1
|
Yamashiro K, Ikegaya Y, Matsumoto N. Automatic detection of foot-strike onsets in a rhythmic forelimb movement. Neurosci Res 2024; 206:41-50. [PMID: 38642677 DOI: 10.1016/j.neures.2024.04.002] [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: 02/01/2024] [Revised: 04/03/2024] [Accepted: 04/09/2024] [Indexed: 04/22/2024]
Abstract
Rhythmic movement is the fundamental motion dynamics characterized by repetitive patterns. Precisely defining onsets in rhythmic movement is essential for a comprehensive analysis of motor functions. Our study introduces an automated method for detecting rat's forelimb foot-strike onsets using deep learning tools. This method demonstrates high accuracy of onset detection by combining two techniques using joint coordinates and behavioral confidence scale. The analysis extends to neural oscillatory responses in the rat's somatosensory cortex, validating the effectiveness of our combined approach. Our technique streamlines experimentation, demanding only a camera and GPU-accelerated computer. This approach is applicable across various contexts and promotes our understanding of brain functions during rhythmic movements.
Collapse
Affiliation(s)
- Kotaro Yamashiro
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuji Ikegaya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan; Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka 565-0871, Japan
| | - Nobuyoshi Matsumoto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
2
|
Liu Y, Nie M, Li X, Wang H, Ren S, Zou D, Liu J, Li R. Garlic-derived Exosomes Alleviate Osteoarthritis Through Inhibiting the MAPK Signaling Pathway. Appl Biochem Biotechnol 2024:10.1007/s12010-024-05047-6. [PMID: 39190086 DOI: 10.1007/s12010-024-05047-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease affecting millions of people worldwide. Garlic-derived exosomes (GDEs) are nanoparticles extracted from garlic that exhibit anti-inflammatory effects on other diseases, but the effect of GDEs on OA has not been elucidated. In this study, GDEs were extracted and characterized. Chondrocytes were treated with IL-1β and incubated with GDEs in vitro, and the expression of cartilage matrix components (collagen II and aggrecan) and matrix degrading enzymes (MMP3 and MMP9) was evaluated via Western blotting. Changes in the MAPK pathway was also examined using Western blotting. The transcriptomic changes associated with GDE intervention were evaluated using high-throughput RNA-seq method. In vivo, we used anterior cruciate ligament transection (ACLT) combined with destabilization of the medial meniscus (DMM) surgery to establish a mouse OA model, and GDEs was intraarticularly injected into the joint cavity. The therapeutic effect of GDE was evaluated by behavioral and histopathological analysis. The results showed that IL-1β treatment inhibited the expression of collagen II and aggrecan, and upregulated the expression of MMP3 and MMP9, while GDE intervention alleviated these effects. GDEs also inhibited the phosphorylation of ERK, JNK, and P38. In vivo, GDE alleviated the sensitivity to heat stimulation and altered walking gait in a mouse OA model. Histopathological analysis indicated that GDE intervention ameliorated joint destruction in the knee joint without obvious toxicity. The results proved that GDEs alleviated the progression of OA in vitro and in vivo, and may be a potential disease-modifying drug for OA.
Collapse
Affiliation(s)
- Yuqin Liu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Ming Nie
- Center for Joint Surgery, Department of Orthopedic Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xueyi Li
- Center for Joint Surgery, Department of Orthopedic Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Hao Wang
- Center for Joint Surgery, Department of Orthopedic Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Shaoju Ren
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Dezheng Zou
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jianhui Liu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
| | - Ruidong Li
- Center for Joint Surgery, Department of Orthopedic Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
3
|
Okazaki Y, Nakagawa Y, Deng XH, Zhang X, Wada S, Album Z, Ying L, Rodeo SA. Establishment of a Posttraumatic Osteoarthritis Model in Mice Induced by Noninvasive Anterior Cruciate Ligament Tear. Am J Sports Med 2024; 52:2008-2020. [PMID: 38828660 DOI: 10.1177/03635465241253225] [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: 06/05/2024]
Abstract
BACKGROUND Animal models that use open surgical transection of the anterior cruciate ligament (ACL) do not accurately simulate the clinical condition regarding the pivot-shift mechanism and the associated inflammatory response that occurs before reconstruction. PURPOSE/HYPOTHESIS The purpose was to characterize a reproducible manual, nonsurgical method to mimic an isolated ACL tear in a clinically relevant model and to evaluate the development of progressive posttraumatic osteoarthritis due to ACL injury. It was hypothesized that the ACL could be reproducibly torn with minimal damage to other ligaments and that there would be progressive development of degenerative joint disease after ACL injury. STUDY DESIGN Controlled laboratory study. METHODS A total of 37 mice (strain C57BL/6) were used to compare the manual procedure with sham surgery (sham group; n = 10) and with the established surgical ACL transection (ACLT) procedure (surgical group; n = 27). In the sham group, a closed manual procedure was performed on the right knee and sham surgery on the left knee. In the surgical group, the closed manual procedure was performed on the right knee and surgical ACLT on the left knee. Dissection using India ink, histological assessment with safranin O and hematoxylin-eosin staining, radiological evaluation through radiographs and microfocus computed tomography scans, and gait analyses were performed to assess cartilage/ligament status. Osteoarthritis Research Society International (OARSI) and synovitis scores, anterior tibial translation, range of motion, bone microstructure, osteophyte volume, and pain were assessed at 2, 4, and 8 weeks postoperatively. RESULTS The manual procedure successfully resulted in an ACL rupture and associated meniscal injury. The posterior cruciate, lateral collateral, and medial collateral ligaments were intact in all dissected knees. Two weeks after ACL tear, the surgical group showed a significantly higher synovitis score, whereas 8 weeks after ACL tear, the manual group showed a significantly higher volume of osteophytes. No significant differences were found between the groups in terms of OARSI score, anterior tibial translation, range of motion, bone microstructure computed tomography values, and stride distance/irregularity. CONCLUSION This procedure can be used to create an ACL tear model without causing grossly evident injuries to other ligaments and avoiding the risk of cartilage damage from surgical instruments. CLINICAL RELEVANCE This procedure offers a more clinically relevant ACL tear model and facilitates simple, inexpensive, and reproducible development of posttraumatic osteoarthritis.
