1
|
Rieger R, Kaderli S, Boulocher C. In vivo impact on rabbit subchondral bone of viscosupplementation with a hyaluronic acid antioxidant conjugate. BMC Musculoskelet Disord 2024; 25:1018. [PMID: 39702245 DOI: 10.1186/s12891-024-07921-0] [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: 11/16/2023] [Accepted: 10/04/2024] [Indexed: 12/21/2024] Open
Abstract
OBJECTIVE This study aimed to assess the effects of an antioxidant-conjugated Hyaluronic Acid (HA), specifically HA-4-aminoresorcinol (HA4AR), on articular cartilage and subchondral bone in osteoarthritis (OA). We conducted a comparative analysis between HA4AR and a commercially available high molecular weight HA formulation in a rabbit model of OA. MATERIALS AND METHODS Eighteen rabbits underwent unilateral anterior cruciate ligament transection (ACLT) and were divided into three groups of six: Saline-group, HA-group, and HA4AR-group, based on the type of intra-articular injection received. Additionally, eight non-operated contralateral knees served as reference points (Contralateral-group). Six weeks post-surgery, iodine-enhanced micro-computed tomography imaging was used to evaluate articular cartilage volume and thickness, and to assess subchondral bone microarchitecture and mineral density. RESULTS Cartilage thickness in both the HA and HA4AR groups was comparable to that of the Contralateral group. Notably, there was a significant reduction in subchondral bone plate tissue mineral density in the HA-group when compared to both the HA4AR and Saline groups (p < 0.05). However, no significant differences in trabecular subchondral bone microarchitectural parameters and mineral densities were observed between the HA4AR-group and the Saline-group. When compared to the Contralateral, Saline, and HA4AR groups, the HA-group exhibited a marked decrease in subchondral bone plate tissue mineral density (p < 0.05). Additionally, a significant reduction in trabecular bone volume fraction was noted in the HA-group compared to the Contralateral-group (p < 0.05). CONCLUSIONS The HA-4AR hydrogel demonstrated significant preservation of subchondral bone plate tissue mineral density compared to HA alone, while other bone microarchitectural parameters remained largely unchanged. These findings indicate that HA-4AR may provide enhanced osteoprotective benefits in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- R Rieger
- Université de Lyon, VetAgro Sup, UPSP ICE 2021.A104, Marcy-l'Etoile, 69280, France.
- Université de Lyon, École Centrale de Lyon, Ecully, 69134, France.
| | - S Kaderli
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, Quai Ernest-Ansermet 30, Geneva, 1211, Switzerland
| | - C Boulocher
- Université de Lyon, VetAgro Sup, UPSP ICE 2021.A104, Marcy-l'Etoile, 69280, France
- UniLaSalle Polytechnic Institute, Veterinary College, Campus of Rouen, Mon Saint Aignan, 76130, France
| |
Collapse
|
2
|
Jin Y, Hu C, Xia J, Xie D, Ye L, Ye X, Jiang L, Song H, Zhu Y, Jiang S, Li W, Qi W, Yang Y, Hu Z. Bimetallic clusterzymes-loaded dendritic mesoporous silica particle regulate arthritis microenvironment via ROS scavenging and YAP1 stabilization. Bioact Mater 2024; 42:613-627. [PMID: 39314862 PMCID: PMC11417149 DOI: 10.1016/j.bioactmat.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Clusterzymes are synthetic enzymes exhibiting substantial catalytic activity and selectivity, which are uniquely driven by single-atom constructs. A dramatic increase in antioxidant capacity, 158 times more than natural trolox, is noted when single-atom copper is incorporated into gold-based clusterzymes to form Au24Cu1. Considering the inflammatory and mildly acidic microenvironment characteristic of osteoarthritis (OA), pH-dependent dendritic mesoporous silica nanoparticles (DMSNs) coupled with PEG have been employed as a delivery system for the spatial-temporal release of clusterzymes within active articular regions, thereby enhancing the duration of effectiveness. Nonetheless, achieving high therapeutic efficacy remains a significant challenge. Herein, we describe the construction of a Clusterzymes-DMSNs-PEG complex (CDP) which remarkably diminishes reactive oxygen species (ROS) and stabilizes the chondroprotective protein YAP by inhibiting the Hippo pathway. In the rabbit ACLT (anterior cruciate ligament transection) model, the CDP complex demonstrated inhibition of matrix metalloproteinase activity, preservation of type II collagen and aggregation protein secretion, thus prolonging the clusterzymes' protective influence on joint cartilage structure. Our research underscores the efficacy of the CDP complex in ROS-scavenging, enabled by the release of clusterzymes in response to an inflammatory and slightly acidic environment, leading to the obstruction of the Hippo pathway and downstream NF-κB signaling pathway. This study illuminates the design, composition, and use of DMSNs and clusterzymes in biomedicine, thus charting a promising course for the development of novel therapeutic strategies in alleviating OA.
Collapse
Affiliation(s)
- Yang Jin
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Chuan Hu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Jiechao Xia
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Dingqi Xie
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Lin Ye
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Xinyi Ye
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Honghai Song
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Yutao Zhu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Sicheng Jiang
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| | - Weiqing Li
- Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Weiming Qi
- Zhejiang Center for Medical Device Evaluation, Zhejiang Medical Products Administration Hangzhou 310009, Zhejiang, China
| | - Yannan Yang
- Institute of Optoelectronics, Fudan University, Shanghai, 200433, China
- South Australian ImmunoGENomics Cancer Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Zhijun Hu
- Department of Orthopaedic Surgery, Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, 310016, China
| |
Collapse
|
3
|
Deng Z, Zeng X, Lin B, Chen L, Wu J, Zheng J, Ma Y, Lyu FJ, Zheng Q. Human umbilical cord mesenchymal stem cells on treating osteoarthritis in a rabbit model: Injection strategies. Heliyon 2024; 10:e38384. [PMID: 39430502 PMCID: PMC11489144 DOI: 10.1016/j.heliyon.2024.e38384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/22/2024] Open
Abstract
Human umbilical cord mesenchymal stem cells (UCMSCs) are a novel stem-cell source to treat osteoarthritis (OA). Here we investigated the therapeutic effects of UCMSCs injection strategies on knee OA in a rabbit model. Thirty OA rabbits randomly received normal saline, a single dose of 1 × 106 UCMSCs, or three injections of 1 × 106 UCMSCs at 2, 4, 6 weeks. Articular cartilages were collected after 8 weeks. Macroscopic and histological assessments indicated that intra-articular injection of UCMSCs, both single and multiple injection, significantly reduced the formation of periarticular osteophytes and articular cartilage degeneration when compared with the control. Furthermore, both UCMSCs injections increased the expression of chondrogenic markers in the articular cartilage, and reduced the levels of TNF-α and IL-6 in synovium. Micro-CT showed significant reduction of sub-chondral bone degeneration and osteophytes in the multiple-injection group compared to the control and single-injection group. Taken together, intra-articular injection of UCMSCs for OA treatment is safe and effective. Single and multiple injection of UCMSCs had comparable reparative effect on cartilage lesions, while multiple injection of UCMSCs further exerted effect on enhancing subchondral bone volume.
Collapse
Affiliation(s)
- Zhantao Deng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiaoli Zeng
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Bofu Lin
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lixuan Chen
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Jiwei Wu
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Jie Zheng
- Guangdong Xiangxue Stem Cell Regenerative Medicine Technology Co., Ltd, Guangzhou, China
| | - Yuanchen Ma
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Feng-Juan Lyu
- The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiujian Zheng
- Department of Orthopedics, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Zhang S, Gao G, Zhou X, Du C, Zhu Y, He TC, Xu Y. Development of a novel rabbit model for femoroacetabular impingement through surgically induced acetabular overcoverage. J Orthop Res 2024. [PMID: 39396202 DOI: 10.1002/jor.25994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/20/2024] [Accepted: 09/27/2024] [Indexed: 10/14/2024]
Abstract
There is a lack of validated small animal models for femoroacetabular impingement (FAI) that induce intra-articular lesions and cause osteoarthritis (OA) progression. The gene expression profile of articular cartilage in patients with FAI has not been characterized in animal studies. The purpose of this study is to describe a novel rabbit model for FAI with validated induction of intra-articular lesions and OA progression and to characterize the gene expression pattern in impinged cartilage using this model. Thirty 6-month-old New Zealand White rabbits underwent unilateral endobutton implant placement at the acetabular rim to surgically create overcoverage. Radiological assessment confirmed secure placement of endobutton at the acetabular rim for all operated hips with a mean alteration in lateral center-edge angle (ΔLCEA) of 16.2 ± 6.6°. Gross inspection revealed secondary cartilage injuries in the anterosuperior region of the femoral head for the operated hips. Cartilage injuries were shown to exacerbate with increased impingement duration, as demonstrated by the modified Outerbridge scores and Mankin scores. Immunostaining and quantitative real-time polymerase chain reaction revealed elevated expression of inflammatory, anabolic and catabolic genes in impinged cartilage. RNA sequencing analysis of cartilage tissue revealed a distinct transcriptome profile and identified C-KIT, CD86, and CD68 as central markers. Our study confirmed that the novel rabbit FAI model created acetabular overcoverage and produced articular cartilage injury at the impingement zone. Cartilage from the impingement zone demonstrated a heightened metabolic state, corroborating with the gene expression pattern observed in patients with FAI.
Collapse
Affiliation(s)
- Siqi Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Guanying Gao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Xiang Zhou
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Cancan Du
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yichuan Zhu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, Illinois, USA
| | - Yan Xu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| |
Collapse
|
5
|
Angrisani N, von der Ahe C, Willumeit-Römer R, Windhagen H, Scheper V, Schwarze M, Wiese B, Helmholz H, Reifenrath J. Treatment of osteoarthritis by implantation of Mg- and WE43-cylinders - A preclinical study on bone and cartilage changes and their influence on pain sensation in rabbits. Bioact Mater 2024; 40:366-377. [PMID: 38978802 PMCID: PMC11228885 DOI: 10.1016/j.bioactmat.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/20/2024] [Accepted: 06/01/2024] [Indexed: 07/10/2024] Open
Abstract
With its main features of cartilage degeneration, subchondral bone sclerosis and osteophyte formation, osteoarthritis represents a multifactorial disease with no effective treatment options. As biomechanical shift in the trabecular network may be a driver for further cartilage degeneration, bone enhancement could possibly delay OA progression. Magnesium is known to be osteoconductive and already showed positive effects in OA models. We aimed to use magnesium cylinders to enhance subchondral bone quality, condition of cartilage and pain sensation compared to sole drilling in vivo. After eight weeks of implantation in rabbits, significant increase in subchondral bone volume and trabecular thickness with constant bone mineral density was found indicating favored biomechanics. As representative for pain, a higher number of CD271+ vessels were present in control samples without magnesium. However, this result could not be confirmed by sensitive, objective lameness evaluation using a pressure sensing mat and no positive effect could be shown on either cartilage degeneration evaluated by OARSI score nor the presence of regenerative cells in CD271-stained samples. The presented results show a relevant impact of implanted magnesium on key structures in OA pain with missing clinical relevance regarding pain. Further studies with shifted focus should examine additional structures as joint capsule or osteophytes.
Collapse
Affiliation(s)
- Nina Angrisani
- Hannover Medical School, Clinic for Orthopaedic Surgery, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| | - Christin von der Ahe
- Hannover Medical School, Clinic for Orthopaedic Surgery, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| | | | - Henning Windhagen
- Hannover Medical School, Clinic for Orthopaedic Surgery, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| | - Verena Scheper
- Hannover Medical School, Department of Otolaryngology, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| | - Michael Schwarze
- Hannover Medical School, Clinic for Orthopaedic Surgery, Laboratory for Biomechanics and Biomaterials, Hannover, Lower Saxony, Germany
| | - Björn Wiese
- Helmholtz-Zentrum Hereon, Institute of Metallic Biomaterials, Geesthacht, Germany
| | - Heike Helmholz
- Helmholtz-Zentrum Hereon, Institute of Metallic Biomaterials, Geesthacht, Germany
| | - Janin Reifenrath
- Hannover Medical School, Clinic for Orthopaedic Surgery, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| |
Collapse
|
6
|
Mei S, Jiang F, Liu N, Feng Z, Zheng Y, Yang W, Zhang W, Cui Y, Wang W, Xie J, Zhang N. Sol-gel synthesis of magnesium oxide nanoparticles and their evaluation as a therapeutic agent for the treatment of osteoarthritis. Nanomedicine (Lond) 2024; 19:1867-1878. [PMID: 39109508 PMCID: PMC11457622 DOI: 10.1080/17435889.2024.2382421] [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: 03/26/2024] [Accepted: 07/16/2024] [Indexed: 10/05/2024] Open
Abstract
Aim: We synthesized MgO NPs via sol-gel reaction and investigated them as carriers to deliver Mg2+ to the affected joint for osteoarthritis (OA).Materials & methods: The physicochemical properties of samples were characterized by transmission electron microscope (TEM), dynamic light scattering (DLS) and x-ray diffraction (XRD). The release of Mg2+ was monitored by ICP-MS. The potential cytotoxicity was evaluated using MTT assay. The efficacy and biosafety were evaluated in a rabbit OA model.Results: MgO NPs can prolong the Mg2+ release time from 0.5 h to 12 h. No significant cytotoxicity was observed when concentrations below 250 μg/ml. Intra-articular samples could effectively alleviate the degeneration and destruction of the cartilage.Conclusion: this study demonstrates the potential of MgO NPs as a safe and effective treatment of OA. Simultaneously, the size of the particles may play a significant role in influencing the therapeutic outcome.
Collapse
Affiliation(s)
- Sen Mei
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Fangchao Jiang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Na Liu
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Zhizi Feng
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Yu Zheng
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Wei Yang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Weizhong Zhang
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Yingna Cui
- Department of Chemistry, Dalian University, Dalian, Liaoning, 116000, China
| | - Weiming Wang
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| | - Jin Xie
- Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Nan Zhang
- Department of Orthopedics, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, 116000, China
| |
Collapse
|
7
|
Zhu T, Ruan H, Wang T, Guo Y, Liu Y. An HFD negatively influences both joint and liver health in rabbits with and without an enzymatically-induced model of arthritis. Vet J 2024; 306:106197. [PMID: 38996961 DOI: 10.1016/j.tvjl.2024.106197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/06/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024]
Abstract
Osteoarthritis (OA) is a common arthritis types in animals that causes persistent pain and reduces quality of life. Although a high-fat diet (HFD) is widely believed to induce obesity and have adverse effects on the body, the connection between HFD and joint health is not well understood. Therefore, in this study, 32 healthy male New Zealand rabbits were randomly divided into four groups: healthy rabbits fed a standard diet (NDG, n=8) or an HFD (HDG, n=8), rabbits fed a standard diet (OAG, n=8) and an HFD (HOG, n=8), and arthritis was induced by intra-articular enzyme injection. After 12 weeks of HFD feeding, articular cartilage, synovium, and subchondral bone were isolated and collected. Joint tissue damage was evaluated using histopathological and imaging tests. The results showed that there was no significant difference in body weight between rabbits fed a normal diet and those fed an HFD. However, the HFD led to an increase in joint injuries in both induced and non-induced arthritis rabbits. Specifically, the HFD induced lipid metabolism disorders and liver damage in vivo, significantly elevating the levels of serum inflammatory cytokines and bone metabolism markers. Moreover, HFD exacerbated articular cartilage damage in the joints and increased the accumulation of inflammatory cells in synovial tissue, resulting in a notable increase in synovial macrophages and inflammatory cytokines. Additionally, HFD accelerated the bone resorption process in subchondral bone, leading to the destruction of bone mass and subchondral bone microstructure. In summary, the results of this study indicate that an HFD can cause histological damage to the articular cartilage, synovium, and subchondral bone in rabbits, exacerbating arthritis in pre-existing joint damage. Notably, weight is not the primary factor in this effect.
