY10 7AG, UK.,School of Pharmacy and Bioengineering (PhaB), Keele University, Keele, ST4 7QB, UK
| | - G Bou-Gharios
- Institute of Ageing and Chronic Disease, University of Liverpool, L7 8TX, UK
| | - R van 't Hof
- Institute of Ageing and Chronic Disease, University of Liverpool, L7 8TX, UK
| | - P I Milner
- Institute of Ageing and Chronic Disease, University of Liverpool, L7 8TX, UK
| | - C Mennan
- Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, SY10 7AG, UK.,School of Pharmacy and Bioengineering (PhaB), Keele University, Keele, ST4 7QB, UK
| | - S Roberts
- Robert Jones & Agnes Hunt Orthopaedic Hospital NHS Foundation Trust, Oswestry, SY10 7AG, UK.,School of Pharmacy and Bioengineering (PhaB), Keele University, Keele, ST4 7QB, UK
| |
Collapse
|
50
|
Pulsed Electromagnetic Field Inhibits Synovitis via Enhancing the Efferocytosis of Macrophages. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4307385. [PMID: 32596310 PMCID: PMC7273431 DOI: 10.1155/2020/4307385] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/06/2020] [Indexed: 01/15/2023]
Abstract
Synovitis plays an important role in the pathogenesis of arthritis, which is closely related to the joint swell and pain of patients. The purpose of this study was to investigate the anti-inflammatory effects of pulsed electromagnetic fields (PEMF) on synovitis and its underlying mechanisms. Destabilization of the medial meniscus (DMM) model and air pouch inflammation model were established to induce synovitis in C57BL/6 mice. The mice were then treated by PEMF (pulse waveform, 1.5 mT, 75 Hz, 10% duty cycle). The synovitis scores as well as the levels of IL-1β and TNF-α suggested that PEMF reduced the severity of synovitis in vivo. Moreover, the proportion of neutrophils in the synovial-like layer was decreased, while the proportion of macrophages increased after PEMF treatment. In addition, the phagocytosis of apoptotic neutrophils by macrophages (efferocytosis) was enhanced by PEMF. Furthermore, the data from western blot assay showed that the phosphorylation of P38 was inhibited by PEMF. In conclusion, our current data show that PEMF noninvasively exhibits the anti-inflammatory effect on synovitis via upregulation of the efferocytosis in macrophages, which may be involved in the phosphorylation of P38.
Collapse
|