Collapse
Affiliation(s)
- Yuki Okazaki
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
| | - Yusuke Nakagawa
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
- Cartilage Regeneration, Tokyo Medical and Dental University, Tokyo, Japan
| | - Xiang-Hua Deng
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
| | - Xueying Zhang
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
| | - Susumu Wada
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
| | - Zoe Album
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
| | - Liang Ying
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
| | - Scott A Rodeo
- Orthopaedic Soft Tissue Research Program, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
4
|
Kaiser JM, Bernard FC, Pucha K, Raval SK, Eng T, Fulton T, Anderson SE, Allen KD, Dixon JB, Willett NJ. Mild exercise expedites joint clearance and slows joint degradation in a joint instability model of osteoarthritis in male rats. Osteoarthritis Cartilage 2024; 32:912-921. [PMID: 38642879 DOI: 10.1016/j.joca.2024.03.120] [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: 06/28/2023] [Revised: 03/10/2024] [Accepted: 03/12/2024] [Indexed: 04/22/2024]
Abstract
OBJECTIVE Exercise remains a hallmark treatment for post-traumatic osteoarthritis (PTOA) and may maintain joint homeostasis in part by clearing inflammatory cytokines, cells, and particles. It remains largely unknown whether exercise-induced joint clearance can provide therapeutic relief of PTOA. In this study, we hypothesized that exercise could slow the progression of preclinical PTOA in part by enhancing knee joint clearance. DESIGN Surgical medial meniscal transection was used to induce PTOA in 3-month-old male Lewis rats. A sham surgery was used as a control. Mild treadmill walking was introduced 3 weeks post-surgery and maintained to 6 weeks post-surgery. Gait and isometric muscle torque were measured at the study endpoint. Near-infrared imaging tracked how exercise altered lymphatic and venous knee joint clearance during discrete time points of PTOA progression. RESULTS Exercise mitigated joint degradation associated with PTOA by preserving glycosaminoglycan content and reducing osteophyte volume (effect size (95% Confidence Interval (CI)); 1.74 (0.71-2.26)). PTOA increased hind step widths (0.57 (0.18-0.95) cm), but exercise corrected this gait dysfunction (0.54 (0.16-0.93) cm), potentially indicating pain relief. Venous, but not lymphatic, clearance was quicker 1-, 3-, and 6-weeks post-surgery compared to baseline. The mild treadmill walking protocol expedited lymphatic clearance rate in moderate PTOA (3.39 (0.20-6.59) hrs), suggesting exercise may play a critical role in restoring joint homeostasis. CONCLUSIONS We conclude that mild exercise has the potential to slow disease progression in part by expediting joint clearance in moderate PTOA.
Collapse
Affiliation(s)
- Jarred M Kaiser
- Atlanta Veterans Affairs Hospital, Decatur, GA, USA; Emory University School of Medicine, Decatur, GA, USA.
| | - Fabrice C Bernard
- Emory University School of Medicine, Decatur, GA, USA; Georgia Institute of Technology, Atlanta, GA, USA.
| | - Krishna Pucha
- Emory University School of Medicine, Decatur, GA, USA.
| | | | - Tracy Eng
- Atlanta Veterans Affairs Hospital, Decatur, GA, USA; Emory University School of Medicine, Decatur, GA, USA.
| | - Travis Fulton
- Atlanta Veterans Affairs Hospital, Decatur, GA, USA; Emory University School of Medicine, Decatur, GA, USA.
| | - Shannon E Anderson
- Emory University School of Medicine, Decatur, GA, USA; Georgia Institute of Technology, Atlanta, GA, USA.
| | | | - J Brandon Dixon
- Emory University School of Medicine, Decatur, GA, USA; Georgia Institute of Technology, Atlanta, GA, USA.
| | - Nick J Willett
- Atlanta Veterans Affairs Hospital, Decatur, GA, USA; Emory University School of Medicine, Decatur, GA, USA; Georgia Institute of Technology, Atlanta, GA, USA; Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA.
| |
Collapse
|
5
|
Liu H, Huang R, Zhuo Z, Zhang X, Wu L, Guo Z, Wen F, An L, Yuan H, Zhang Y, Xu Y. Activation of kappa opioid receptor suppresses post-traumatic osteoarthritis via sequestering STAT3 on the plasma membrane. Cell Commun Signal 2024; 22:335. [PMID: 38890746 PMCID: PMC11186255 DOI: 10.1186/s12964-024-01709-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/09/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE Kappa opioid receptor (KOR) signaling is involved in joint development and inflammation in Osteoarthritis (OA), while the biochemical mechanism remains unclarified. This study aims to investigate downstream molecular events of KOR activation, to provide novel perspectives in OA pathology. METHODS U50,488H, a selective KOR agonist, was intra-articularly injected in mice upon destabilization of the medial meniscus (DMM) as OA models, with PBS injection as control. The behavioral and histological evaluation was assessed by hot plate test and red solid green staining, respectively. Alterations in mRNA and protein expression were assessed by RNA-seq, RT-qPCR, immunohistochemistry and western blotting (WB) in chondrocytes treated with TNF-α or TNF-α + U50,488H. Proteins interacted with KOR were explored using proximity labeling followed by mass spectrometry and then testified by co-immunoprecipitation (Co-IP) assay and immunofluorescence (IF). RESULTS OA-induced pain was reduced and cartilage degeneration was alleviated upon KOR activation in DMM mice. In chondrocytes, activation of KOR reversed the upregulation of MMPs, IL-6, IL-1β and phosphorylated(p-) STAT3, stimulated by TNF-α, while the expression of NF-κB, MAPKs and AKT signaling weren't reversed. RNA-seq and IF results presented that KOR activation evidently reduced STAT3 nuclear translocation in chondrocytes upon TNF-α stimuli. The reduction may be resulted from the binding of KOR and STAT3 in the plasma membrane, revealed by proximity labeling and Co-IP results. CONCLUSIONS KOR activation protects cartilage from OA, and this protective effect is mainly exerted via sequestering STAT3 on the plasma membrane, resulting in inactivation of STAT3-dependent immune responses which otherwise contributes to OA.