Collapse
Affiliation(s)
- Tingting Zhu
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Department of Veterinary Surgery, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Hongri Ruan
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Department of Veterinary Surgery, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Tiantian Wang
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Department of Veterinary Surgery, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yingchao Guo
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Department of Veterinary Surgery, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yun Liu
- Heilongjiang Provincial Key Laboratory of Pathogenic Mechanism for Animal Disease and Comparative Medicine, Department of Veterinary Surgery, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
8
|
Dzidzishvili L, Fernández-Valle ME, Moreno Molera D, Calvo E, López-Torres II. High-resolution magnetic resonance imaging can predict osteoarthritic progression after medial meniscus posterior root injury: randomized in vivo experimental study in a rabbit model. J ISAKOS 2024; 9:526-533. [PMID: 38583525 DOI: 10.1016/j.jisako.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/25/2024] [Accepted: 03/29/2024] [Indexed: 04/09/2024]
Abstract
IMPORTANCE The field of meniscal root preservation has undergone significant advancement over the past decades; however, the challenge remains to fully understand whether meniscal root repair can ultimately arrest or delay osteoarthritic changes. OBJECTIVE To assess longitudinal changes in articular cartilage, subchondral bone, and progression to meniscal extrusion (ME) using high-resolution magnetic resonance imaging (MRI). METHODS Medial meniscus posterior root tear was surgically induced in 39 New Zealand white rabbits. Animals were randomly assigned into three experimental groups: partial meniscectomy after root tear (PM, n = 13); root tear left in situ (CT, n = 13); and transtibial root repair (RR, n = 13). Contralateral limbs were used as healthy controls. High resolution 4.7 Tesla MRI of the knee joint was performed at baseline, after 2-, and 4-months of post-surgery. Cartilage thickness was calculated in medial and lateral compartments. In addition, the evaluation of ME, subchondral bone edema and healing potential after root repair were assessed too. RESULTS Progressive cartilage thinning, ME, and subchondral bone edema were evident in all 3 study groups after 4-months of follow-up. The mean cartilage thickness in the PM group was 0.53 mm (±0.050), 0.57 mm (±0.05) in the CT group, and 0.60 mm (±0.08) in the RR group. The PM group exhibited significantly higher cartilage loss when compared to the CT and RR groups (p < 0.001). Moreover, progressive ME and subchondral bone edema were associated with a more severe cartilage loss at the final follow-up. CONCLUSION Meniscal root repair did not halt but rather reduced the progression of osteoarthritis (OA). Degenerative changes worsened at a rapid rate in the PM group compared to the RR and CT groups. Early cartilage swelling, persistent subchondral edema, and progressive ME predicted a more severe progression to knee OA in the CT and RR groups. LEVEL OF EVIDENCE II.
Collapse
Affiliation(s)
- Lika Dzidzishvili
- Department of Orthopaedic Surgery and Traumatology, Hospital Universitario Fundación Jiménez Díaz, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Av. De los Reyes Católicos, 2, 28040 Madrid, Spain.
| | | | - David Moreno Molera
- Bioimaging Research Support Center- Universidad Complutense Madrid, Paseo Juan XXIII, 1, 28040 Madrid, Spain
| | - Emilio Calvo
- Department of Orthopaedic Surgery and Traumatology, Hospital Universitario Fundación Jiménez Díaz, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Av. De los Reyes Católicos, 2, 28040 Madrid, Spain
| | - Irene Isabel López-Torres
- Department of Orthopaedic Surgery and Traumatology, Hospital Universitario Fundación Jiménez Díaz, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Av. De los Reyes Católicos, 2, 28040 Madrid, Spain
| |
Collapse
|
9
|
Chapman JH, Ghosh D, Attari S, Ude CC, Laurencin CT. Animal Models of Osteoarthritis: Updated Models and Outcome Measures 2016-2023. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:127-146. [PMID: 38983776 PMCID: PMC11233113 DOI: 10.1007/s40883-023-00309-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 07/11/2024]
Abstract
Purpose Osteoarthritis (OA) is a global musculoskeletal disorder that affects primarily the knee and hip joints without any FDA-approved disease-modifying therapies. Animal models are essential research tools in developing therapies for OA; many animal studies have provided data for the initiation of human clinical trials. Despite this, there is still a need for strategies to recapitulate the human experience using animal models to better develop treatments and understand pathogenesis. Since our last review on animal models of osteoarthritis in 2016, there have been exciting updates in OA research and models. The main purpose of this review is to update the latest animal models and key features of studies in OA research. Method We used our existing classification method and screened articles in PubMed and bibliographic search for animal OA models between 2016 and 2023. Relevant and high-cited articles were chosen for inclusion in this narrative review. Results Recent studies were analyzed and classified. We also identified ex vivo models as an area of ongoing research. Each animal model offers its own benefit in the study of OA and there are a full range of outcome measures that can be assessed. Despite the vast number of models, each has its drawbacks that have limited translating approved therapies for human use. Conclusion Depending on the outcome measures and objective of the study, researchers should pick the best model for their work. There have been several exciting studies since 2016 that have taken advantage of regenerative engineering techniques to develop therapies and better understand OA. Lay Summary Osteoarthritis (OA) is a chronic debilitating disease without any cure that affects mostly the knee and hip joints and often results in surgical joint replacement. Cartilage protects the joint from mechanical forces and degrades with age or in response to injury. The many contributing causes of OA are still being investigated, and animals are used for preclinical research and to test potential new treatments. A single consensus OA animal model for preclinical studies is non-existent. In this article, we review the many animal models for OA and provide a much-needed update on studies and model development since 2016.
Collapse
Affiliation(s)
- James H. Chapman
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Debolina Ghosh
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Seyyedmorteza Attari
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Chinedu C. Ude
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, 263 Farmington Avenue, Farmington, CT 06030-3711, USA
- Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, UConn Health, Farmington, CT 06030, USA
- Department of Orthopedic Surgery, UConn Health, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical and Bimolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
10
|
Liu X, Chen R, Song Z, Sun Z. Exercise following joint distraction inhibits muscle wasting and delays the progression of post-traumatic osteoarthritis in rabbits by activating PGC-1α in skeletal muscle. J Orthop Surg Res 2024; 19:325. [PMID: 38822418 PMCID: PMC11141044 DOI: 10.1186/s13018-024-04803-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/21/2024] [Indexed: 06/03/2024] Open
Abstract
OBJECTIVE Muscle wasting frequently occurs following joint trauma. Previous research has demonstrated that joint distraction in combination with treadmill exercise (TRE) can mitigate intra-articular inflammation and cartilage damage, consequently delaying the advancement of post-traumatic osteoarthritis (PTOA). However, the precise mechanism underlying this phenomenon remains unclear. Hence, the purpose of this study was to examine whether the mechanism by which TRE following joint distraction delays the progression of PTOA involves the activation of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), as well as its impact on muscle wasting. METHODS Quadriceps samples were collected from patients with osteoarthritis (OA) and normal patients with distal femoral fractures, and the expression of PGC-1α was measured. The hinged external fixator was implanted in the rabbit PTOA model. One week after surgery, a PGC-1α agonist or inhibitor was administered for 4 weeks prior to TRE. Western blot analysis was performed to detect the expression of PGC-1α and Muscle atrophy gene 1 (Atrogin-1). We employed the enzyme-linked immunosorbent assay (ELISA) technique to examine pro-inflammatory factors. Additionally, we utilized quantitative real-time polymerase chain reaction (qRT-PCR) to analyze genes associated with cartilage regeneration. Synovial inflammation and cartilage damage were evaluated through hematoxylin-eosin staining. Furthermore, we employed Masson's trichrome staining and Alcian blue staining to analyze cartilage damage. RESULTS The decreased expression of PGC-1α in skeletal muscle in patients with OA is correlated with the severity of OA. In the rabbit PTOA model, TRE following joint distraction inhibited the expressions of muscle wasting genes, including Atrogin-1 and muscle ring finger 1 (MuRF1), as well as inflammatory factors such as interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) in skeletal muscle, potentially through the activation of PGC-1α. Concurrently, the production of IL-1β, IL-6, TNF-α, nitric oxide (NO), and malondialdehyde (MDA) in the synovial fluid was down-regulated, while the expression of type II collagen (Col2a1), Aggrecan (AGN), SRY-box 9 (SOX9) in the cartilage, and superoxide dismutase (SOD) in the synovial fluid was up-regulated. Additionally, histological staining results demonstrated that TRE after joint distraction reduced cartilage degeneration, leading to a significant decrease in OARSI scores.TRE following joint distraction could activate PGC-1α, inhibit Atrogin-1 expression in skeletal muscle, and reduce C-telopeptides of type II collagen (CTX-II) in the blood compared to joint distraction alone. CONCLUSION Following joint distraction, TRE might promote the activation of PGC-1α in skeletal muscle during PTOA progression to exert anti-inflammatory effects in skeletal muscle and joint cavity, thereby inhibiting muscle wasting and promoting cartilage regeneration, making it a potential therapeutic intervention for treating PTOA.
Collapse
Affiliation(s)
- Xinghui Liu
- School of Basic Medical Sciences, Hubei University of Arts and Science, Xiangyang, Hubei, 441000, China
| | - Rong Chen
- Department of Traumatic Orthopedics, Xiangyang Hospital of Traditional Chinese Medicine (Xiangyang Institute of Traditional Chinese Medicine), No. 24 Changzheng Road, Xiangyang, Hubei, 441001, China
| | - Zhenfei Song
- Department of Traumatic Orthopedics, Xiangyang Hospital of Traditional Chinese Medicine (Xiangyang Institute of Traditional Chinese Medicine), No. 24 Changzheng Road, Xiangyang, Hubei, 441001, China
| | - Zhibo Sun
- Department of Traumatic Orthopedics, Xiangyang Hospital of Traditional Chinese Medicine (Xiangyang Institute of Traditional Chinese Medicine), No. 24 Changzheng Road, Xiangyang, Hubei, 441001, China.
| |
Collapse
|
11
|
Li J, Zhang F, Ga X, Gao G, Guo T. Total meniscus replacement with a 3D printing of network hydrogel composite scaffold in a rabbit model. Knee Surg Sports Traumatol Arthrosc 2024; 32:1187-1198. [PMID: 38506124 DOI: 10.1002/ksa.12139] [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: 09/28/2023] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 03/21/2024]
Abstract
PURPOSE The aim of this study was to evaluate the role of a novel total meniscal implant in promoting meniscal regeneration and protecting articular cartilage in a rabbit model for 3 and 6 months. METHODS Thirty-six New Zealand rabbits were selected and divided into poly(ɛ-caprolactone) (PG-Pg) scaffold group, meniscectomy group and sham group. In this study, it was investigated whether PG-Pg scaffold can prevent articular cartilage degeneration and promote tissue degeneration, and its mechanical properties at 3 and 6 months after surgery were also explored. RESULT The degree of articular cartilage degeneration was significantly lower in the PG-Pg scaffold group than in the meniscectomy group. The number of chondrocytes increased in the PG-Pg scaffold at 3 and 6 months, while a gradual increase in the mechanical properties of the PG-Pg stent was observed from 6 months. CONCLUSION The PG-Pg scaffold slows down the degeneration of articular cartilage, promotes tissue regeneration and improves biomechanical properties after meniscectomy. This novel meniscus scaffold holds promise for enhancing surgical strategies and delivering superior long-term results for individuals with severe meniscus tears. LEVEL OF EVIDENCE NA.
Collapse
Affiliation(s)
- Jiaran Li
- Guizhou Provincial People's Hospital, Guiyang, China
- The Third Affiliated Hospital of Xinxiang Medical College, Xinxiang, China
| | | | - Xisijia Ga
- Guizhou Provincial People's Hospital, Guiyang, China
| | - Gan Gao
- Guizhou Provincial People's Hospital, Guiyang, China
| | - Tao Guo
- Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
12
|
Adam AO, Benea HRC, Fotescu HM, Alcalá Ruiz M, Cimpean GC, Ciornei V, Cernacovschi A, Edves AR, Crisan M. Recent Trends in Adipose Tissue-Derived Injectable Therapies for Osteoarthritis: A Scoping Review of Animal Models. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:707. [PMID: 38792890 PMCID: PMC11123108 DOI: 10.3390/medicina60050707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024]
Abstract
Background and Objectives: This scoping review investigates recent trends in adipose tissue-derived injectable therapies for osteoarthritis (OA) in animal models, focusing on minimally manipulated or lightly processed adipose tissue. By evaluating and examining the specific context in which these therapies were investigated across diverse animal OA models, this review aims to provide valuable insights that will inform and guide future research and clinical applications in the ongoing pursuit of effective treatments for osteoarthritis. Materials and Methods: This research conducted a comprehensive literature review of PubMed and Embase to determine studies about minimally manipulated adipose tissue-derived injectable therapies for osteoarthritis investigated using animal models. The primary search found 530 results. After excluding articles that focused on spontaneous osteoarthritis; on transfected, preconditioned, cultured, or co-cultured adipose-derived stem cells; and articles with unavailable full text, we included 11 articles in our review. Results: The examined therapies encompassed mechanical micro-fragmented adipose tissue (MFAT) and stromal vascular fraction (SVF) obtained via collagenase digestion and centrifugation. These interventions were evaluated across various animal models, including mice, rats, rabbits, and sheep with induced OA. Notably, more studies concentrated on surgically induced OA rather than chemically induced OA. The assessment of these therapies focused on elucidating their protective immunomodulatory, anti-inflammatory, and chondroregenerative potential through comprehensive evaluations, including macroscopic assessments, histological analyses, immunohistochemical examinations, and biochemical assays. Conclusions: This review provides a comprehensive analysis of adipose tissue-derived injectable therapies for osteoarthritis across diverse animal models. While revealing potential benefits and insights, the heterogeneity of data and the limited number of studies highlight the need for further research to formulate conclusive recommendations for clinical applications.
Collapse
Affiliation(s)
- Alina Otilia Adam
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Horea Rares Ciprian Benea
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Horia Mihnea Fotescu
- Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Miriam Alcalá Ruiz
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - George Claudiu Cimpean
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Vladimir Ciornei
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Arsenii Cernacovschi
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Andrei Rares Edves
- Department of Orthopedics and Traumatology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400132 Cluj-Napoca, Romania; (A.O.A.)
| | - Maria Crisan
- Department of Histology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| |
Collapse
|
13
|
Suderman R, Hurtig M, Grynpas M, Kuzyk P, Changoor A. Effect of Press-Fit Size on Insertion Mechanics and Cartilage Viability in Human and Ovine Osteochondral Grafts. Cartilage 2024:19476035241247297. [PMID: 38651510 PMCID: PMC11569632 DOI: 10.1177/19476035241247297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/25/2024] Open
Abstract
OBJECTIVE The osteochondral allograft procedure uses grafts constructed larger than the recipient site to stabilize the graft, in what is known as the press-fit technique. This research aims to characterize the relationships between press-fit size, insertion forces, and cell viability in ovine and human osteochondral tissue. DESIGN Human (4 donors) and ovine (5 animals) articular joints were used to harvest osteochondral grafts (4.55 mm diameter, N = 33 Human, N = 35 Ovine) and create recipient sites with grafts constructed to achieve varying degrees of press fit (0.025-0.240 mm). Donor grafts were inserted into recipient sites while insertion forces were measured followed by quantification of chondrocyte viability and histological staining to evaluate the extracellular matrix. RESULTS Both human and ovine tissues exhibited similar mechanical and cellular responses to changes in press-fit. Insertion forces (Human: 3-169 MPa, Ovine: 36-314 MPa) and cell viability (Human: 16%-89% live, Ovine: 2%-76% live) were correlated to press-fit size for both human (force: r = 0.539, viability: r = -0.729) and ovine (force: r = 0.655, viability: r = -0.714) tissues. In both species, a press-fit above 0.14 mm resulted in reduced cell viability below a level acceptable for transplantation, increased insertion forces, and reduced linear correlation to press-fit size compared to samples with a press-fit below 0.14 mm. CONCLUSIONS Increasing press-fit size required increased insertion forces and resulted in reduced cell viability. Ovine and human osteochondral tissues responded similarly to impact insertion and varying press-fit size, providing evidence for the use of the ovine model in allograft-related research.
Collapse
Affiliation(s)
- R.P. Suderman
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Kierans Janigan Biomechanics Research Program, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - M.B. Hurtig
- Comparative Orthopaedic Research Laboratory, Department of Clinical Studies, University of Guelph, Guelph, ON, Canada
| | - M.D. Grynpas
- Kierans Janigan Biomechanics Research Program, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Material Science & Engineering, University of Toronto, Toronto, ON, Canada
| | - P.R.T. Kuzyk
- Kierans Janigan Biomechanics Research Program, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - A. Changoor
- Kierans Janigan Biomechanics Research Program, Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Material Science & Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Fainor M, Orozco BS, Muir VG, Mahindroo S, Gupta S, Mauck RL, Burdick JA, Smith HE, Gullbrand SE. Mechanical crosstalk between the intervertebral disc, facet joints, and vertebral endplate following acute disc injury in a rabbit model. JOR Spine 2023; 6:e1287. [PMID: 38156057 PMCID: PMC10751980 DOI: 10.1002/jsp2.1287] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/21/2023] [Accepted: 09/06/2023] [Indexed: 12/30/2023] Open
Abstract
Background Vertebral endplate sclerosis and facet osteoarthritis have been documented in animals and humans. However, it is unclear how these adjacent pathologies engage in crosstalk with the intervertebral disc. This study sought to elucidate this crosstalk by assessing each compartment individually in response to acute disc injury. Methods Eleven New Zealand White rabbits underwent annular disc puncture using a 16G or 21G needle. At 4 and 10 weeks, individual compartments of the motion segment were analyzed. Discs underwent T 1 relaxation mapping with MRI contrast agent gadodiamide as well T 2 mapping. Both discs and facets underwent mechanical testing via vertebra-disc-vertebra tension-compression creep testing and indentation testing, respectively. Endplate bone density was quantified via μCT. Discs and facets were sectioned and stained for histology scoring. Results Intervertebral discs became more degenerative with increasing needle diameter and time post-puncture. Bone density also increased in endplates adjacent to both 21G and 16G punctured discs leading to reduced gadodiamide transport at 10 weeks. The facet joints, however, did not follow this same trend. Facets adjacent to 16G punctured discs were less degenerative than facets adjacent to 21G punctured discs at 10 weeks. 16G facets were more degenerative at 4 weeks than at 10, suggesting the cartilage had recovered. The formation of severe disc osteophytes in 16G punctured discs between 4 and 10 weeks likely offloaded the facet cartilage, leading to the recovery observed. Conclusions Overall, this study supports that degeneration spans the whole spinal motion segment following disc injury. Vertebral endplate thickening occurred in response to disc injury, which limited the diffusion of small molecules into the disc. This work also suggests that altered disc mechanics can induce facet degeneration, and that extreme bony remodeling adjacent to the disc may promote facet cartilage recovery through offloading of the articular cartilage.
Collapse
Affiliation(s)
- Matthew Fainor
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Brianna S. Orozco
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Victoria G. Muir
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sonal Mahindroo
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
- Department of BiologySt. Bonaventure UniversitySt. BonaventureNew YorkUSA
| | - Sachin Gupta
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Robert L. Mauck
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jason A. Burdick
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- BioFrontiers Institute and Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderColoradoUSA
| | - Harvey E. Smith
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| | - Sarah E. Gullbrand
- Department of Orthopaedic Surgery, McKay Orthopaedic Research Laboratory, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Translational Musculoskeletal Research CenterCorporal Michael J. Crescenz VA Medical CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
15
|
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: 4.0] [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
|
16
|
Anjiki K, Matsumoto T, Kuroda Y, Fujita M, Hayashi S, Nakano N, Tsubosaka M, Kamenaga T, Takashima Y, Kikuchi K, Ikuta K, Onoi Y, Tachibana S, Suda Y, Wada K, Matsushita T, Kuroda R. Heterogeneous Cells as well as Adipose-Derived Stromal Cells in Stromal Vascular Fraction Contribute to Enhance Anabolic and Inhibit Catabolic Factors in Osteoarthritis. Stem Cell Rev Rep 2023; 19:2407-2419. [PMID: 37477775 DOI: 10.1007/s12015-023-10589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2023] [Indexed: 07/22/2023]
Abstract
The stromal-vascular fraction (SVF), comprising heterogeneous cell populations and adipose-derived stromal cells (ADSCs), has therapeutic potential against osteoarthritis (OA); however, the underlying mechanism remains elusive. This study aimed to investigate the therapeutic effects of heterogeneous cells in rabbit SVF on rabbit chondrocytes. Rabbit SVF and ADSCs were autografted into knees at OA onset. The SVF (1 × 105) and low-dose ADSCs (lADSC; 1 × 104) groups adjusted for their stromal cell content were compared. Animals were euthanized 8 and 12 weeks after OA onset for macroscopic and histological analyses of OA progression and synovitis. Immunohistochemical and real-time polymerase chain reaction assessments were conducted. In vitro, immune-fluorescent double staining was performed for SVF to stain macrophages with F4/80, CD86(M1), and CD163(M2). OA progression was markedly suppressed, and synovitis was reduced in the SVF groups (OARSI histological score 8 W: 6.8 ± 0.75 vs. 3.8 ± 0.75, p = 0.001; 12 W: 8.8 ± 0.4 vs. 5.4 ± 0.49, p = 0.0002). The SVF groups had higher expression of collagen II and SOX9 in cartilage and TGF-β and IL-10 in the synovium, lower expression of MMP-13, and lower macrophage M1/M2 ratio than the lADSC groups. Immunofluorescent double staining revealed a markedly higher number of M2 than that of M1 macrophages in the SVF. The therapeutic effects of SVF on chondrocytes were superior than those of lADSCs, with enhanced anabolic and inhibited catabolic factors. Heterogeneous cells, mainly M2 macrophages in the SVF, enhanced growth factor secretion and chondrocyte-protective cytokines, thus benefiting chondrocytes and knee joint homeostasis. Overall, the SVF is a safe, relatively simple, and a useful treatment option for OA.
Collapse
Affiliation(s)
- Kensuke Anjiki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masahiro Fujita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yoshinori Takashima
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kenichi Kikuchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kenmei Ikuta
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Yoshihito Suda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kensuke Wada
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takehiko Matsushita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1, Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
17
|
Wen S, Iturriaga V, Vásquez B, del Sol M. Comparison of Four Treatment Protocols with Intra-Articular Medium Molecular Weight Hyaluronic Acid in Induced Temporomandibular Osteoarthritis: An Experimental Study. Int J Mol Sci 2023; 24:14130. [PMID: 37762430 PMCID: PMC10531553 DOI: 10.3390/ijms241814130] [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: 08/14/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
The aim was to compare the effect between a single intra-articular infiltration (1i) and two infiltrations (2i) of medium molecular weight hyaluronic acid (MMW-HA) of high viscosity (HV) and low viscosity (LV) on the histopathological characteristics of temporomandibular joint (TMJ) osteoarthritis (OA) induced in rabbits. An experimental study was conducted on Oryctolagus cuniculus rabbits, including 42 TMJs, distributed between (1) TMJ-C, control group; (2) TMJ-OA, group with OA; (3) TMJ-OA-wt, group with untreated OA; (4) group treated with HA-HV-1i; (5) group treated with HA-HV-2i; (6) group treated with HA-LV-1i; and (7) group treated with HA-LV-2i. The results were evaluated using the Osteoarthritis Research Society International (OARSI) scale and descriptive histology considering the mandibular condyle (MC), the articular disc (AD), and the mandibular fossa (MF). The Kruskal-Wallis test was used for the statistical analysis, considering p < 0.05 significant. All treated groups significantly decreased the severity of OA compared to the TMJ-OA-wt group. The HA-HV-2i group showed significant differences in the degree of OA from the TMJ-OA group. The degree of OA in the HA-HV-2i group was significantly lower than in the HA-LV-1i, HA-LV-2i, and HA-HV-1i groups. The protocol that showed better results in repairing the joint was HA-HV-2i. There are histological differences depending on the protocol of the preparation used: two infiltrations seem to be better than one, and when applying two doses, high viscosity shows better results.
Collapse
Affiliation(s)
- Schilin Wen
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile;
- Grupo de Investigación de Pregrado en Odontología, Facultad de Ciencias de la Salud (FACSA), Universidad Autónoma de Chile, Temuco 4810101, Chile
| | - Veronica Iturriaga
- Temporomandibular Disorder and Orofacial Pain Program, Department of Integral Adult Care Dentistry, Universidad de La Frontera, Temuco 4780000, Chile;
- Sleep & Pain Research Group, Faculty of Dentistry, Universidad de La Frontera, Temuco 4780000, Chile
- Center of Excellence in Morphological and Surgical Studies (CEMyQ), Universidad de La Frontera, Temuco 4780000, Chile
| | - Bélgica Vásquez
- Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile;
| | - Mariano del Sol
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4780000, Chile;
- Center of Excellence in Morphological and Surgical Studies (CEMyQ), Universidad de La Frontera, Temuco 4780000, Chile
| |
Collapse
|
18
|
Ruscitto A, Chen P, Tosa I, Wang Z, Zhou G, Safina I, Wei R, Morel MM, Koch A, Forman M, Reeve G, Lecholop MK, Wilson M, Bonthius D, Chen M, Ono M, Wang TC, Yao H, Embree MC. Lgr5-expressing secretory cells form a Wnt inhibitory niche in cartilage critical for chondrocyte identity. Cell Stem Cell 2023; 30:1179-1198.e7. [PMID: 37683603 PMCID: PMC10790417 DOI: 10.1016/j.stem.2023.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 06/06/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023]
Abstract
Osteoarthritis is a degenerative joint disease that causes pain, degradation, and dysfunction. Excessive canonical Wnt signaling in osteoarthritis contributes to chondrocyte phenotypic instability and loss of cartilage homeostasis; however, the regulatory niche is unknown. Using the temporomandibular joint as a model in multiple species, we identify Lgr5-expressing secretory cells as forming a Wnt inhibitory niche that instruct Wnt-inactive chondroprogenitors to form the nascent synovial joint and regulate chondrocyte lineage and identity. Lgr5 ablation or suppression during joint development, aging, or osteoarthritis results in depletion of Wnt-inactive chondroprogenitors and a surge of Wnt-activated, phenotypically unstable chondrocytes with osteoblast-like properties. We recapitulate the cartilage niche and create StemJEL, an injectable hydrogel therapy combining hyaluronic acid and sclerostin. Local delivery of StemJEL to post-traumatic osteoarthritic jaw and knee joints in rabbit, rat, and mini-pig models restores cartilage homeostasis, chondrocyte identity, and joint function. We provide proof of principal that StemJEL preserves the chondrocyte niche and alleviates osteoarthritis.
Collapse
Affiliation(s)
- Angela Ruscitto
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Peng Chen
- Clemson University-Medical University of South Carolina Joint Bioengineering Program, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ikue Tosa
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ziyi Wang
- Department of Molecular Biology and Biochemistry, Okayama University Graduate, School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 7008525, Japan
| | - Gan Zhou
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ingrid Safina
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ran Wei
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mallory M Morel
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Alia Koch
- Section of Hospital Dentistry, Division of Oral & Maxillofacial Surgery, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Michael Forman
- Section of Hospital Dentistry, Division of Oral & Maxillofacial Surgery, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Gwendolyn Reeve
- Division of Oral and Maxillofacial Surgery, New York Presbyterian Weill Cornell Medicine, New York, NY 10065, USA
| | - Michael K Lecholop
- Department of Oral and Maxillofacial Surgery, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Marshall Wilson
- Clemson University-Medical University of South Carolina Joint Bioengineering Program, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Daniel Bonthius
- Clemson University-Medical University of South Carolina Joint Bioengineering Program, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mo Chen
- Wnt Scientific, LLC, Harlem Biospace, New York, NY 10027, USA
| | - Mitsuaki Ono
- Department of Molecular Biology and Biochemistry, Okayama University Graduate, School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 7008525, Japan; Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 7008525, Japan
| | - Timothy C Wang
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Digestive and Liver Diseases, Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Hai Yao
- Clemson University-Medical University of South Carolina Joint Bioengineering Program, Department of Bioengineering, Clemson University, Clemson, SC 29634, USA; Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mildred C Embree
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA; Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
19
|
Dzidzishvili L, Calvo E, López-Torres II. Medial Meniscus Posterior Root Repair Reduces but Does Not Avoid Histologic Progression of Osteoarthritis: Randomized In Vivo Experimental Study in a Rabbit Model. Am J Sports Med 2023; 51:2964-2974. [PMID: 37589243 DOI: 10.1177/03635465231188527] [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] [Indexed: 08/18/2023]
Abstract
BACKGROUND The optimal treatment option for meniscus root tears is still challenging, and whether the meniscus root repair ultimately can arrest or delay osteoarthritic changes is still a concern. PURPOSE/HYPOTHESIS The purpose of this study was 2-fold: (1) to describe and compare histopathologic findings of 3 different therapeutic options for medial meniscus posterior root tear: nonoperative management, partial meniscectomy, and meniscus root repair; and (2) to test the hypothesis that meniscus root tears treated nonoperatively predispose to a lower risk of osteoarthritic progression compared with partial meniscectomy. STUDY DESIGN Controlled laboratory study. METHODS Posteromedial meniscus root tears were carried out in 39 New Zealand White rabbits. Animals were randomly assigned into 3 experimental groups: partial meniscectomy after root tear (PM; n = 13), root tears treated conservatively (CT; n = 13), and transtibial root repair (RR; n = 13). Contralateral limbs were used as healthy controls. The animals were euthanized at 16 weeks postoperatively; tissue samples of femoral and tibial articular cartilage were collected and processed for macro- and microscopic assessment to detect signs of early osteoarthritis (OA). Each sample was histopathologically assessed using the Osteoarthritis Research Society International grading and staging system. RESULTS Osteoarthritic changes were the hallmark in all 3 experimental groups. The RR group had the lowest scores for cartilage damage (mean, 2.5; range, 2-3), and the PM group exhibited higher and more severe signs of OA (mean, 16; range, 9-16) compared with the CT group (mean, 5; range, 4-6). The between-group comparison revealed significant differences, as the PM group showed a significantly higher rate of macro- and microscopic osteoarthritic changes compared with the RR (P < .001) and CT (P < .001) groups. The weightbearing area of the medial femoral condyle was the most severely affected, and tidemark disruption was evident in all tissue samples. CONCLUSION Meniscus root repair cannot completely arrest the histopathologic progression of knee OA but leads to significantly less severe degenerative changes than partial meniscectomy and nonoperative treatment. Partial meniscectomy leads to the most severe osteoarthritic progression, while stable radial tears left in situ have lower progression compared with partial meniscectomy. CLINICAL RELEVANCE Histologic assessment is an essential tool and metric for guiding and understanding osteoarthritic features, providing insight into the disease development and progression. This study provides histopathologic evidence on osteoarthritic progression after medial meniscus posterior root repair. This knowledge can help to set more realistic expectations and can lead to the future development of augmented techniques.