Collapse
Affiliation(s)
- Haixia Liu
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Renhuan Huang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ziang Zhuo
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinru Zhang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ling Wu
- iView Therapeutics, Inc., Cranbury, NJ, USA
| | - Zhen Guo
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fuping Wen
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liwei An
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hang Yuan
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Yiming Zhang
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
6
|
Guan Z, Liu Y, Luo L, Jin X, Guan Z, Yang J, Liu S, Tao K, Pan J. Sympathetic innervation induces exosomal miR-125 transfer from osteoarthritic chondrocytes, disrupting subchondral bone homeostasis and aggravating cartilage damage in aging mice. J Adv Res 2024:S2090-1232(24)00122-X. [PMID: 38554999 DOI: 10.1016/j.jare.2024.03.022] [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: 10/12/2023] [Revised: 03/16/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is a progressive disease that poses a significant threat to human health, particularly in aging individuals: Although sympathetic activation has been implicated in bone metabolism, its role in the development of OA related to aging remains poorly understood. Therefore, this study aimed to investigate how sympathetic regulation impacts aging-related OA through experiments conducted both in vivo and in vitro. METHODS To analyze the effect of sympathetic regulation on aging-related OA, we conducted experiments using various mouse models. These models included a natural aging model, a medial meniscus instability model, and a load-induced model, which were used to examine the involvement of sympathetic nerves. In order to evaluate the expression levels of β1-adrenergic receptor (Adrβ1) and sirtuin-6 (Sirt6) in chondrocytes of naturally aging OA mouse models, we performed assessments. Additionally, we investigated the influence of β1-adrenergic receptor knockout or treatment with a β1-adrenergic receptor blocker on the progression of OA in aging mice and detected exosome release and detected downstream signaling expression by inhibiting exosome release. Furthermore, we explored the impact of sympathetic depletion through tyrosine hydroxylase (TH) on OA progression in aging mice. Moreover, we studied the effects of norepinephrine(NE)-induced activation of the β1-adrenergic receptor signaling pathway on the release of exosomes and miR-125 from chondrocytes, subsequently affecting osteoblast differentiation in subchondral bone. RESULTS Our findings demonstrated a significant increase in sympathetic activity, such as NE levels, in various mouse models of OA including natural aging, medial meniscus instability, and load-induced models. Notably, we observed alterations in the expression levels of β1-adrenergic receptor and Sirt6 in chondrocytes in OA mouse models associated with natural aging, leading to an improvement in the progression of OA. Critically, we found that the knockout of β1-adrenergic receptor or treatment with a β1-adrenergic receptor blocker attenuated OA progression in aging mice and the degraded cartilage explants produced more exosome than the nondegraded ones, Moreover, sympathetic depletion through TH was shown to ameliorate OA progression in aging mice. Additionally, we discovered that NE-induced activation of the β1-adrenergic receptor signaling pathway facilitated the release of exosomes and miR-125 from chondrocytes, promoting osteoblast differentiation in subchondral bone. CONCLUSION In conclusion, our study highlights the role of sympathetic innervation in facilitating the transfer of exosomal miR-125 from osteoarthritic chondrocytes, ultimately disrupting subchondral bone homeostasis and exacerbating cartilage damage in aging mice. These findings provide valuable insights into the potential contribution of sympathetic regulation to the pathogenesis of aging-related OA.
Collapse
Affiliation(s)
- Zhiyuan Guan
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China; Science and Technology Center, Fenyang College of Shanxi Medical University, Shanxi 032200, China
| | - Yanbin Liu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiaotong University, No. 100 Haining Road, Shanghai 200080, China
| | - Liying Luo
- Department of Ophthalmology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Jin
- Department of Rheumatology and Immunology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zhiqiang Guan
- Department of Dermatology, Xuzhou Municipal Hospital Affiliated with Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Jianjun Yang
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Shengfu Liu
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Kun Tao
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Jianfeng Pan
- Science and Technology Center, Fenyang College of Shanxi Medical University, Shanxi 032200, China.
| |
Collapse
|
7
|
Choi SH, Kim HC, Jang SG, Lee YJ, Heo JY, Kweon GR, Ryu MJ. Effects of a Combination of Polynucleotide and Hyaluronic Acid for Treating Osteoarthritis. Int J Mol Sci 2024; 25:1714. [PMID: 38338992 PMCID: PMC10855695 DOI: 10.3390/ijms25031714] [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: 12/28/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Knee osteoarthritis (OA), an age-related degenerative disease characterized by severe pain and disability, is treated using polynucleotides (PNs) and hyaluronic acid (HA). The intra-articular (IA) injection of HA has been studied extensively in both animal models and in humans; however, the efficacy and mechanisms of action remain unclear. In addition, there has been a paucity of research regarding the use of PN alone or in combination with HA in OA. To investigate the effect of the combined injection of PN and HA in vivo, pathological and behavioral changes were assessed in an OA model. Anterior cruciate ligament transection and medial meniscectomy were performed in Sprague-Dawley rats to create the OA animal model. The locomotor activity improved following PNHA injection, while the OARSI grade improved in the medial tibia and femur. In mild OA, TNFα levels decreased histologically in the PN, HA, and PNHA groups but only the PNHA group showed behavioral improvement in terms of distance. In conclusion, PNHA exhibited anti-inflammatory effects during OA progression and improved locomotor activity regardless of the OARSI grade.
Collapse
Affiliation(s)
- Seung Hee Choi
- Joonghun Pharmaceutical Co., Ltd., 15 Gukhoe-daero 62-gil, Yeongdeungpo-gu, Seoul 07236, Republic of Korea; (S.H.C.); (H.C.K.); (S.G.J.); (Y.J.L.)
| | - Hyun Chul Kim
- Joonghun Pharmaceutical Co., Ltd., 15 Gukhoe-daero 62-gil, Yeongdeungpo-gu, Seoul 07236, Republic of Korea; (S.H.C.); (H.C.K.); (S.G.J.); (Y.J.L.)
| | - Seul Gi Jang
- Joonghun Pharmaceutical Co., Ltd., 15 Gukhoe-daero 62-gil, Yeongdeungpo-gu, Seoul 07236, Republic of Korea; (S.H.C.); (H.C.K.); (S.G.J.); (Y.J.L.)
| | - Yeon Jae Lee
- Joonghun Pharmaceutical Co., Ltd., 15 Gukhoe-daero 62-gil, Yeongdeungpo-gu, Seoul 07236, Republic of Korea; (S.H.C.); (H.C.K.); (S.G.J.); (Y.J.L.)
| | - Jun Young Heo
- Department of Biochemistry, College of Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; (J.Y.H.); (G.R.K.)
| | - Gi Ryang Kweon
- Department of Biochemistry, College of Medicine, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea; (J.Y.H.); (G.R.K.)
| | - Min Jeong Ryu
- Joonghun Pharmaceutical Co., Ltd., 15 Gukhoe-daero 62-gil, Yeongdeungpo-gu, Seoul 07236, Republic of Korea; (S.H.C.); (H.C.K.); (S.G.J.); (Y.J.L.)