Collapse
Affiliation(s)
- Lika Dzidzishvili
- Department of Orthopaedic Surgery and Traumatology, Hospital Universitario Fundación Jiménez Díaz, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Emilio Calvo
- Department of Orthopaedic Surgery and Traumatology, Hospital Universitario Fundación Jiménez Díaz, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Irene Isabel López-Torres
- Department of Orthopaedic Surgery and Traumatology, Hospital Universitario Fundación Jiménez Díaz, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
20
|
Mert L, Bilgiç B, Şenol BK, Zülfikar OB, Durmaz H, Polat G. What is the Effect of Bevacizumab on Cartilage and Synovium in a Rabbit Model of Hemophilic Arthropathy? Clin Orthop Relat Res 2023; 481:1634-1647. [PMID: 37036937 PMCID: PMC10344489 DOI: 10.1097/corr.0000000000002628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/27/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Hemophilic arthropathy can cause recurrent hemarthroses and severe damage to the synovium and articular cartilage. Previous studies have shown that vascular endothelial growth factor (VEGF) plays an essential role in neoangiogenesis. Bevacizumab, a monoclonal VEGF inhibitor, is used clinically to prevent angiogenesis. However, its effects on hemophilic arthropathy are unknown. QUESTIONS/PURPOSES Using a hemophilic arthropathy rabbit model, we asked: Does an intra-articular injection of bevacizumab (1) inhibit VEGF, (2) decrease signal intensity in dynamic contrast-enhanced MRI (DCE-MRI) as an assessment of capillary permeability and neoangiogenesis, (3) reduce cartilage damage, (4) reduce synovial changes, and (5) affect macroscopic changes during the development of hemophilic arthropathy? METHODS Twenty-five male New Zealand rabbits were divided into four groups. Eight knees from four rabbits were used as the control group. We used an established animal model for hemophilic arthropathy in the remaining 21 rabbits. Animals were assigned randomly to three groups with seven rabbits in each group. One group was used to establish mild arthropathy, and the other two were used to establish severe arthropathy. Autologous blood from the rabbits' ears was injected into the right and left knees twice per week for 8 weeks to represent mild arthropathy and for 16 weeks to represent severe arthropathy. In the mild arthropathy group, bevacizumab was injected into the right knee once every 2 weeks. Bevacizumab was injected into the right knee of rabbits in one of the severe arthropathy groups once every 2 weeks for 16 weeks, and intra-articular bevacizumab injections were administered to the right knees of rabbits in the other severe arthropathy group once every 2 weeks after the eighth week. An equal volume of 0.9% saline was injected into the left knee of rabbits in all arthropathy groups. To explore the efficacy of bevacizumab, joint diameters were quantitatively measured, and cartilage and synovial changes were examined. Degeneration of articular cartilage was evaluated with the semiquantitative Osteoarthritis Research Society International grading system. Synovial damage was analyzed with a semiquantitative microscopic scoring system. In addition, we evaluated perfusion and angiogenesis using DCE-MRI (quantitative signal intensity changes). Immunohistochemical testing was used to measure VEGF levels (analyzed by Western blotting). RESULTS Intra-articular bevacizumab treatment inhibited VEGF in our rabbit model of hemophilic arthropathy. VEGF protein expression levels were lower in the mild arthropathy group that received intra-articular bevacizumab (0.89 ± 0.45) than the mild arthropathy control group (1.41 ± 0.61) (mean difference -0.52 [95% CI -0.898 to -0.143]; p = 0.02). VEGF levels were lower in the severe arthropathy group that received treatment for 16 weeks (0.94 ± 0.27) than in the control knees (1.49 ± 0.36) (mean difference -0.55 [95% CI -0.935 to -0.161]; p = 0.01). In the severe arthropathy group, the Osteoarthritis Research Society International score indicating cartilage damage was lower in the group that received intra-articular bevacizumab treatment from the beginning than in the control group (median 17 [range 13 to 18] versus 18 [range 17 to 20]; difference of medians 1; p = 0.02). Additionally, the scores indicated synovial damage was lower in the group that received intra-articular bevacizumab treatment from the beginning than the control group (median 5 [range 4 to 9] versus 9 [range 8 to 12]; difference of medians 4; p = 0.02). The mean of mean values for signal intensity changes was higher in the nontreated severe groups than in the group of healthy knees. The signal intensity changes were higher in the severe arthropathy control groups (Groups BC and CC) (median 311.6 [range 301.4 to 361.2] and 315.1 [range 269.7 to 460.4]) than in the mild arthropathy control group (Group AC) (median 234.1 [range 212.5 to 304.2]; difference of medians 77.5 and 81, respectively; p = 0.02 and p = 0.04, respectively). In the severe arthropathy group, discoloration caused by hemosiderin deposition in the cartilage and synovium was more pronounced than in the mild arthropathy group. In the severe arthropathy group treated with intra-articular bevacizumab, joint diameters were smaller than in the control group (Group BT median 12.7 mm [range 12.3 to 14.0] versus Group BC median 14.0 mm [range 13.1 to 14.5]; difference of medians 1.3 mm; p = 0.02). CONCLUSION Hemarthrosis damages the synovial tissues and cartilage in the knees of rabbits, regardless of whether they are treated with intra-articular bevacizumab. However, intra-articular injection of bevacizumab may reduce cartilage and synovial damage in rabbits when treatment is initiated early during the development of hemophilic arthropathy. CLINICAL RELEVANCE If the findings in this study are replicated in larger-animal models that consider the limitations of our work, then a trial in humans might be appropriate to ascertain whether intra-articular injection of bevacizumab could reduce cartilage damage and synovial changes in patients with hemophilia whose hemarthroses cannot otherwise be controlled.
Collapse
Affiliation(s)
- Lezgin Mert
- Department of Orthopedics and Traumatology, Istanbul University, İstanbul Faculty of Medicine, İstanbul, Turkey
| | - Bilge Bilgiç
- Department of Pathology, Istanbul University, İstanbul Faculty of Medicine, İstanbul, Turkey
| | - Başak Koç Şenol
- Department of Pediatric Hematology-Oncology, İstanbul University, Oncology Institute, İstanbul, Turkey
| | - Osman Bülent Zülfikar
- Department of Pediatric Hematology-Oncology, İstanbul University, Oncology Institute, İstanbul, Turkey
| | - Hayati Durmaz
- Department of Orthopedics and Traumatology, Istanbul University, İstanbul Faculty of Medicine, İstanbul, Turkey
| | - Gökhan Polat
- Department of Orthopedics and Traumatology, Istanbul University, İstanbul Faculty of Medicine, İstanbul, Turkey
| |
Collapse
|
21
|
Menges S, Michaelis M, Kleinschmidt-Dörr K. Anti-NGF treatment worsens subchondral bone and cartilage measures while improving symptoms in floor-housed rabbits with osteoarthritis. Front Physiol 2023; 14:1201328. [PMID: 37435308 PMCID: PMC10331818 DOI: 10.3389/fphys.2023.1201328] [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: 04/06/2023] [Accepted: 06/14/2023] [Indexed: 07/13/2023] Open
Abstract
Objective: Osteoarthritis (OA) is a common joint disorder often affecting the knee. It is characterized by alterations of various joint tissues including subchondral bone and by chronic pain. Anti-nerve growth factor (NGF) antibodies have demonstrated improvement in pain associated with OA in phase 3 clinical trials but have not been approved due to an increased risk of developing rapidly progressive OA. The aim of this study was to investigate effects of systemic anti-NGF-treatment on structure and symptoms in rabbits with surgically induced joint instability. Methods: This was elicited by anterior cruciate ligament transection and partial resection of the medial meniscus in right knee of 63 female rabbits, housed altogether in a 56 m2 floor husbandry. Rabbits received either 0.1, 1 or 3 mg/kg anti-NGF antibody intra-venously at weeks 1, 5 and 14 after surgery or vehicle. During in-life phase, static incapacitance tests were performed and joint diameter was measured. Following necropsy, gross morphological scoring and micro-computed tomography analysis of subchondral bone and cartilage were performed. Results: After surgery, rabbits unloaded operated joints, which was improved with 0.3 and 3 mg/kg anti-NGF compared to vehicle injection during the first half of the study. The diameter of operated knee joints increased over contralateral measures. This increase was bigger in anti-NGF treated rabbits beginning 2 weeks after the first IV injection and became dose-dependent and more pronounced with time. In the 3 mg/kg anti-NGF group, the bone volume fraction and trabecular thickness increased in the medio-femoral region of operated joints compared to contralateral and to vehicle-treated animals, while cartilage volume and to a lesser extent thickness decreased. Enlarged bony areas were found in right medio-femoral cartilage surfaces of animals receiving 1 and 3 mg/kg anti-NGF. Alterations of all structural parameters were particularly distinct in a subgroup of three rabbits, which also exhibited more prominent symptomatic improvement. Conclusion: This study showed that anti-NGF administration exerted negative impact on structure in destabilized joints of rabbits, while pain-induced unloading of joints was improved. Our findings open up the possibility to better understand the effects of systemic anti-NGF, particularly on subchondral bone, and thus the occurrence of rapidly progressive OA in patients.
Collapse
|
22
|
Shen P, Gao J, Huang S, You C, Wang H, Chen P, Yao T, Gao T, Zhou B, Shen S, Zhao X, Ma J. LncRNA AC006064.4-201 serves as a novel molecular marker in alleviating cartilage senescence and protecting against osteoarthritis by destabilizing CDKN1B mRNA via interacting with PTBP1. Biomark Res 2023; 11:39. [PMID: 37055817 PMCID: PMC10099822 DOI: 10.1186/s40364-023-00477-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/20/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is the most prevalent age-related disease in the world. Chondrocytes undergo an age-dependent decline in their proliferation and synthetic capacity, which is the main cause of OA development. However, the intrinsic mechanism of chondrocyte senescence is still unclear. This study aimed to investigate the role of a novel long non-coding RNA (lncRNA), AC006064.4-201 in the regulation of chondrocyte senescence and OA progression and to elucidate the underlying molecular mechanisms. METHODS The function of AC006064.4-201 in chondrocytes was assessed using western blotting, quantitative real-time polymerase chain reaction (qRT-PCR), immunofluorescence (IF) and β-galactosidase staining. The interaction between AC006064.4-201 and polypyrimidine tract-binding protein 1 (PTBP1), as well as cyclin-dependent kinase inhibitor 1B (CDKN1B), was evaluated using RPD-MS, fluorescence in situ hybridization (FISH), RNA immunoprecipitation (RIP) and RNA pull-down assays. Mice models were used to investigate the role of AC006064.4-201 in post-traumatic and age-related OA in vivo. RESULTS Our research revealed that AC006064.4-201 was downregulated in senescent and degenerated human cartilage, which could alleviate senescence and regulate metabolism in chondrocytes. Mechanically, AC006064.4-201 directly interacts with PTBP1 and blocks the binding between PTBP1 and CDKN1B mRNA, thereby destabilizing CDKN1B mRNA and decreasing the translation of CDKN1B. The in vivo experiments were consistent with the results of the in vitro experiments. CONCLUSIONS The AC006064.4-201/PTBP1/CDKN1B axis plays an important role in OA development and provides new molecular markers for the early diagnosis and treatment of OA in the future. Schematic diagram of AC006064.4-201 mechanism. A schematic diagram of the mechanism underlying the effect of AC006064.4-201.
Collapse
Affiliation(s)
- Panyang Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Jun Gao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Shaohan Huang
- Department of Endocrinology, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Chenan You
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Haitao Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Pengyu Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Teng Yao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Tianyou Gao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Bohao Zhou
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China
| | - Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
- Department of Endocrinology, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
| | - Xing Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
- Department of Endocrinology, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
| | - Jianjun Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University & Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
- Department of Endocrinology, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, China.
| |
Collapse
|
23
|
Ojanen SP, Finnilä MAJ, Herzog W, Saarakkala S, Korhonen RK, Rieppo L. Micro-computed Tomography-Based Collagen Orientation and Anisotropy Analysis of Rabbit Articular Cartilage. Ann Biomed Eng 2023:10.1007/s10439-023-03183-4. [PMID: 37005948 DOI: 10.1007/s10439-023-03183-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/27/2023] [Indexed: 04/04/2023]
Abstract
The collagen network is the highly organized backbone of articular cartilage providing tissue tensile stiffness and restricting proteoglycan bleaching out of the tissue. Osteoarthritis (OA) diminishes proper collagen network adaptation. Our aim was to provide quantitative three-dimensional (3D) information of the cartilage collagen network adaptation in early osteoarthritis using high resolution micro-computed tomography (µCT)-imaging. Osteochondral samples from the femoral condyles were collected from healthy (N = 8, both legs) and experimental OA rabbit model with anterior cruciate ligament transection (N = 14, single leg). Samples were processed for cartilage µCT-imaging and histological evaluation with polarized light microscopy (PLM). Structure tensor analysis was used to analyse the collagen fibre orientation and anisotropy of the µCT-images, and PLM was used as a validation for structural changes. Depth-wise comparison of collagen fibre orientation acquired with µCT-imaging and PLM correlated well, but the values obtained with PLM were systematically greater than those measured with µCT-imaging. Structure tensor analysis allowed for 3D quantification of collagen network anisotropy. Finally, µCT-imaging revealed only minor differences between the control and experimental groups.
Collapse
Affiliation(s)
- Simo P Ojanen
- Department of Technical Physics, University of Eastern Finland, P.O. Box 1627, 70210, Kuopio, Finland.
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland.
| | - Mikko A J Finnilä
- Department of Technical Physics, University of Eastern Finland, P.O. Box 1627, 70210, Kuopio, Finland
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | - Simo Saarakkala
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Rami K Korhonen
- Department of Technical Physics, University of Eastern Finland, P.O. Box 1627, 70210, Kuopio, Finland
| | - Lassi Rieppo
- Research Unit of Health Sciences and Technology, University of Oulu, Oulu, Finland
| |
Collapse
|
24
|
Orozco GA, Ristaniemi A, Haghighatnejad M, Mohammadi A, Finnilä MAJ, Saarakkala S, Herzog W, Isaksson H, Korhonen RK. Adaptation of Fibril-Reinforced Poroviscoelastic Properties in Rabbit Collateral Ligaments 8 Weeks After Anterior Cruciate Ligament Transection. Ann Biomed Eng 2023; 51:726-740. [PMID: 36129552 PMCID: PMC10023629 DOI: 10.1007/s10439-022-03081-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022]
Abstract
Ligaments of the knee provide stability and prevent excessive motions of the joint. Rupture of the anterior cruciate ligament (ACL), a common sports injury, results in an altered loading environment for other tissues in the joint, likely leading to their mechanical adaptation. In the collateral ligaments, the patterns and mechanisms of biomechanical adaptation following ACL transection (ACLT) remain unknown. We aimed to characterize the adaptation of elastic and viscoelastic properties of the lateral and medial collateral ligaments eight weeks after ACLT. Unilateral ACLT was performed in six rabbits, and collateral ligaments were harvested from transected and contralateral knee joints after eight weeks, and from an intact control group (eight knees from four animals). The cross-sectional areas were measured with micro-computed tomography. Stepwise tensile stress-relaxation testing was conducted up to 6% final strain, and the elastic and viscoelastic properties were characterized with a fibril-reinforced poroviscoelastic material model. We found that the cross-sectional area of the collateral ligaments in the ACL transected knees increased, the nonlinear elastic collagen network modulus of the LCL decreased, and the amount of fast relaxation in the MCL decreased. Our results indicate that rupture of the ACL leads to an early adaptation of the elastic and viscoelastic properties of the collagen fibrillar network in the collateral ligaments. These adaptations may be important to consider when evaluating whole knee joint mechanics after ACL rupture, and the results aid in understanding the consequences of ACL rupture on other tissues.
Collapse
Affiliation(s)
- Gustavo A Orozco
- Department of Applied Physics, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland.
- Department of Biomedical Engineering, Lund University, Box 188, 221 00, Lund, Sweden.
| | - Aapo Ristaniemi
- Department of Applied Physics, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
- AO Research Institute Davos, Davos, Switzerland
| | - Mehrnoush Haghighatnejad
- Department of Applied Physics, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| | - Ali Mohammadi
- Department of Applied Physics, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| | - Mikko A J Finnilä
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, Finland
| | - Simo Saarakkala
- Research Unit of Medical Imaging, Physics and Technology, University of Oulu, Oulu, Finland
- Department of Diagnostic Radiology, Oulu University Hospital, Oulu, Finland
| | - Walter Herzog
- Human Performance Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Canada
| | - Hanna Isaksson
- Department of Biomedical Engineering, Lund University, Box 188, 221 00, Lund, Sweden
| | - Rami K Korhonen
- Department of Applied Physics, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| |
Collapse
|
25
|
Nativel F, Smith A, Boulestreau J, Lépine C, Baron J, Marquis M, Vignes C, Le Guennec Y, Veziers J, Lesoeur J, Loll F, Halgand B, Renard D, Abadie J, Legoff B, Blanchard F, Gauthier O, Vinatier C, Rieux AD, Guicheux J, Le Visage C. Micromolding-based encapsulation of mesenchymal stromal cells in alginate for intraarticular injection in osteoarthritis. Mater Today Bio 2023; 19:100581. [PMID: 36896417 PMCID: PMC9988569 DOI: 10.1016/j.mtbio.2023.100581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/27/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Osteoarthritis (OA) is an inflammatory joint disease that affects cartilage, subchondral bone, and joint tissues. Undifferentiated Mesenchymal Stromal Cells are a promising therapeutic option for OA due to their ability to release anti-inflammatory, immuno-modulatory, and pro-regenerative factors. They can be embedded in hydrogels to prevent their tissue engraftment and subsequent differentiation. In this study, human adipose stromal cells are successfully encapsulated in alginate microgels via a micromolding method. Microencapsulated cells retain their in vitro metabolic activity and bioactivity and can sense and respond to inflammatory stimuli, including synovial fluids from OA patients. After intra-articular injection in a rabbit model of post-traumatic OA, a single dose of microencapsulated human cells exhibit properties matching those of non-encapsulated cells. At 6 and 12 weeks post-injection, we evidenced a tendency toward a decreased OA severity, an increased expression of aggrecan, and a reduced expression of aggrecanase-generated catabolic neoepitope. Thus, these findings establish the feasibility, safety, and efficacy of injecting cells encapsulated in microgels, opening the door to a long-term follow-up in canine OA patients.
Collapse
Affiliation(s)
- Fabien Nativel
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Audrey Smith
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France.,UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200, Bruxelles, Belgium
| | - Jeremy Boulestreau
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Charles Lépine
- Nantes Université, CHU Nantes, Department of Pathology, F-44000 Nantes, France
| | - Julie Baron
- Nantes Université, CHU Nantes, Department of Pathology, F-44000 Nantes, France
| | - Melanie Marquis
- UR1268 BIA (Biopolymères Interactions Assemblages), INRAE, F-44300 Nantes, France
| | - Caroline Vignes
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Yoan Le Guennec
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Joelle Veziers
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Julie Lesoeur
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - François Loll
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Boris Halgand
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Denis Renard
- UR1268 BIA (Biopolymères Interactions Assemblages), INRAE, F-44300 Nantes, France
| | - Jerome Abadie
- LabONIRIS, ONIRIS (Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering), F-44300 Nantes, France
| | - Benoit Legoff
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Frederic Blanchard
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Olivier Gauthier
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France.,ONIRIS Nantes-Atlantic College of Veterinary Medicine, Centre de Recherche et D'investigation Préclinique (CRIP), F-44300 Nantes, France
| | - Claire Vinatier
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Anne des Rieux
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200, Bruxelles, Belgium
| | - Jerome Guicheux
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Catherine Le Visage
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| |
Collapse
|
26
|
Torres-Torrillas M, Damia E, del Romero A, Pelaez P, Miguel-Pastor L, Chicharro D, Carrillo JM, Rubio M, Sopena JJ. Intra-osseous plasma rich in growth factors enhances cartilage and subchondral bone regeneration in rabbits with acute full thickness chondral defects: Histological assessment. Front Vet Sci 2023; 10:1131666. [PMID: 37065219 PMCID: PMC10095833 DOI: 10.3389/fvets.2023.1131666] [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: 12/25/2022] [Accepted: 03/13/2023] [Indexed: 04/18/2023] Open
Abstract
Background Intra-articular (IA) combined with intra-osseous (IO) infiltration of plasma rich in growth factors (PRGF) have been proposed as an alternative approach to treat patients with severe osteoarthritis (OA) and subchondral bone damage. The aim of the study is to evaluate the efficacy of IO injections of PRGF to treat acute full depth chondral lesion in a rabbit model by using two histological validated scales (OARSI and ICRS II). Methodology A total of 40 rabbits were included in the study. A full depth chondral defect was created in the medial femoral condyle and then animals were divided into 2 groups depending on the IO treatment injected on surgery day: control group (IA injection of PRGF and IO injection of saline) and treatment group (IA combined with IO injection of PRGF). Animals were euthanized 56 and 84 days after surgery and the condyles were processed for posterior histological evaluation. Results Better scores were obtained in treatment group in both scoring systems at 56- and 84-days follow-up than in control group. Additionally, longer-term histological benefits have been obtained in the treatment group. Conclusions The results suggests that IO infiltration of PRGF enhances cartilage and subchondral bone healing more than the IA-only PRGF infiltration and provides longer-lasting beneficial effects.
Collapse
Affiliation(s)
- Marta Torres-Torrillas
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Elena Damia
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Ayla del Romero
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Pau Pelaez
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Laura Miguel-Pastor
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Deborah Chicharro
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - José M. Carrillo
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Mónica Rubio
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| | - Joaquín J. Sopena
- Bioregenerative Medicine and Applied Surgery Research Group, Department of Animal Medicine and Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
- García Cugat Foundation CEU-UCH Chair of Medicine and Regenerative Surgery, CEU Cardenal Herrera University, CEU Universities, Valencia, Spain
| |
Collapse
|
27
|
Feng Z, Huang Q, Zhang X, Xu P, Li S, Ma D, Meng Q. PPAR-γ Activation Alleviates Osteoarthritis through Both the Nrf2/NLRP3 and PGC-1α/Δψm Pathways by Inhibiting Pyroptosis. PPAR Res 2023; 2023:2523536. [PMID: 37020714 PMCID: PMC10070030 DOI: 10.1155/2023/2523536] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/10/2023] [Accepted: 02/27/2023] [Indexed: 03/30/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease with a gradually increasing morbidity in the aging and obese population. Emerging evidence has implicated pyroptosis in the etiology of OA and it may be recognized as a therapeutic target in OA. We have previously reported regarding another disease that peroxisome proliferator-activated receptor gamma (PPAR-γ) activation exerts an anti-inflammatory effect by suppressing the nucleotide-binding and oligomerization domain-like receptor containing protein (NLRP) 3 inflammasome. However, the relationship between PPAR-γ and NLRP3-mediated pyroptosis in OA cartilage and its underlying mechanisms is fully unclear. In this study, we found that the level of NLRP3-mediated pyroptosis in severe lateral femoral condyle cartilage wear in the knee of an OA patient was significantly higher than that in the mild lateral femoral condyle cartilage wear areas. Moreover, in lipopolysaccharide (LPS)/adenosine triphosphate (ATP)-induced primary chondrocytes and knee OA rat models, we demonstrated that activation of PPAR-γ by pioglitazone (Piog) attenuated LPS/ATP-induced chondrocyte pyroptosis and arthritis. These effects were partially counteracted by either blocking the nuclear factor erythroid-2-related factor (Nrf2)/NLRP3 or PGC1-α/Δψm signaling pathway. Simultaneous depression of these two signaling pathways can completely abrogate the protective effects of Piog on OA and chondrocytes. Taken together, Piog protects OA cartilage against pyroptosis-induced damage by simultaneously activating both the Nrf2/NLRP3 and PGC-1α/Δψm pathways, which enhances antioxidative and anti-inflammatory responses as well as mitochondrial biogenesis. Therefore, Piog may be a promising agent for human OA cartilage damage in future clinical treatments.
Collapse
|
28
|
Zhao C, Sun G, Li Y, Kong K, Li X, Kan T, Yang F, Wang L, Wang X. Forkhead box O3 attenuates osteoarthritis by suppressing ferroptosis through inactivation of NF-κB/MAPK signaling. J Orthop Translat 2023; 39:147-162. [PMID: 37188001 PMCID: PMC10175709 DOI: 10.1016/j.jot.2023.02.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/03/2023] [Accepted: 02/20/2023] [Indexed: 05/17/2023] Open
Abstract
Background Ferroptosis is a nonapoptotic cell death process that is characterized by lipid peroxidation and intracellular iron accumulation. As osteoarthritis (OA) progresses, inflammation or iron overload induces ferroptosis of chondrocytes. However, the genes that play a vital role in this process are still poorly studied. Methods Ferroptosis was elicited in the ATDC5 chondrocyte cell line and primary chondrocytes by administration of the proinflammatory cytokines, interleukin (IL)-1β and tumor necrosis factor (TNF)-α, which play key roles in OA. The effect of FOXO3 expression on apoptosis, extracellular matrix (ECM) metabolism, and ferroptosis in ATDC5 cells and primary chondrocytes was verified by western blot, Immunohistochemistry (IMHC), immunofluorescence (IF) and measuring Malondialdehyde (MDA) and Glutathione (GSH) levels. The signal cascades that modulated FOXO3-mediated ferroptosis were identified by using chemical agonists/antagonists and lentivirus. In vivo experiments were performed following destabilization of medial meniscus surgery on 8-week-old C57BL/6 mice and included micro-computed tomography measurements. Results In vitro administration of IL-1β and TNF-α, to ATDC5 cells or primary chondrocytes induced ferroptosis. In addition, the ferroptosis agonist, erastin, and the ferroptosis inhibitor, ferrostatin-1, downregulated or upregulated the protein expression of forkhead box O3 (FOXO3), respectively. This, suggested, for the first time, that FOXO3 may regulate ferroptosis in articular cartilage. Our results further suggested that FOXO3 regulated ECM metabolism via the ferroptosis mechanism in ATDC5 cells and primary chondrocytes. Moreover, a role for the NF-κB/mitogen-activated protein kinase (MAPK) signaling cascade in regulating FOXO3 and ferroptosis was demonstrated. In vivo experiments confirmed the rescue effect of intra-articular injection of a FOXO3-overexpressing lentivirus against erastin-aggravated OA. Conclusions The results of our study show that the activation of ferroptosis promotes chondrocyte death and disrupts the ECM both in vivo and in vitro. In addition, FOXO3 can reduce OA progression by inhibiting ferroptosis through the NF-κB/MAPK signaling pathway. The Translational potential of this article This study highlights the important role of chondrocyte ferroptosis regulated by FOXO3 through the NF-κB/MAPK signaling in the progression of OA. The inhibition of chondrocyte ferroptosis by activating FOXO3 is expected to be a new target for the treatment of OA.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Guantong Sun
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yaxin Li
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Keyu Kong
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiaodong Li
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Tianyou Kan
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Fei Yang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Corresponding author. 639 Zhizaoju Road, Shanghai, 200011, PR China.
| | - Lei Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Corresponding author. 639 Zhizaoju Road, Shanghai, 200011, PR China.
| | - Xiaoqing Wang
- Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Corresponding author. 639 Zhizaoju Road, Shanghai, 200011, PR China.
| |
Collapse
|
29
|
Yang F, Maimaitimin M, He Z, Zhang X, Huang H, Wang J. The Cartilage Protective Effect of Labrum Reconstruction Using Meniscus Allograft Compared with Labrum Resection in a Porcine Model. Cartilage 2023; 14:76-85. [PMID: 36484319 PMCID: PMC10076893 DOI: 10.1177/19476035221141419] [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] [Indexed: 12/13/2022] Open
Abstract
PURPOSE This study aimed to verify the femoral head cartilage protective effect of labral reconstruction in a porcine model. METHODS Twelve pigs (24 hips) were divided into 3 groups: labrum defect group, lateral meniscus (LM) allograft group, and LM allograft wrapped with acellular peritoneum matrix (LM-APM) group before undergoing bilateral hip surgery. The pigs were sacrificed at 12 and 24 weeks postoperatively, while the femoral head cartilage was retrieved and then subjected to imaging measurement, macroscopic observations, and biomechanical and histological assessment. RESULTS Imaging measurement and macroscopic observations revealed that the defect area of the labrum was filled in LM and LM-APM allograft groups after 24 weeks, whereas the labrum defect remained at 24 weeks in the control group. The femoral head cartilage corresponding to the area of labral resection in the labral defect group had worse macroscopic Osteoarthritis Research Society International (OARSI) scores, uneven and discontinuous cartilage on hematoxylin and eosin (H&E) staining and Safranin O staining, decreased histopathology OARSI Osteoarthritis Cartilage Histopathology Assessment System (OOCHAS) scores, and decreased elastic modulus and hardness at 12 and 24 weeks after surgery compared with the meniscus allograft groups. CONCLUSION This study demonstrated that the LM allograft with or without APM for labral reconstruction had a chondroprotective effect on the femoral head in a porcine model.
Collapse
Affiliation(s)
- Fan Yang
- Beijing Key Laboratory of Sports Injuries, Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing, China
| | - Maihemuti Maimaitimin
- Beijing Key Laboratory of Sports Injuries, Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing, China
| | - Ziyi He
- Beijing Key Laboratory of Sports Injuries, Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing, China
| | - Xin Zhang
- Beijing Key Laboratory of Sports Injuries, Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing, China
| | - Hongjie Huang
- Beijing Key Laboratory of Sports Injuries, Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing, China
| | - Jianquan Wang
- Beijing Key Laboratory of Sports Injuries, Department of Sports Medicine, Institute of Sports Medicine of Peking University, Peking University Third Hospital, Beijing, China
| |
Collapse
|
30
|
Functional Loss of Terminal Complement Complex Protects Rabbits from Injury-Induced Osteoarthritis on Structural and Cellular Level. Biomolecules 2023; 13:biom13020216. [PMID: 36830586 PMCID: PMC9953363 DOI: 10.3390/biom13020216] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/25/2023] Open
Abstract
The terminal complement complex (TCC) has been described as a potential driver in the pathogenesis of posttraumatic osteoarthritis (PTOA). However, sublytic TCC deposition might also play a crucial role in bone development and regeneration. Therefore, we elucidated the effects of TCC on joint-related tissues using a rabbit PTOA model. In brief, a C6-deficient rabbit breed was characterized on genetic, protein, and functional levels. Anterior cruciate ligament transection (ACLT) was performed in C6-deficient (C6-/-) and C6-sufficient (C6+/-) rabbits. After eight weeks, the progression of PTOA was determined histologically. Moreover, the structure of the subchondral bone was evaluated by µCT analysis. C6 deficiency could be attributed to a homozygous 3.6 kb deletion within the C6 gene and subsequent loss of the C5b binding site. Serum from C6-/- animals revealed no hemolytic activity. After ACLT surgery, joints of C6-/- rabbits exhibited significantly lower OA scores, including reduced cartilage damage, hypocellularity, cluster formation, and osteophyte number, as well as lower chondrocyte apoptosis rates and synovial prostaglandin E2 levels. Moreover, ACLT surgery significantly decreased the trabecular number in the subchondral bone of C6-/- rabbits. Overall, the absence of TCC protected from injury-induced OA progression but had minor effects on the micro-structure of the subchondral bone.