| |
Collapse
|
8
|
Doucet MR, Laevski AM, Doiron JA, Boudreau LH, Surette ME. Locomotor activity as an effective measure of the severity of inflammatory arthritis in a mouse model. PLoS One 2024; 19:e0291399. [PMID: 38232088 DOI: 10.1371/journal.pone.0291399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 08/28/2023] [Indexed: 01/19/2024] Open
Abstract
OBJECTIVE Mouse models are valuable in preclinical studies of inflammatory arthritis. However, current methods for measuring disease severity or responses to treatment are not optimal. In this study a smart cage system using multiple sensors to measure locomotor activity was evaluated in the K/BxN serum transfer model of inflammatory arthritis. METHODS Arthritis was induced in C57BL/6 mice with injections of K/BxN serum. Clinical index and ankle thickness were measured for 14 days. Locomotor activity was measured in smart cages for 23 h periods on Days 0, 7, and 13. The same measurements were taken in mice consuming diets supplemented or not with fish oil to evaluate a preventative treatment. RESULTS Initiation, peak and resolution phases of disease could be measured with the smart cages. Locomotor activity including speed, travel distance, number of active movements and rear movements were all significantly lower on Days 7-8 of illness (peak) compared to Days 0 and 13-14 (resolution) (one-way repeated measures analyses, p<0.05). The clinical index and ankle thickness measurements did not capture differences between dietary groups. Significantly increased activity was measured in most of the locomotor parameters in the fish oil group compared to the control mice at both Days 8 and 14 (2-way repeated measures ANOVA, p<0.05). CONCLUSION The measurement of locomotor activity provided a more detailed evaluation of the impact of inflammatory arthritis on animal well-being and mobility than that provided by measuring clinical index and ankle thickness, and could be a valuable tool in preclinical studies of inflammatory arthritis.
Collapse
Affiliation(s)
- Mélina R Doucet
- Department of Chemistry and Biochemistry, New Brunswick Centre for Precision Medicine, Université de Moncton, Moncton, New Brunswick, Canada
| | - Angela M Laevski
- Department of Chemistry and Biochemistry, New Brunswick Centre for Precision Medicine, Université de Moncton, Moncton, New Brunswick, Canada
| | - Jérémie A Doiron
- Department of Chemistry and Biochemistry, New Brunswick Centre for Precision Medicine, Université de Moncton, Moncton, New Brunswick, Canada
| | - Luc H Boudreau
- Department of Chemistry and Biochemistry, New Brunswick Centre for Precision Medicine, Université de Moncton, Moncton, New Brunswick, Canada
| | - Marc E Surette
- Department of Chemistry and Biochemistry, New Brunswick Centre for Precision Medicine, Université de Moncton, Moncton, New Brunswick, Canada
| |
Collapse
|
9
|
Nooreen Z, Wal P, Summaiyya F. A Systemic Review on Nutraceutical Supplements used in the Management of Osteoarthritis. RECENT ADVANCES IN FOOD, NUTRITION & AGRICULTURE 2024; 15:33-45. [PMID: 38258782 DOI: 10.2174/012772574x270405231102054920] [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: 07/31/2023] [Revised: 09/19/2023] [Accepted: 09/28/2023] [Indexed: 01/24/2024]
Abstract
Osteoarthritis (OA) is a progressive degenerative joint disease. It basically impairs the structural integrity of articulate cartilage and imbalances the catabolic and anabolic signals in the joint. A degenerative disease is characterized by swelling, pain, and joint stiffness. The treatment and management of osteoarthritis are based on analgesic and anti-inflammatory agents, whereas the exact cause of OA is not known yet. The negative effects of synthetic medications have led to a daily rise in the usage of nutraceuticals and dietary supplements. Clinicians are aware of these treatments, and they also recommend nutraceuticals in addition to the currently preferred therapy. Many in-vitro and in-vivo experiments have been performed in past years to evaluate the function of these on osteoarthritis. The collection of articles was published on search engines like PubMed, Scopus, Google Scholar, ResearchGate, and ScienceDirect. The evaluation covers every potential nutraceutical utilized in osteoarthritis, together with its supporting data and mode of action. The present review discusses nutraceuticals, including devil's claw, vitamin D, boswellic acid, capsaicin, ginger, curcumin, krill oil, ginger, and avocado/soybean unsaponifiable.
Collapse
Affiliation(s)
- Zulfa Nooreen
- Department of Pharmacy, PSIT - Pranveer Singh Institute of Technology (Pharmacy) Bhauti, Kanpur Uttar Pradesh 209305, India
| | - Pranay Wal
- Department of Pharmacy, PSIT - Pranveer Singh Institute of Technology (Pharmacy) Bhauti, Kanpur Uttar Pradesh 209305, India
| | - Fariha Summaiyya
- Department of Biotechnology, Integral Informatic and Research Center-1 (IIRC-1) Intergral University Lucknow Uttar Pradesh-226026, India
| |
Collapse
|
10
|
Fu W, Vasylyev D, Bi Y, Zhang M, Sun G, Khleborodova A, Huang G, Zhao L, Zhou R, Li Y, Liu S, Cai X, He W, Cui M, Zhao X, Hettinghouse A, Good J, Kim E, Strauss E, Leucht P, Schwarzkopf R, Guo EX, Samuels J, Hu W, Attur M, Waxman SG, Liu CJ. Na v1.7 as a chondrocyte regulator and therapeutic target for osteoarthritis. Nature 2024; 625:557-565. [PMID: 38172636 PMCID: PMC10794151 DOI: 10.1038/s41586-023-06888-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Osteoarthritis (OA) is the most common joint disease. Currently there are no effective methods that simultaneously prevent joint degeneration and reduce pain1. Although limited evidence suggests the existence of voltage-gated sodium channels (VGSCs) in chondrocytes2, their expression and function in chondrocytes and in OA remain essentially unknown. Here we identify Nav1.7 as an OA-associated VGSC and demonstrate that human OA chondrocytes express functional Nav1.7 channels, with a density of 0.1 to 0.15 channels per µm2 and 350 to 525 channels per cell. Serial genetic ablation of Nav1.7 in multiple mouse models demonstrates that Nav1.7 expressed in dorsal root ganglia neurons is involved in pain, whereas Nav1.7 in chondrocytes regulates OA progression. Pharmacological blockade of Nav1.7 with selective or clinically used pan-Nav channel blockers significantly ameliorates the progression of structural joint damage, and reduces OA pain behaviour. Mechanistically, Nav1.7 blockers regulate intracellular Ca2+ signalling and the chondrocyte secretome, which in turn affects chondrocyte biology and OA progression. Identification of Nav1.7 as a novel chondrocyte-expressed, OA-associated channel uncovers a dual target for the development of disease-modifying and non-opioid pain relief treatment for OA.