Collapse
|
31
|
Ro DH, Jang MJ, Koh J, Choi WS, Kim HC, Han HS, Choi JW. Mechanism of action of genicular artery embolization in a rabbit model of knee osteoarthritis. Eur Radiol 2023; 33:125-134. [PMID: 35932304 DOI: 10.1007/s00330-022-09006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/02/2022] [Accepted: 06/27/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES To establish a rabbit osteoarthritis model for genicular artery embolization (GAE) experiments and to investigate the cellular mechanism of action of this novel procedure for interventional radiologists. METHODS Rabbit knee osteoarthritis was surgically modeled by anterior cruciate ligament transection and medial partial meniscectomy of the bilateral hindlimbs followed by 10 weeks of incubation. Rabbits exhibiting synovitis on magnetic resonance imaging were randomly divided into two groups: the bilateral GAE group and the control (sham procedure) group. Four weeks later, the rabbits' mobility (moving time, sec/10 min) and the histopathological features of each knee were assessed, and inter-group differences were evaluated using Student's t-test and ordinal/linear logistic models with generalized estimating equations. RESULTS Osteoarthritis modeling and endovascular procedures were successful in 15 of 20 rabbits (8 and 7 in the GAE and control groups, respectively). There was no significant difference in moving times between the two groups (p = .958). The degree of structural cartilage damage was similar in both groups (p = .780). However, the synovial proliferation (p = .016), synovial hypertrophy (p < .001), and villous hyperplasia of the synovial stroma (p = .002) scores were significantly lower in the GAE group than in the control group. The CD3+ cell density (p = .018) and CD3 + cell-infiltrated area (p = .019) were also significantly lower in the GAE group than in the control group. CONCLUSION GAE can limit inflammatory processes in the synovium of osteoarthritis-affected knees. KEY POINTS Surgical transection of the anterior cruciate ligament and medial partial meniscectomy of rabbit knees provides a useful animal model for research of genicular artery embolization. Osteoarthritic knees treated by genicular artery embolization showed milder synovial proliferation (p = .016), synovial hypertrophy (p < .001), and villous hyperplasia of the synovial stroma (p = .002) than the untreated knees. Osteoarthritic knees treated by genicular artery embolization presented lower CD3+ cell density (p = .018) and CD3+ cell-infiltrated area (p = .019) in the synovium than the untreated knees.
Collapse
Affiliation(s)
- Du Hyun Ro
- Department of Orthopedic Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Myoung-Jin Jang
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, Korea
| | - Jaemoon Koh
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Won Seok Choi
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyo-Cheol Kim
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, #101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Hyuk-Soo Han
- Department of Orthopedic Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Woo Choi
- Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, #101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
32
|
Dou H, Wang S, Hu J, Song J, Zhang C, Wang J, Xiao L. Osteoarthritis models: From animals to tissue engineering. J Tissue Eng 2023; 14:20417314231172584. [PMID: 37223125 PMCID: PMC10201005 DOI: 10.1177/20417314231172584] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/13/2023] [Indexed: 05/25/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative osteoarthropathy. Although it has been revealed that a variety of factors can cause or aggravate the symptoms of OA, the pathogenic mechanisms of OA remain unknown. Reliable OA models that accurately reflect human OA disease are crucial for studies on the pathogenic mechanism of OA and therapeutic drug evaluation. This review first demonstrated the importance of OA models by briefly introducing the OA pathological features and the current limitations in the pathogenesis and treatment of OA. Then, it mainly discusses the development of different OA models, including animal and engineered models, highlighting their advantages and disadvantages from the perspective of pathogenesis and pathology analysis. In particular, the state-of-the-art engineered models and their potential were emphasized, as they may represent the future direction in the development of OA models. Finally, the challenges in obtaining reliable OA models are also discussed, and possible future directions are outlined to shed some light on this area.
Collapse
Affiliation(s)
- Hongyuan Dou
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Shuhan Wang
- Shenzhen Institute for Drug Control, Shenzhen Testing Center of Medical Devices, Shenzhen, China
| | - Jiawei Hu
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Jian Song
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Jiali Wang
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
33
|
Panizzi L, Vignes M, Dittmer K, Waterland M, Rogers C, Sano H, McIlwraith C, Pemberton S, Owen M, Riley C. Infrared spectroscopy of serum fails to identify early biomarker changes in an equine model of traumatic osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100297. [DOI: 10.1016/j.ocarto.2022.100297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022] Open
|
34
|
Zhao C, Li X, Sun G, Liu P, Kong K, Chen X, Yang F, Wang X. CircFOXO3 protects against osteoarthritis by targeting its parental gene FOXO3 and activating PI3K/AKT-mediated autophagy. Cell Death Dis 2022; 13:932. [PMID: 36344492 PMCID: PMC9640610 DOI: 10.1038/s41419-022-05390-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/09/2022]
Abstract
Osteoarthritis (OA) is a degenerative joint disorder causing pain and functional disability. Emerging evidence reveals that circular RNAs (circRNAs) play essential roles in OA progression and development. This study aimed to investigate the role of a novel circRNA factor, circFOXO3, in the progression of OA and elucidate its underlying molecular mechanism. The function of circFOXO3 in OA and interaction between circFOXO3 and its downstream mRNA target, forkhead box O3 (FOXO3), were evaluated by western blot (WB), immunofluorescence (IF), RNA immunoprecipitation, reverse transcription-quantitative PCR (RT-qPCR), and fluorescence in situ hybridization (FISH). Upregulation of circFOXO3 and autophagic flux were detected both in vivo and in vitro by WB, transmission electron microscopy (TEM), IF, and immunohistochemistry (IHC). A mouse model of OA was also used to confirm the role of circFOXO3 in OA pathogenesis in vivo. Decreased expression of circFOXO3 in OA cartilage tissues was directly associated with excessive apoptosis and imbalance between anabolic and catabolic factors of the extracellular matrix (ECM). Mechanistically, circFOXO3 functioned in cartilage by targeting its parental gene FOXO3 and activating autophagy. Intra-articular injection of lentivirus-circFOXO3 alleviated OA in the mouse model. In conclusion, our results reveal the key role played by circFOXO3 in OA progression; circFOXO3 overexpression may alleviate apoptosis of chondrocytes and promote anabolism of the ECM via activation of FOXO3 and autophagy, providing a potentially effective novel therapeutic strategy for OA.
Collapse
Affiliation(s)
- Chen Zhao
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| | - Xiaodong Li
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| | - Guantong Sun
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| | - Pengcheng Liu
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| | - Keyu Kong
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| | - Xuzhuo Chen
- grid.16821.3c0000 0004 0368 8293Department of Oral Surgery, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| | - Fei Yang
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| | - Xiaoqing Wang
- grid.16821.3c0000 0004 0368 8293Department of Orthopedics, Shanghai Key Laboratory of Orthopedic Implant, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011 China
| |
Collapse
|
35
|
Hosokawa Y, Onodera T, Homan K, Yamaguchi J, Kudo K, Kameda H, Sugimori H, Iwasaki N. Establishment of a New Qualitative Evaluation Method for Articular Cartilage by Dynamic T2w MRI Using a Novel Contrast Medium as a Water Tracer. Cartilage 2022; 13:19476035221111503. [PMID: 36072990 PMCID: PMC9459471 DOI: 10.1177/19476035221111503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE In the early stages of cartilage damage, diagnostic methods focusing on the mechanism of maintaining the hydrostatic pressure of cartilage are thought to be useful. 17O-labeled water, which is a stable isotope of oxygen, has the advantage of no radiation exposure or allergic reactions and can be detected by magnetic resonance imaging (MRI). This study aimed to evaluate MRI images using 17O-labeled water in a rabbit model. DESIGN Contrast MRI with 17O-labeled water and macroscopic and histological evaluations were performed 4 and 8 weeks after anterior cruciate ligament transection surgery in rabbits. A total of 18 T2-weighted images were acquired, and 17O-labeled water was manually administered on the third scan. The 17O concentration in each phase was calculated from the signal intensity at the articular cartilage. Macroscopic and histological grades were evaluated and compared with the 17O concentration. RESULTS An increase in 17O concentration in the macroscopic and histologically injured areas was observed by MRI. Macroscopic evaluation showed that the 17O concentration significantly increased in the damaged site group. Histological evaluations also showed that 17O concentrations significantly increased at 36 minutes 30 seconds after initiating MRI scanning in the Osteoarthritis Research Society International (OARSI) grade 3 (0.493 in grade 0, 0.659 in grade 1, 0.4651 in grade 2, and 0.9964 in grade 3, P < 0.05). CONCLUSION 17O-labeled water could visualize earlier articular cartilage damage, which is difficult to detect by conventional methods.
Collapse
Affiliation(s)
- Yoshiaki Hosokawa
- Department of Orthopaedic Surgery,
Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo,
Japan
| | - Tomohiro Onodera
- Department of Orthopaedic Surgery,
Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo,
Japan,Tomohiro Onodera, Department of Orthopaedic
Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido
University, Sapporo 060-8648, Japan.
| | - Kentaro Homan
- Department of Orthopaedic Surgery,
Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo,
Japan
| | - Jun Yamaguchi
- Department of Orthopaedic Surgery,
Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo,
Japan
| | - Kohsuke Kudo
- Department of Diagnostic Imaging,
Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroyuki Kameda
- Department of Diagnostic Imaging,
Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | | | - Norimasa Iwasaki
- Department of Orthopaedic Surgery,
Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo,
Japan
| |
Collapse
|
36
|
Huang K, Wu B, Hou Z, Ahmad A, Ahmed M, Khan AA, Tian F, Cheng F, Chu W, Deng K. Psoralen downregulates osteoarthritis chondrocyte inflammation via an estrogen-like effect and attenuates osteoarthritis. Aging (Albany NY) 2022; 14:6716-6726. [PMID: 36036756 PMCID: PMC9467404 DOI: 10.18632/aging.204245] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022]
Abstract
Estrogen and its receptor play a positive role in the development of osteoarthritis (OA). Psoralen is a plant-derived estrogen analog. This study aimed to verify whether psoralen inhibits OA through an estrogen-like effect. First, human primary chondrocytes in the late stage of OA were extracted to complete collagen type II immunofluorescence staining and cell proliferation experiments. Subsequently, estrogen, psoralen and estrogen receptor antagonists were co-cultured with OA chondrocytes, and RT-PCR was performed to detect the gene expression. A rabbit OA model was subsequently made by anterior cruciate ligament transection (ACLT). They were set as Sham group, OA group and Psoralen group, respectively. The articular cartilage samples were taken after 5 weeks of treatment, and the effect was observed by gross observation, histological staining, micro-CT scanning of subchondral bone. The results of cellular experiments displayed that the cultured cells were positive for collagen II fluorescence staining and 12 μg/mL psoralen was selected as the optimal concentration. In addition, psoralen had effects similar to estrogen, promoting the expression of estrogen tar-get genes CTSD, PGR and TFF1 and decreasing the expression of the inflammation-related gene TNF- α, IL-1β and IL-6. The effect of psoralen was blocked after the use of an estrogen receptor antagonist. Further animal experiments indicated that the psoralen group showed less destruction of cartilage tissue and decreased OASRI scores compared with the OA group. A subchondral bone CT scan demonstrated that psoralen significantly increased subchondral bone mineral density (BMD), trabecular thickness and trabecular number and decreased trabecular separation. In summary, psoralen inhibits the inflammatory production of chondrocytes, which is related to estrogen-like effect, and can be used to attenuate the progression of OA.
Collapse
Affiliation(s)
- Kui Huang
- Departments of Orthopedics, The First Hospital of Yangtze University, Jingzhou, China
| | - Bo Wu
- Departments of Orthopedics, The First Hospital of Yangtze University, Jingzhou, China
| | - Zhuhu Hou
- Departments of Orthopedics, The Jiangling County People’s Hospital, Jingzhou, China
| | - Akhlaq Ahmad
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Mushtaq Ahmed
- Department of Biotechnology, University of Science and Technology, Bannu, Pakistan
| | - Ayesha Ali Khan
- Department of Biochemistry and Molecular Biology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Feng Tian
- Departments of Orthopedics, The First Hospital of Yangtze University, Jingzhou, China
| | - Fan Cheng
- Departments of Orthopedics, The First Hospital of Yangtze University, Jingzhou, China
| | - Wei Chu
- Departments of Orthopedics, The First Hospital of Yangtze University, Jingzhou, China
| | - Ke Deng
- Departments of Orthopedics, The First Hospital of Yangtze University, Jingzhou, China
| |
Collapse
|
37
|
Sifre V, Ten-Esteve A, Serra CI, Soler C, Alberich-Bayarri Á, Segarra S, Martí-Bonmatí L. Knee Cartilage and Subchondral Bone Evaluations by Magnetic Resonance Imaging Correlate with Histological Biomarkers in an Osteoarthritis Rabbit Model. Cartilage 2022; 13:19476035221118166. [PMID: 36004407 PMCID: PMC9421031 DOI: 10.1177/19476035221118166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE To evaluate pathological changes in cartilage and subchondral bone MRI biomarkers in a rabbit model of osteoarthritis (OA) and correlate these with histological variations. DESIGN Transection of the anterior cruciate ligament was performed on the right knee of eighteen 12-week-old New Zealand white rabbits to induce OA. 3-Tesla MR images were obtained from 18 healthy control knees (left) and 18 knees with OA (right). Imaging biomarkers included volume, thickness, T1 and T2* cartilage parametric maps, and several subchondral bone features: bone volume to total volume ratio, trabecular thickness, trabecular spacing, trabecular number (TbN), 2D and 3D fractal dimensions, and quality of trabecular score (QTS). Microscopic analysis of the lateral femoral condyles was set as the ground truth. RESULTS When healthy and osteoarthritic knees were compared, significant differences were seen in the T1 and T2* values of the femur and tibia cartilage and in the subchondral bone volume to total volume, TbN, and QTS of both the lateral and medial aspects of the femur and tibia. Histological findings revealed significant osteoarthritic changes between healthy and osteoarthritic knees in stain, structure, chondrocyte density, total score, and subchondral bone biomarker levels. A positive correlation was found between histological staining, structure, chondrocyte density, and total score variables in T1 and T2* cartilage biomarkers. A negative correlation was observed between histological subchondral bone variables and magnetic resonance D2D and QTS biomarkers. CONCLUSION Quantification of several cartilage and subchondral bone imaging biomarkers in a rabbit model of OA allows the detection of significant changes, which are correlated with histological findings.
Collapse
Affiliation(s)
- Vicente Sifre
- Programa de Doctorado en Ciencias de la Vida y del Medio Natural, Escuela de Doctorado, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain,Hospital Veterinario UCV, Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain,Vicente Sifre, Programa de Doctorado en Ciencias de la vida y del medio natural, Escuela de Doctorado, Universidad Católica de Valencia San Vicente Mártir, Avenida Pérez Galdós 51, Valencia 46018, Spain.
| | - Amadeo Ten-Esteve
- Biomedical Imaging Research Group (GIBI230-PREBI), La Fe Health Research Institute and Imaging La Fe node at Distributed Network for Biomedical Imaging, Unique Scientific and Technical Infrastructures, Valencia, Spain
| | - C. Iván Serra
- Hospital Veterinario UCV, Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Carme Soler
- Hospital Veterinario UCV, Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria y Ciencias Experimentales, Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - Ángel Alberich-Bayarri
- Biomedical Imaging Research Group (GIBI230-PREBI), La Fe Health Research Institute and Imaging La Fe node at Distributed Network for Biomedical Imaging, Unique Scientific and Technical Infrastructures, Valencia, Spain,Quantitative Imaging Biomarkers in Medicine, QUIBIM SL, Valencia, Spain
| | | | - Luis Martí-Bonmatí
- Biomedical Imaging Research Group (GIBI230-PREBI), La Fe Health Research Institute and Imaging La Fe node at Distributed Network for Biomedical Imaging, Unique Scientific and Technical Infrastructures, Valencia, Spain
| |
Collapse
|
38
|
Wu G, Ma F, Liu Z, Liu J, Xue Y, Zhang M, Wen C, Tang B, Lin L. Hybrid composites with magnesium-containing glycosaminoglycans as a chondroconducive matrix for osteoarthritic cartilage repair. Int J Biol Macromol 2022; 220:1104-1113. [PMID: 35981680 DOI: 10.1016/j.ijbiomac.2022.08.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/29/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022]
Abstract
The alteration of the extracellular matrix (ECM) homeostasis plays an important role in the development of osteoarthritis (OA). The pathological changes of OA are mainly manifested in the large reduction of components in ECM, like type II collagen and aggrecan, especially hyaluronic acid and chondroitin sulfate and often accompanied by inflammation. Rebuilding ECM and inhibiting inflammation may reverse OA progression. In this work, we developed new magnesium-containing glycosaminoglycans (Mg-GAGs), to create a positive ECM condition for promoting cartilage regeneration and alleviating OA. In vitro results suggested that the introduction of Mg-GAGs contributed to promoting chondrocyte proliferation and facilitated upregulating chondrogenic genes and suppressed inflammation-related factors. Moreover, Mg-GAGs exhibited positive effects on suppressing synovial inflammation, reducing chondrocyte apoptosis and preserving the subchondral bone in the ACLT-induced OA rabbit model. This study provides new insight into ECM-based therapeutic strategy and opens a new avenue for the development of novel OA treatment.
Collapse
Affiliation(s)
- Guofeng Wu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Orthopedics, Southern University of Science and Technology Hospital, Shenzhen, Guangdong, PR China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Fenbo Ma
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Zhengwei Liu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Jiayi Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Yizhebang Xue
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Mengdi Zhang
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China
| | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China; Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, PR China.
| | - Lijun Lin
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
39
|
Delbaldo C, Tschon M, Martini L, Fini M, Codispoti G. Benefits of Applying Nanotechnologies to Hydrogels in Efficacy Tests in Osteoarthritis Models-A Systematic Review of Preclinical Studies. Int J Mol Sci 2022; 23:ijms23158236. [PMID: 35897805 PMCID: PMC9368605 DOI: 10.3390/ijms23158236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 12/09/2022] Open
Abstract
Osteoarthritis (OA) is a severe musculoskeletal disease with an increasing incidence in the worldwide population. Recent research has focused on the development of innovative strategies to prevent articular cartilage damage and slow down OA progression, and nanotechnologies applied to hydrogels have gained particular interest. The aim of this systematic review is to investigate the state of the art on preclinical in vitro and in vivo efficacy studies applying nanotechnologies to hydrogels in OA models to elucidate the benefits of their applications. Three databases were consulted for eligible papers. The inclusion criteria were in vitro and in vivo preclinical studies, using OA cells or OA animal models, and testing hydrogels and nanoparticles (NPs) over the last ten years. Data extraction and quality assessment were performed. Eleven papers were included. In vitro studies evidenced that NP-gels do not impact on cell viability and do not cause inflammation in OA cell phenotypes. In vivo research on rodents showed that these treatments could increase drug retention in joints, reducing inflammation and preventing articular cartilage damage. Nanotechnologies in preclinical efficacy tests are still new and require extensive studies and technical hits to determine the efficacy, safety, fate, and localization of NPs for translation into an effective therapy for OA patients.
Collapse
|
40
|
Wang L, Zhao L, Shen L, Fang Q, Yang Z, Wang R, Wu Q, Xie Y. Comparison of the effects of autologous and allogeneic purified platelet-rich plasma on cartilage damage in a rabbit model of knee osteoarthritis. Front Surg 2022; 9:911468. [PMID: 35910465 PMCID: PMC9334772 DOI: 10.3389/fsurg.2022.911468] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Background Purified platelet-rich plasma (P-PRP) is gradually being used in the treatment of osteoarthritis (OA), and its sources are mainly divided into autologous and allogeneic blood. However, it is unclear whether autologous PRP is more effective or allogeneic PRP is superior. Objective In this study, autologous and allogeneic P-PRP was injected at early stage of KOA in rabbits, and then the differences in the efficacy of the two P-PRPs against KOA were compared from several perspectives, including pathological histology and immunohistochemistry. Method Experimental rabbits were divided into normal group (n = 8), model group (n = 8), autologous P-PRP group (n = 8), and allogeneic P-PRP group (n = 8) using a random number table method. The normal and model groups did not receive any treatment, and the autologous P-PRP and allogeneic P-PRP groups received intra-articular injections of autologous and allogeneic P-PRP, respectively, to observe the changes in the gross specimens of the knee joints of the experimental rabbits in each group. The histopathological changes of chondrocytes were also observed by HE-stained sections of articular cartilage, and the expression of chondrocytes Bone morphogenetic protein-2 (BMP-2) and Sox9 were detected by immunohistochemistry. Results Compared with the allogeneic P-PRP group, the differences were statistically significant (P < 0.05) in the gross specimens and pathological histological findings in the autologous PRP group. Immunohistochemical results showed that the expression of BMP-2 and Sox9 was elevated in both the autologous P-PRP group and the allogeneic P-PRP group compared with the model group, and the expression of BMP-2 was higher in the autologous P-PRP group than in the allogeneic P-PRP group, with a statistically significant difference (P < 0.05), while there was no difference in the expression of Sox9 between the two groups (P > 0.05). Conclusion Intra-articular injection of autologous P-PRP activated the expression of BMP-2 and Sox9 in chondrocytes and effectively improved KOA cartilage repair and reduced bone redundancy and joint fluid formation, and its efficacy was superior to that of intra-articular injection of allogeneic P-PRP.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Luting Zhao
- Department of Rehabilitation Medicine, The First People’s Hospital of Ziyang, Ziyang, China
| | - Lianwei Shen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
- Department of Rehabilitation Medicine, Shandong University Cheeloo College of Medicine, Jinan, China; Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qilin Fang
- Department of Rehabilitation Medicine, The First People’s Hospital of Ziyang, Ziyang, China
| | - Zhenglei Yang
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Rongrong Wang
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qing Wu
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Correspondence: Qing Wu Yulei Xie
| | - Yulei Xie
- Department of Rehabilitation Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Faculty of Rehabilitation Medicine, Capital Medical University, Beijing, China
- Correspondence: Qing Wu Yulei Xie
| |
Collapse
|
41
|
Liu NQ, Chen S, Geng D, Lei J, Zhang J, Li L, Lin Y, Ouyang Y, Shkhyan R, Van Handel B, Bian F, Mkaratigwa T, Chai Y, Evseenko D. Local Drug-Induced Modulation of gp130 Receptor Signaling Delays Disease Progression in a Pig Model of Temporo-Mandibular Joint Osteoarthritis. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.937819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Temporomandibular joint disorders (TMJs) are a multifaceted group of chronic disorders characterized by stiffness in the jaw, limited jaw mobility and pain when opening or closing the mouth. TMJs are relatively common, with incidence rates in the range of 5–12%, with nearly twice as many women as men being affected. One of the primary causes of TMJs is a degenerative disease of joints, such as osteoarthritis (OA), characterized by progressive loss of cartilage which causes stiffness, swelling, and pain. Currently, there are no disease-modifying agents on the market for OA. We have recently discovered a small molecule, R805 acting as a modulator of glycoprotein 130 (gp130) receptor for IL-6 family of cytokines. R805 enables regenerative outputs of endogenous joint stem and progenitor cells through immunomodulation in the joint microenvironment by reducing the levels of destructive cytokines and supporting chondrocyte survival and anabolism. Extensive testing has shown R805 to be safe at doses far above the therapeutic level. Here, we have conducted a pivotal efficacy study in our newly-established pig model of TMJ post-traumatic OA. IA injection of R805 has shown a highly significant reduction of articular cartilage degeneration, reduced synovitis and degenerative changes in subchondral bone in the mandibular condyle compared to the vehicle-treated group. These data will support additional pre-clinical development of R805 as a first-in-class injectable therapeutic for TMJ osteoarthritis.
Collapse
|
42
|
Mukherjee P, Roy S, Ghosh D, Nandi SK. Role of animal models in biomedical research: a review. Lab Anim Res 2022; 38:18. [PMID: 35778730 PMCID: PMC9247923 DOI: 10.1186/s42826-022-00128-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
The animal model deals with the species other than the human, as it can imitate the disease progression, its’ diagnosis as well as a treatment similar to human. Discovery of a drug and/or component, equipment, their toxicological studies, dose, side effects are in vivo studied for future use in humans considering its’ ethical issues. Here lies the importance of the animal model for its enormous use in biomedical research. Animal models have many facets that mimic various disease conditions in humans like systemic autoimmune diseases, rheumatoid arthritis, epilepsy, Alzheimer’s disease, cardiovascular diseases, Atherosclerosis, diabetes, etc., and many more. Besides, the model has tremendous importance in drug development, development of medical devices, tissue engineering, wound healing, and bone and cartilage regeneration studies, as a model in vascular surgeries as well as the model for vertebral disc regeneration surgery. Though, all the models have some advantages as well as challenges, but, present review has emphasized the importance of various small and large animal models in pharmaceutical drug development, transgenic animal models, models for medical device developments, studies for various human diseases, bone and cartilage regeneration model, diabetic and burn wound model as well as surgical models like vascular surgeries and surgeries for intervertebral disc degeneration considering all the ethical issues of that specific animal model. Despite, the process of using the animal model has facilitated researchers to carry out the researches that would have been impossible to accomplish in human considering the ethical prohibitions.
Collapse
Affiliation(s)
- P Mukherjee
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Mohanpur, Nadia, India
| | - S Roy
- Department of Veterinary Clinical Complex, West Bengal University of Animal and Fishery Sciences, Mohanpur, Nadia, India
| | - D Ghosh
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, India
| | - S K Nandi
- Department of Veterinary Surgery and Radiology, West Bengal University of Animal and Fishery Sciences, Kolkata, India.
| |
Collapse
|
43
|
Keita-Alassane S, Otis C, Bouet E, Guillot M, Frezier M, Delsart A, Moreau M, Bédard A, Gaumond I, Pelletier JP, Martel-Pelletier J, Beaudry F, Lussier B, Lecomte R, Marchand S, Troncy E. Estrogenic impregnation alters pain expression: analysis through functional neuropeptidomics in a surgical rat model of osteoarthritis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:703-715. [PMID: 35318491 DOI: 10.1007/s00210-022-02231-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/11/2022] [Indexed: 12/20/2022]
Abstract
PURPOSE Several observational studies suggest that estrogens could bias pain perception. To evaluate the influence of estrogenic impregnation on pain expression, a prospective, randomized, controlled, blinded study was conducted in a Sprague-Dawley rat model of surgically induced osteoarthritis (OA). METHODS Female rats were ovariectomized and pre-emptive 17β-estradiol (0.025 mg, 90-day release time) or placebo pellets were installed subcutaneously during the OVX procedures. Thirty-five days after, OA was surgically induced on both 17β-estradiol (OA-E) and placebo (OA-P) groups. Mechanical hypersensitivity was assessed by static weight-bearing (SWB) and paw withdrawal threshold (PWT) tests. Mass spectrometry coupled with high-performance liquid chromatography (HPLC-MS) was performed to quantify the spinal pronociceptive neuropeptides substance P (SP), calcitonin gene-related peptide (CGRP), bradykinin (BK), somatostatin (SST), and dynorphin-A (Dyn-A). RESULTS Compared to control, ovariectomized rats presented higher SP (P = 0.009) and CGRP (P = 0.017) concentrations. OA induction increased the spinal level of SP (+ 33%, P < 0.020) and decreased the release of BK (- 20%, (P < 0.037)). The OA-E rats at functional assessment put more % body weight on the affected hind limb than OA-P rats at D7 (P = 0.027) and D56 (P = 0.033), and showed higher PWT at D56 (P = 0.009), suggesting an analgesic and anti-allodynic effect of 17β-estradiol. Interestingly, the 17β-estradiol treatment counteracted the increase of spinal concentration of Dyn-A (P < 0.016) and CGRP (P < 0.018). CONCLUSION These results clearly indicate that 17β-estradiol interfers with the development of central sensitization and confirm that gender dimorphism should be considered when looking at pain evaluation.
Collapse
Affiliation(s)
- Sokhna Keita-Alassane
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
| | - Colombe Otis
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Emilie Bouet
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
| | - Martin Guillot
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Charles River Laboratories Montreal ULC, Senneville, QC, Canada
| | - Marilyn Frezier
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
| | - Aliénor Delsart
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
| | - Maxim Moreau
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Agathe Bédard
- Charles River Laboratories Montreal ULC, Senneville, QC, Canada
| | - Isabelle Gaumond
- Département de Chirurgie, Département d'anesthésie, Faculté de Médecine Et Des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Francis Beaudry
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Bertrand Lussier
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada
| | - Roger Lecomte
- Sherbrooke Molecular Imaging Center, Department of Nuclear Medicine and Radiobiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Serge Marchand
- Département de Chirurgie, Département d'anesthésie, Faculté de Médecine Et Des Sciences de La Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, QC, Canada
| | - Eric Troncy
- Research Group in Animal Pharmacology of Quebec (GREPAQ), Department of Veterinary Biomedicine, Faculty of Veterinary Medicine, Université de Montréal, Saint Hyacinthe, QC, Canada.