Collapse
Affiliation(s)
- Wenyu Fu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Dmytro Vasylyev
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT, USA
| | - Yufei Bi
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Mingshuang Zhang
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Guodong Sun
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Asya Khleborodova
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Guiwu Huang
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Libo Zhao
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Renpeng Zhou
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Yonggang Li
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Shujun Liu
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT, USA
| | - Xianyi Cai
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Wenjun He
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Min Cui
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Xiangli Zhao
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Aubryanna Hettinghouse
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Julia Good
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Ellen Kim
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Eric Strauss
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Philipp Leucht
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Ran Schwarzkopf
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
| | - Edward X Guo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jonathan Samuels
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Wenhuo Hu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mukundan Attur
- Division of Rheumatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Center for Neuroscience and Regeneration Research, Veterans Affairs Connecticut Healthcare, West Haven, CT, USA.
| | - Chuan-Ju Liu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Karuppagounder V, Chung J, Abdeen A, Thompson A, Bouboukas A, Pinamont WJ, Yoshioka NK, Sepulveda DE, Raup-Konsavage WM, Graziane NM, Vrana KE, Elbarbary RA, Kamal F. Therapeutic Effects of Non-Euphorigenic Cannabis Extracts in Osteoarthritis. Cannabis Cannabinoid Res 2023; 8:1030-1044. [PMID: 35994012 PMCID: PMC10714119 DOI: 10.1089/can.2021.0244] [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] [Indexed: 11/13/2022] Open
Abstract
Introduction: Osteoarthritis (OA) is disabling and degenerative disease of the joints that is clinically characterized by pain and loss of function. With no disease-modifying treatment available, current therapies aim at pain management but are of limited efficacy. Cannabis products, specifically cannabinoids, are widely used to control pain and inflammation in many diseases with no scientific evidence demonstrating their efficacy in OA. Objective: We investigated the effects of non-euphorigenic cannabis extracts, CBD oil and cannabigerol oil (CBG oil), on pain and disease progression in OA mice. Methods and Results: Twelve-week-old male C57BL/6J mice received either sham or destabilization of the medial meniscus (DMM) surgery. DMM mice were treated with vehicle, CBD oil, or CBG oil. The gait of DMM mice was impaired as early as 2 weeks following surgery and continued deteriorating until week 8, which was restored by CBD oil and CBG oil treatments throughout the disease course. Mechanical allodynia developed in DMM mice, however, was not ameliorated by any of the treatments. On the other hand, both CBD oil and CBG oil ameliorated cold allodynia. In open field test, both oil treatments normalized changes in the locomotor activity of DMM mice. CBD oil and CBG oil treatments significantly reduced synovitis in DMM mice. Only CBG oil reduced cartilage degeneration, chondrocyte loss, and matrix metalloproteinase 13 expression, with a significant increase in the number of anabolic chondrocytes. Subchondral bone remodeling found in vehicle-treated DMM mice was not ameliorated by either CBD or CBG oil. Conclusions: Our results show evidence for the therapeutic efficacy of CBD oil and CBG oil, where both oils ameliorate pain and inflammation, and improve gait and locomotor activity in OA mice, representing clinical pain and function. Importantly, only CBG oil is chondroprotective, which may provide superior efficacy in future studies in OA patients.
Collapse
Affiliation(s)
- Vengadeshprabhu Karuppagounder
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Juliet Chung
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Ahmed Abdeen
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Amy Thompson
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Andreas Bouboukas
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - William J. Pinamont
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Natalie K. Yoshioka
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Diana E. Sepulveda
- Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Anesthesiology and Perioperative Medicine, and Penn State College of Medicine, Hershey, Pennsylvania, USA
| | | | - Nicholas M. Graziane
- Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Anesthesiology and Perioperative Medicine, and Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Kent E. Vrana
- Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Reyad A. Elbarbary
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
- Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Fadia Kamal
- Center for Orthopedic Research and Translational Science (CORTS), Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Orthopedics and Rehabilitation, Departments of Penn State College of Medicine, Hershey, Pennsylvania, USA
- Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
12
|
Pang Y, Ma Y, Zheng K, Zhu S, Sui H, Ren H, Liu K, Li W, Huang Y, Du D, Gao J, Zhang C. Costal Cartilage Graft Repair Osteochondral Defect in a Mouse Model. Cartilage 2023:19476035231209404. [PMID: 37881954 DOI: 10.1177/19476035231209404] [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] [Indexed: 10/27/2023] Open
Abstract
OBJECTIVE Osteochondral defects develop into osteoarthritis without intervention. Costal cartilage can be utilized as an alternative source for repairing osteochondral defect. Our previous clinical study has shown the successful osteochondral repair by costal cartilage graft with integration into host bone bed. In this study, we investigate how cartilaginous graft adapt to osteochondral environment and the mechanism of bone-cartilage interface formation. DESIGN Costal cartilage grafting was performed in C57BL/6J mice and full-thickness osteochondral defect was made as control. 3D optical profiles and micro-CT were applied to evaluate the reconstruction of articular cartilage surface and subchondral bone as well as gait analysis to evaluate articular function. Histological staining was performed at 2, 4, and 8 weeks after surgery. Moreover, costal cartilage from transgenic mice with fluorescent markers were transplanted into wild-type mice to observe the in vivo changes of costal chondrocytes. RESULTS At 8 weeks after surgery, 3D optical profiles and micro-CT showed that in the graft group, the articular surface and subchondral bone were well preserved. Gait analysis and International Cartilage Repair Society (ICRS) score evaluation showed a good recovery of joint function and histological repair in the graft group. Safranin O staining showed the gradual integration of graft and host tissue. Costal cartilage from transgenic mice with fluorescent markers showed that donor-derived costal chondrocytes turned into osteocytes in the subchondral area of host femur. CONCLUSION Costal cartilage grafting shows both functional and histological repair of osteochondral defect in mice. Graft-derived costal chondrocytes differentiate into osteocytes and contribute to endochondral ossification.
Collapse
Affiliation(s)
- Yidan Pang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiyang Ma
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaiwen Zheng
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyuan Zhu
- Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyu Sui
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hao Ren
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kang Liu
- Beixcell (Beijing) Biotechnology Ltd, Beijing, China
| | - Wei Li
- Beixcell (Beijing) Biotechnology Ltd, Beijing, China
| | - Yigang Huang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dajiang Du
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjie Gao
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Jinjiang Municipal Hospital (Shanghai Sixth People's Hospital Fujian), Jinjiang City, Quanzhou, China
| | - Changqing Zhang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Habib YH, Sheta E, Khattab M, Gowayed MA. Diminazene aceturate or losartan ameliorates the functional, radiological and histopathological alterations in knee osteoarthritis rodent model: repurposing of the ACE2/Ang1-7/MasR cascade. J Exp Orthop 2023; 10:107. [PMID: 37878123 PMCID: PMC10600085 DOI: 10.1186/s40634-023-00673-1] [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: 06/18/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023] Open
Abstract
PURPOSE Current therapies for osteoarthritis (OA) are limited to analgesics and anti-inflammatory drugs. Considering the importance of oxidative stress and inflammatory mediators in OA etiology, we tested the hypothesis that targeting the renin-angiotensin-aldosterone system (RAAS) can improve OA anomalies. Diminazene (DIZE), an activator of angiotensin-converting enzyme 2 and the angiotensin 2 type-1 receptor blocker losartan (LOS) were used for this purpose. METHODS OA was induced by a single intra-articular injection of monosodium iodoacetate. The effects of exposure to DIZE or LOS for 21 days on OA anomalies in rats' knees were investigated. Evaluation of motor function, nociception, and inflammatory response was done using rotarod, knee bend and knee swelling tests. Markers of knee joint inflammation, and cellular oxidation in addition to the RAAS biomarkers, were assessed in knee tissues, along with radiological and histopathological investigations. RESULTS Elevations in inflammatory and oxidative markers in knee tissues of OA rats were mostly improved by the two therapeutic drugs. Such effect was also reflected in the rotarod, knee bend and knee swelling tests. Treatment with DIZE has shown a more prominent effect than LOS in controlling OA-associated inflammation and cellular oxidation. Markers of RAAS have also shown better responsiveness to DIZE over LOS. CONCLUSIONS DIZE has shown a prominent increase in the angiotensin 1-7 amount, highlighting the involvement of the signaling pathway in the immunomodulatory effect. The radiological and histopathology examination came to confirm the outcome of biochemical markers, nominating diminazene aceturate as a possible therapeutic option for OA.