- Osteoarthritis Research Unit, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC, Canada.
| |
Collapse
|
44
|
Deng Z, Gao X, Utsunomiya H, Arner JW, Ruzbarsky JJ, Huard M, Ravuri S, Philippon MJ, Huard J. Effects of oral losartan administration on homeostasis of articular cartilage and bone in a rabbit model. Bone Rep 2022; 16:101526. [PMID: 35372645 PMCID: PMC8971351 DOI: 10.1016/j.bonr.2022.101526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 11/26/2022] Open
Abstract
Background and aims Previous work has shown that oral losartan can enhance microfracture-mediated cartilage repair in a rabbit osteochondral defect injury model. In this study, we aimed to determine whether oral losartan would have a detrimental effect on articular cartilage and bone homeostasis in the uninjured sides. Methods New Zealand rabbits were divided into 4 groups including normal uninjured (Normal), contralateral uninjured side of osteochondral defect (Defect), osteochondral defect plus microfracture (Microfracture) and osteochondral defect plus microfracture and losartan oral administration (10 mg/kg/day) (Losartan). Rabbits underwent different surgeries and treatment and were sacrificed at 12 weeks. Both side of the normal group and uninjured side of treatment groups tibias were harvested for Micro-CT and histological analysis for cartilage and bone including H&E staining, Herovici's staining (bone and cartilage) Alcian blue and Safranin O staining (cartilage) as well as immunohistochemistry of losartan related signaling pathways molecules for both cartilage and bone. Results Our results showed losartan oral treatment at 10 mg/kg/day slightly increase Alcian blue positive matrix as well as decrease collagen type 3 in articular cartilage while having no significant effect on articular cartilage structure, cellularity, and other matrix. Losartan treatment also did not affect angiotensin receptor type 1 (AGTR1), angiotensin receptor type 2 (AGTR2) and phosphorylated transforming factor β1 activated kinase 1 (pTAK1) expression level and pattern in the articular cartilage. Furthermore, losartan treatment did not affect microarchitecture of normal cancellous bone and cortical bone of tibias compared to normal and other groups. Losartan treatment slightly increased osteocalcin positive osteoblasts on the surface of cancellous bone and did not affect bone matrix collagen type 1 content and did not change AGTR1, AGTR2 and pTAK1 signal molecule expression. Conclusion Oral losartan used as a microfracture augmentation therapeutic does not have significant effect on uninjured articular cartilage and bone based on our preclinical rabbit model. These results provided further evidence that the current regimen of using losartan as a microfracture augmentation therapeutic is safe with respect to bone and cartilage homeostasis and support clinical trials for its application in human cartilage repair.
Collapse
|
45
|
Improved Joint Health Following Oral Administration of Glycosaminoglycans with Native Type II Collagen in a Rabbit Model of Osteoarthritis. Animals (Basel) 2022; 12:ani12111401. [PMID: 35681865 PMCID: PMC9179918 DOI: 10.3390/ani12111401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/26/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Osteoarthritis is an incurable chronic disease. For this reason, new therapies are constantly emerging to improve clinical signs and the quality of life of our pets. Chondroitin sulfate, glucosamine and hyaluronic acid have been proven effective and are the most widely used in many formulations. In the present study, adding native type II collagen to the combination of chondroitin sulfate, glucosamine and hyaluronic acid showed improvements on osteoarthritis progression in an experimental model of osteoarthritis induced by transection of the cranial cruciate ligament of the knee in New Zealand white rabbits. Disease progression was monitored at different time points using magnetic resonance imaging biomarkers, measurement of hyaluronic acid in synovial fluid, and macroscopic and microscopic evaluations of cartilage, synovial membrane and subchondral bone. Overall, our results showed that adding native type II collagen to a combination of glycosaminoglycans allows a significantly slower osteoarthritis progression, compared to glycosaminoglycans alone. Abstract A prospective, experimental, randomized, double blinded study was designed to evaluate the effects of glycosaminoglycans, with or without native type II collagen (NC), in an osteoarthritis model induced by cranial cruciate ligament transection. The following compounds were tested: chondroitin sulfate (CS), glucosamine hydrochloride (GlHCl), hyaluronic acid (HA) and NC. Fifty-four female 12-week-old New Zealand rabbits were classified into three groups: CTR (control–no treatment), CGH (CS + GlHCl + HA) and CGH-NC (CS + GlHCl + HA + NC). Each group was subdivided into three subgroups according to survival times of 24, 56 and 84 days. Over time, all rabbits developed degenerative changes associated with osteoarthritis. CGH-NC showed significantly improved values on macroscopic evaluation, compared to CTR and CGH. Microscopically, significantly better results were seen with CGH and CGH-NC, compared to CTR, and synovial membrane values were significantly better with CGH-NC compared to CGH. A significant improvement in magnetic resonance imaging biomarkers was also observed with CGH-NC in cartilage transversal relaxation time (T2) and subchondral bone D2D fractal dimension in the lateral condyle. In conclusion, our results show beneficial effects on joint health of CGH and CGH-NC and also supports that adding NC to CGH results in even greater efficacy.
Collapse
|
46
|
Senter R, Boyce R, Repic M, Martin EW, Chabicovsky M, Langevin-Carpentier G, Bédard A, Bodick N. Efficacy and Safety of FX201, a Novel Intra-Articular IL-1Ra Gene Therapy for Osteoarthritis Treatment, in a Rat Model. Hum Gene Ther 2022; 33:541-549. [PMID: 34963343 PMCID: PMC9142767 DOI: 10.1089/hum.2021.131] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/16/2021] [Indexed: 11/22/2022] Open
Abstract
Osteoarthritis (OA) is a disabling, degenerative disease characterized by progressive cartilage and bone damage. There remains a need for local therapies that, following a single injection, can provide long-term pain relief and functional improvement and potentially delay disease progression. FX201 is a novel, intra-articular (IA), interleukin-1 receptor antagonist (IL-1Ra) gene therapy in development for the treatment of OA. In this study, we assessed the efficacy, biodistribution, and safety of helper-dependent adenovirus (HDAd)-ratIL-1Ra, the rat surrogate of FX201, and the biodistribution of FX201, in the anterior cruciate ligament transection (ACLT) rat OA model. A single IA injection of HDAd-ratIL-1Ra administered 7 days post-ACLT mitigated OA-related changes to cartilage, bone, and the synovial membrane at week 12 following surgery. Furthermore, FX201 and HDAd-ratIL-1Ra persisted for at least 92 days in the injected joint and proximal tissues with minimal evidence of vector spreading peripherally. Finally, HDAd-ratIL-1Ra showed a favorable safety profile without any local or systemic adverse effects. In conclusion, HDAd-ratIL-1Ra demonstrated local therapeutic and disease-modifying effects and was well tolerated, supporting further clinical development of FX201.
Collapse
Affiliation(s)
- Rebecca Senter
- Flexion Therapeutics, Inc., Burlington, Massachusetts, USA
| | - Rogely Boyce
- Beechy Ridge ToxPath, LLC, Clay, West Virginia, USA
| | | | | | | | | | | | - Neil Bodick
- Gate Science, Inc., Moultonborough, New Hampshire, USA
| |
Collapse
|
47
|
Bosser C, Auregan JC, Bensidhoum M, Hoc T, Bégué T, Vialle R, Nouguier-Lehon C, Bachy M. Early impairment of cartilage poroelastic properties in an animal model of ACL tear. Orthop Traumatol Surg Res 2022; 108:103116. [PMID: 34666200 DOI: 10.1016/j.otsr.2021.103116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/16/2021] [Accepted: 04/23/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND In more than 50% of cases, anterior cruciate ligament (ACL) lesions lead to post-traumatic osteoarthritis. Ligament reconstruction stabilizes the joint, but the tear seems to impair the poroelasticity of the cartilage: synovial membrane fluid inflammation is observed 3 weeks after tearing. There have been some descriptions of visible cartilage changes, but poroelasticity has never been analyzed at this early stage. The present animal study aimed to determine (1) whether cartilage showed early poroelastic deterioration after ACL tear; (2) whether an impairment correlated with macroscopic changes; and (3) whether cartilage poroelasticity deteriorated over time. HYPOTHESIS In the days following trauma, cartilage poroelasticity is greatly impaired, without macroscopically visible change. MATERIAL AND METHODS ACL tear was surgically induced in 18 New-Zealand rabbits. Cartilage poroelasticity was assessed on indentation-relaxation test in 3 groups: "early", at 2 weeks postoperatively (n=6), "mid-early" at 6 weeks (n=6) and in a non-operated control group ("non-op"). Macroscopic changes were scored in the same groups. RESULTS Poroelastic impairment was greatest at the early time-point (2 weeks). Permeability ranged from a mean 0.08±0.05×10-15 m4/Ns (range, 0.028-0.17) in the "non-op" group to 1.03±0.60×10-15 m4/Ns (range, 0.24-2.15) in the "early" group (p=0.007). Shear modulus ranged from 0.53±0.11MPa (range, 0.36-0.66) to 0.23±0.10MPa (range, 0.12-0.43), respectively (p=0.013). Macroscopic deterioration, on the other hand, differed significantly only between the "mid-early" and the "non-op" groups: p=0.011 for cartilage deterioration and p=0.008 for osteophyte formation. At the "mid-early" time point, poroelastic deterioration was less marked, with 0.33±0.33×10-15 m4/Ns permeability (range, 0.06-1.06) and shear modulus 0.30±0.10MPa (range, 0.13-0.41: respectively p=0.039 and p=0.023 compared to the "non-op" group. DISCUSSION The severe rapid deterioration in poroelasticity following ACL tear in an animal model, as notably seen in increased permeability, corresponds to changes in cartilage microstructure, with easier outflows of interstitial fluid. This mechanical degradation may underlie onset of microcracks within the cartilage, leading to physiological loading that the cartilage by its nature is unable to repair. Further investigations are needed to correlate these experimental data with clinical findings. LEVEL OF EVIDENCE III; comparative study with control group.
Collapse
Affiliation(s)
- Cartherine Bosser
- HealthDataSciences, 45, chemin du Barthélémy, 69260 Charbonnières-les-Bains, France
| | - Jean Charles Auregan
- Université de Paris, B3OA, UMR CNRS 7052, Inserm U1271, 10, avenue de Verdun, 75010 Paris, France; Service d'orthopédie, université Paris-Saclay, AP-HP, Antoine Béclère, 157, rue de la Porte-de-Trivaux, 92140 Clamart, France
| | - Morad Bensidhoum
- Université de Paris, B3OA, UMR CNRS 7052, Inserm U1271, 10, avenue de Verdun, 75010 Paris, France
| | - Thierry Hoc
- Université de Paris, B3OA, UMR CNRS 7052, Inserm U1271, 10, avenue de Verdun, 75010 Paris, France; École centrale de Lyon, département de mécanique, MSGMGC, 36, avenue Guy-de-Collongue, 69134 Ecully cedex, France
| | - Thierry Bégué
- Service d'orthopédie, université Paris-Saclay, AP-HP, Antoine Béclère, 157, rue de la Porte-de-Trivaux, 92140 Clamart, France
| | - Raphaël Vialle
- Service de chirurgie orthopédique et réparatrice de l'enfant, AP-HP-Sorbonne université, hôpital Trousseau, 26, avenue du Docteur-Arnold-Netter, 75012 Paris, France
| | - Cécile Nouguier-Lehon
- Université de Lyon, LTDS UMR CNRS 5513, école centrale de Lyon, 36, avenue Guy-de-Collongue, 69134 Ecully cedex, France
| | - Manon Bachy
- Université de Paris, B3OA, UMR CNRS 7052, Inserm U1271, 10, avenue de Verdun, 75010 Paris, France; Service de chirurgie orthopédique et réparatrice de l'enfant, AP-HP-Sorbonne université, hôpital Trousseau, 26, avenue du Docteur-Arnold-Netter, 75012 Paris, France.
| |
Collapse
|
48
|
Hua B, Qiu J, Ye X, Liu X. Intra-articular injection of a novel Wnt pathway inhibitor, SM04690, upregulates Wnt16 expression and reduces disease progression in temporomandibular joint osteoarthritis. Bone 2022; 158:116372. [PMID: 35218985 DOI: 10.1016/j.bone.2022.116372] [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: 12/28/2021] [Revised: 02/14/2022] [Accepted: 02/19/2022] [Indexed: 11/20/2022]
Abstract
Abnormal Wnt signaling has been shown to be involved in the pathogenesis of temporomandibular joint osteoarthritis (TMJOA). Recent studies demonstrates that SM04690, a small-molecule inhibitor of the Wnt signaling pathway, is able to promote cartilage regeneration in a rat model of knee joint osteoarthritis. However, whether SM04690 has any effect on TMJOA is unknown. Here we first performed partial TMJ discectomy to induce TMJOA in rabbit and rat. Histology, TRAP staining, immunohistochemistry and μCT analysis showed intra-articular injection of SM04690 protected condylar cartilage from degeneration and attenuated abnormal subchondral bone remodeling of TMJ condylar in both rabbit and rat model TMJOA. We isolated and cultured primary condylar chondrocytes for in vitro studies to investigate molecular mechanisms and downstream effects of SM04690. We found that SM04690 inhibited the canonical Wnt pathway, upregulated the expression of Wnt16 and cartilage anabolic factors including COL2A1, SOX9 and aggrecan, suppressed the expression of cartilage catabolic factor MMP13 and protected chondrocytes from TNF-α-induced inflammatory response. Previous studies have identified fibrocartilage stem cells (FCSCs) localized within the TMJ condyle superficial zone niche that regenerate cartilage and repair joint injury. Here we showed that intra-articular injection of SM04690 increased the number of the TMJ condyle superficial zone (SZ) cells in vivo. Further in vitro studies revealed that SM04690 enhanced FCSCs chondrogenesis and formation of cartilaginous-like tissue in pellet cultures. Taken together, our work demonstrates that SM04690 treatment might be able to promote FCSCs chondrogenesis and repair TMJ cartilage, highlighting the therapeutic potential of intra-articular injection of SM04690 in TMJOA.
Collapse
Affiliation(s)
- Bingqiang Hua
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Jin Qiu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoping Ye
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xianwen Liu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
49
|
Terracciano R, Carcamo-Bahena Y, Royal ALR, Demarchi D, Labis JS, Harris JD, Weiner BK, Gupta N, Filgueira CS. Quantitative high-resolution 7T MRI to assess longitudinal changes in articular cartilage after anterior cruciate ligament injury in a rabbit model of post-traumatic osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100259. [DOI: 10.1016/j.ocarto.2022.100259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/28/2022] [Accepted: 03/30/2022] [Indexed: 11/25/2022] Open
|
50
|
Zaki S, Blaker CL, Little CB. OA foundations - experimental models of osteoarthritis. Osteoarthritis Cartilage 2022; 30:357-380. [PMID: 34536528 DOI: 10.1016/j.joca.2021.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is increasingly recognised as a disease of diverse phenotypes with variable clinical presentation, progression, and response to therapeutic intervention. This same diversity is readily apparent in the many animal models of OA. However, model selection, study design, and interpretation of resultant findings, are not routinely done in the context of the target human (or veterinary) patient OA sub-population or phenotype. This review discusses the selection and use of animal models of OA in discovery and therapeutic-development research. Beyond evaluation of the different animal models on offer, this review suggests focussing the approach to OA-animal model selection on study objective(s), alignment of available models with OA-patient sub-types, and the resources available to achieve valid and translatable results. How this approach impacts model selection is discussed and an experimental design checklist for selecting the optimal model(s) is proposed. This approach should act as a guide to new researchers and a reminder to those already in the field, as to issues that need to be considered before embarking on in vivo pre-clinical research. The ultimate purpose of using an OA animal model is to provide the best possible evidence if, how, when and where a molecule, pathway, cell or process is important in clinical disease. By definition this requires both model and study outcomes to align with and be predictive of outcomes in patients. Keeping this at the forefront of research using pre-clinical OA models, will go a long way to improving the quality of evidence and its translational value.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Australia.
| | - C L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Australia; Murray Maxwell Biomechanics Laboratory, The Kolling Institute, University of Sydney Faculty of Medicine and Health, At Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Australia.
| |
Collapse
|