Collapse
Affiliation(s)
- Yasser H Habib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Mahmoud Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mennatallah A Gowayed
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Canal El- Mahmoudia Str., Smouha, Alexandria, Egypt.
| |
Collapse
|
14
|
Ni W, Zhang H, Mei Z, Hongyi Z, Wu Y, Xu W, Ma Y, Yang W, Liang Y, Gu T, Su Y, Fan S, Shen S, Hu Z. An inducible long noncoding RNA, LncZFHX2, facilitates DNA repair to mediate osteoarthritis pathology. Redox Biol 2023; 66:102858. [PMID: 37633048 PMCID: PMC10472307 DOI: 10.1016/j.redox.2023.102858] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/06/2023] [Accepted: 08/17/2023] [Indexed: 08/28/2023] Open
Abstract
Cartilage homeostasis is essential for chondrocytes to maintain proper phenotype and metabolism. Because adult articular cartilage is avascular, chondrocytes must survive in low oxygen conditions, and changing oxygen tension can significantly affect metabolism and proteoglycan synthesis in these cells. However, whether long noncoding RNA participate in cartilage homeostasis under hypoxia has not been reported yet. Here, we first identified LncZFHX2 as a lncRNA upregulated under physiological hypoxia in cartilage, specifically by HIF-1α. LncZFHX2 knockdown simultaneously accelerated cellular senescence, targeted multiple components of extracellular matrix metabolism, and increased DNA damage in chondrocytes. Through a series of in vitro and in vivo experiments, we identified that LncZFHX2 performed a novel function that regulated RIF1 expression through forming a transcription complex with KLF4 and promoting chondrocyte DNA repair. Moreover, chondrocyte-conditional knockout of LncZFHX2 accelerated injury-induced cartilage degeneration in vivo. In conclusion, we identified a hypoxia-activated DNA repair pathway that maintains matrix homeostasis in osteoarthritis cartilage.
Collapse
Affiliation(s)
- Weiyu Ni
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Haitao Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Zixuan Mei
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Zhou Hongyi
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Yizheng Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Wenbin Xu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Yan Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Wentao Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Yi Liang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Tianyuan Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Yingfeng Su
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China.
| | - Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China.
| | - Ziang Hu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang Province, China; Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China.
| |
Collapse
|
15
|
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: 1.0] [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
|
16
|
Lian WS, Wu RW, Ko JY, Chen YS, Wang SY, Jahr H, Wang FS. Inhibition of histone lysine demethylase 6A promotes chondrocytic activity and attenuates osteoarthritis development through repressing H3K27me3 enhancement of Wnt10a. Int J Biochem Cell Biol 2023; 158:106394. [PMID: 36871937 DOI: 10.1016/j.biocel.2023.106394] [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: 10/19/2022] [Revised: 02/08/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Histone hypermethylation represses gene transcription, which affects cartilage homeostasis or joint remodeling. Trimethylation of lysine 27 of histone 3 (H3K27me3) changes epigenome signatures, regulating tissue metabolism. This study aimed to investigate whether loss of H3K27me3 demethylase Kdm6a function affected osteoarthritis development. We revealed that chondrocyte-specific Kdm6a knockout mice developed relatively long femurs and tibiae as compared to wild-type mice. Kdm6a deletion mitigated osteoarthritis symptoms, including articular cartilage loss, osteophyte formation, subchondral trabecular bone loss, and irregular walking patterns of destabilized medial meniscus-injured knees. In vitro, loss of Kdm6a function compromised the loss in expression of key chondrocyte markers Sox9, collagen II, and aggrecan and improved glycosaminoglycan production in inflamed chondrocytes. RNA sequencing showed that Kdm6a loss changed transcriptomic profiles, which contributed to histone signaling, NADPH oxidase, Wnt signaling, extracellular matrix, and cartilage development in articular cartilage. Chromatin immunoprecipitation sequencing uncovered that Kdm6a knockout affected H3K27me3 binding epigenome, repressing Wnt10a and Fzd10 transcription. Wnt10a was, among others, functional molecules regulated by Kdm6a. Forced Wnt10a expression attenuated Kdm6a deletion-induced glycosaminoglycan overproduction. Intra-articular administration with Kdm6a inhibitor GSK-J4 attenuated articular cartilage erosion, synovitis, and osteophyte formation, improving gait profiles of injured joints. In conclusion, Kdm6a loss promoted transcriptomic landscapes contributing to extracellular matrix synthesis and compromised epigenetic H3K27me3-mediated promotion of Wnt10a signaling, preserving chondrocytic activity to attenuate osteoarthritic degeneration. We highlighted the chondroprotective effects of Kdm6a inhibitor for mitigating the development of osteoarthritic disorders.
Collapse
Affiliation(s)
- Wei-Shiung Lian
- Core Laboratory for Phenomics and Diagnostics, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Memorial Hospital, Kaohsiung, Taiwan; Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Re-Wen Wu
- Department of Orthopedic Surgery, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Jih-Yang Ko
- Department of Orthopedic Surgery, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yu-Shan Chen
- Core Laboratory for Phenomics and Diagnostics, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Memorial Hospital, Kaohsiung, Taiwan; Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Shao-Yu Wang
- Core Laboratory for Phenomics and Diagnostics, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Memorial Hospital, Kaohsiung, Taiwan; Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Holger Jahr
- Department of Anatomy and Cell Biology, University Hospital RWTH Aachen, Germany; Department of Orthopedic Surgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Feng-Sheng Wang
- Core Laboratory for Phenomics and Diagnostics, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan; Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Memorial Hospital, Kaohsiung, Taiwan; Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
17
|
Maestas DR, Chung L, Han J, Wang X, Sommerfeld SD, Kelly SH, Moore E, Nguyen HH, Mejías JC, Peña AN, Zhang H, Hooks JST, Chin AF, Andorko JI, Berlinicke CA, Krishnan K, Choi Y, Anderson AE, Mahatme R, Mejia C, Eric M, Woo J, Ganguly S, Zack DJ, Zhao L, Pearce EJ, Housseau F, Pardoll DM, Elisseeff JH. Helminth egg derivatives as proregenerative immunotherapies. Proc Natl Acad Sci U S A 2023; 120:e2211703120. [PMID: 36780522 PMCID: PMC9974432 DOI: 10.1073/pnas.2211703120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 01/11/2023] [Indexed: 02/15/2023] Open
Abstract
The immune system is increasingly recognized as an important regulator of tissue repair. We developed a regenerative immunotherapy from the helminth Schistosoma mansoni soluble egg antigen (SEA) to stimulate production of interleukin (IL)-4 and other type 2-associated cytokines without negative infection-related sequelae. The regenerative SEA (rSEA) applied to a murine muscle injury induced accumulation of IL-4-expressing T helper cells, eosinophils, and regulatory T cells and decreased expression of IL-17A in gamma delta (γδ) T cells, resulting in improved repair and decreased fibrosis. Encapsulation and controlled release of rSEA in a hydrogel further enhanced type 2 immunity and larger volumes of tissue repair. The broad regenerative capacity of rSEA was validated in articular joint and corneal injury models. These results introduce a regenerative immunotherapy approach using natural helminth derivatives.
Collapse
Affiliation(s)
- David R. Maestas
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Liam Chung
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, School of Medicine, Baltimore, MD21287
| | - Jin Han
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Xiaokun Wang
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Sven D. Sommerfeld
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Sean H. Kelly
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Erika Moore
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
- Materials Science and Engineering, University of Florida, Gainesville, FL32611
| | - Helen Hieu Nguyen
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Joscelyn C. Mejías
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Alexis N. Peña
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Hong Zhang
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Joshua S. T. Hooks
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Alexander F. Chin
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - James I. Andorko
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, School of Medicine, Baltimore, MD21287
| | - Cynthia A. Berlinicke
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Kavita Krishnan
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Younghwan Choi
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Amy E. Anderson
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Ronak Mahatme
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Christopher Mejia
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Marie Eric
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - JiWon Woo
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
| | - Sudipto Ganguly
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, School of Medicine, Baltimore, MD21287
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Liang Zhao
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, School of Medicine, Baltimore, MD21287
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Edward J. Pearce
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, School of Medicine, Baltimore, MD21287
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD21287
| | - Franck Housseau
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, School of Medicine, Baltimore, MD21287
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Drew M. Pardoll
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, School of Medicine, Baltimore, MD21287
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD21287
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, School of Medicine, Baltimore, MD21287
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD21287
| |
Collapse
|
18
|
Mima Z, Wang K, Liang M, Wang Y, Liu C, Wei X, Luo F, Nie P, Chen X, Xu Y, Ma Q. Blockade of JAK2 retards cartilage degeneration and IL-6-induced pain amplification in osteoarthritis. Int Immunopharmacol 2022; 113:109340. [DOI: 10.1016/j.intimp.2022.109340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/27/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
|
19
|
Zhao R, Wei X, Zhang C, Wu H, Xiang C, Li H, Duan W, Duan Z, Li C, Zhao Y, Huang L. α2-macroglobulin-rich serum as a master inhibitor of inflammatory factors attenuates cartilage degeneration in a mini pig model of osteoarthritis induced by “idealized” anterior cruciate ligament reconstruction. Front Pharmacol 2022; 13:849102. [PMID: 36133821 PMCID: PMC9483147 DOI: 10.3389/fphar.2022.849102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Post-traumatic osteoarthritis is a special type of osteoarthritis and a common disease, with few effective treatments available. α2-Macroglobulin (α2M) is important to chondral protection in post-traumatic osteoarthritis. However, its injection into xenogeneic joint cavities involves safety hazards, limiting clinical applications. Exploring serum α2M-enriching strategies and the therapeutic effect and mechanism of α2M-rich serum (α2MRS) autologous joint injection to treat post-traumatic osteoarthritis has significant value. In the present study, a unique filtration process was used to obtain α2MRS from human and mini pig serum. We evaluated the potential of α2MRS in protecting against post-surgery cartilage degeneration. We identify the potential of α2MRS in reducing the expression of inflammatory cytokines and factors that hasten cartilage degeneration in post-operative conditions leading to post-traumatic osteoarthritis. The potential of α2MRS was analyzed in interleukin-1β induced human chondrocytes and mini pig models. In the chondrocyte model, α2MRS significantly promoted human chondrocyte proliferation and reduced apoptosis and chondrocyte catabolic cytokine gene transcription and secretion. The anterior cruciate ligament autograft reconstruction model of mini pigs was randomized into groups, operated on, and injected with α2MRS or saline. The results showed that α2MRS injection significantly suppressed the levels of inflammatory factors, improved gait, and showed significantly lower cartilage degeneration than the groups that did not receive α2MRS injections. This study highlights the chondroprotective effects of α2MRS, elucidated its potential applications against cartilage degeneration, and could provide a basis for the clinical translation of α2MRS.
Collapse
Affiliation(s)
- Ruipeng Zhao
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University. Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Xiaochun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University. Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Chengming Zhang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University. Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Hongru Wu
- Shanxi Institute of Sports Science, Taiyuan, China
| | - Chuan Xiang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University. Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Haoqian Li
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University. Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Wangping Duan
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University. Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Zhiqing Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Chunjiang Li
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University. Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Yu Zhao
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University. Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
| | - Lingan Huang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University. Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Taiyuan, China
- Department of Pain Medicine, Sanya Central Hospital of Hainan Medical College, Sanya, China
- *Correspondence: Lingan Huang,
| |
Collapse
|
20
|
Jin Y, Liu Z, Li Z, Li H, Zhu C, Li R, Zhou T, Fang B. Histone demethylase JMJD3 downregulation protects against aberrant force-induced osteoarthritis through epigenetic control of NR4A1. Int J Oral Sci 2022; 14:34. [PMID: 35831280 PMCID: PMC9279410 DOI: 10.1038/s41368-022-00190-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/27/2022] [Accepted: 06/21/2022] [Indexed: 11/09/2022] Open
Abstract
Osteoarthritis (OA) is a prevalent joint disease with no effective treatment strategies. Aberrant mechanical stimuli was demonstrated to be an essential factor for OA pathogenesis. Although multiple studies have detected potential regulatory mechanisms underlying OA and have concentrated on developing novel treatment strategies, the epigenetic control of OA remains unclear. Histone demethylase JMJD3 has been reported to mediate multiple physiological and pathological processes, including cell differentiation, proliferation, autophagy, and apoptosis. However, the regulation of JMJD3 in aberrant force-related OA and its mediatory effect on disease progression are still unknown. In this work, we confirmed the upregulation of JMJD3 in aberrant force-induced cartilage injury in vitro and in vivo. Functionally, inhibition of JMJD3 by its inhibitor, GSK-J4, or downregulation of JMJD3 by adenovirus infection of sh-JMJD3 could alleviate the aberrant force-induced chondrocyte injury. Mechanistic investigation illustrated that aberrant force induces JMJD3 expression and then demethylates H3K27me3 at the NR4A1 promoter to promote its expression. Further experiments indicated that NR4A1 can regulate chondrocyte apoptosis, cartilage degeneration, extracellular matrix degradation, and inflammatory responses. In vivo, anterior cruciate ligament transection (ACLT) was performed to construct an OA model, and the therapeutic effect of GSK-J4 was validated. More importantly, we adopted a peptide-siRNA nanoplatform to deliver si-JMJD3 into articular cartilage, and the severity of joint degeneration was remarkably mitigated. Taken together, our findings demonstrated that JMJD3 is flow-responsive and epigenetically regulates OA progression. Our work provides evidences for JMJD3 inhibition as an innovative epigenetic therapy approach for joint diseases by utilizing p5RHH-siRNA nanocomplexes.
Collapse
Affiliation(s)
- Yu Jin
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhen Liu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhenxia Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hairui Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Cheng Zhu
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Ruomei Li
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Ting Zhou
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China.
| | - Bing Fang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China.
| |
Collapse
|
21
|
Batista TSC, Oliveira AFR, Santana LB, Nascimento V, Cândido EAF, Batista MVA. Gait analysis with muscular fibrosis and treatment with Alpinia zerumbet essential oil in immobilized rats. AN ACAD BRAS CIENC 2022; 94:e20211164. [PMID: 35703698 DOI: 10.1590/0001-3765202220211164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022] Open
Abstract
The analysis of gait in animals is important for understanding movement disorders in various human pathologies, especially those that develop muscle fibrosis. In the search for treatment alternatives for this problem, essential oils have been studied. Among them, research involving the essential oil of Alpinia zerumbet (EOAz) has been shown to promote relaxation and improve muscle function. Therefore, this study aimed to evaluate the effect of EOAz on gait with muscle fibrosis in immobilized rats. 30 rats (Wistar) were divided into five groups of six animals each: control group (without fibrosis and without treatment), immobilization group (with fibrosis and without treatment), and EOAz treatment groups (with fibrosis and with treatment). The animals were immobilized for 15 days with an ankle plantar flexion orthosis. After this period, they were treated with the oil cutaneously for 30 days. The analysis of behavioral tests before treatment indicated a significant increase in the means of the immobilized groups about to with concerning the control. We conclude that EOAz was effective in improving gait after inducing muscle fibrosis in immobilized rats. Studies are needed to assess the oil's effectiveness in the treatment of muscle fibrosis in human pathologies.
Collapse
Affiliation(s)
- Thaisa S C Batista
- Universidade Federal de Sergipe, Departamento de Fisioterapia, Av. Governador Marcelo Déda, s/n, 49400-000 Lagarto, SE, Brazil.,Universidade Tiradentes, Instituto de Pesquisa de Sergipe, Laboratório de Estudos Biológicos e Produtos Naturais, Av. Murilo Dantas, 300, 49010-390 Aracaju, SE, Brazil
| | - Amanda F R Oliveira
- Universidade Tiradentes, Instituto de Pesquisa de Sergipe, Laboratório de Estudos Biológicos e Produtos Naturais, Av. Murilo Dantas, 300, 49010-390 Aracaju, SE, Brazil
| | - Luana B Santana
- Universidade Tiradentes, Instituto de Pesquisa de Sergipe, Laboratório de Estudos Biológicos e Produtos Naturais, Av. Murilo Dantas, 300, 49010-390 Aracaju, SE, Brazil
| | - Vítor Nascimento
- Universidade Tiradentes, Instituto de Pesquisa de Sergipe, Laboratório de Estudos Biológicos e Produtos Naturais, Av. Murilo Dantas, 300, 49010-390 Aracaju, SE, Brazil
| | - Edna A F Cândido
- Universidade Tiradentes, Instituto de Pesquisa de Sergipe, Laboratório de Estudos Biológicos e Produtos Naturais, Av. Murilo Dantas, 300, 49010-390 Aracaju, SE, Brazil
| | - Marcus V A Batista
- Universidade Federal de Sergipe, Departamento de Biologia, Centro de Ciências Biológicas e da Saúde, Laboratório de Genética Molecular e Biotecnologia (GMBio), Av. Marechal Rondon, s/n, Jd. Rosa Elze, 49100-000 São Cristóvão, SE, Brazil
| |
Collapse
|
22
|
Fouasson-Chailloux A, Menu P, Dauty M. Lower-Limb Arthropathies and Walking: The Use of 3D Gait Analysis as a Relevant Tool in Clinical Practice. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19116785. [PMID: 35682370 PMCID: PMC9179954 DOI: 10.3390/ijerph19116785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/01/2023]
Affiliation(s)
- Alban Fouasson-Chailloux
- Service de Médecine Physique et Réadaptation Locomotrice et Respiratoire, CHU Nantes, Nantes Université, 44093 Nantes, France; (P.M.); (M.D.)
- Service de Médecine du Sport, CHU Nantes, Nantes Université, 44093 Nantes, France
- Institut Régional de Médecine du Sport (IRMS), 44093 Nantes, France
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, ONIRIS, Nantes Université, 44042 Nantes, France
- Correspondence:
| | - Pierre Menu
- Service de Médecine Physique et Réadaptation Locomotrice et Respiratoire, CHU Nantes, Nantes Université, 44093 Nantes, France; (P.M.); (M.D.)
- Service de Médecine du Sport, CHU Nantes, Nantes Université, 44093 Nantes, France
- Institut Régional de Médecine du Sport (IRMS), 44093 Nantes, France
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, ONIRIS, Nantes Université, 44042 Nantes, France
| | - Marc Dauty
- Service de Médecine Physique et Réadaptation Locomotrice et Respiratoire, CHU Nantes, Nantes Université, 44093 Nantes, France; (P.M.); (M.D.)
- Service de Médecine du Sport, CHU Nantes, Nantes Université, 44093 Nantes, France
- Institut Régional de Médecine du Sport (IRMS), 44093 Nantes, France
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, ONIRIS, Nantes Université, 44042 Nantes, France
| |
Collapse
|
23
|
Abstract
Joint pain is the hallmark symptom of osteoarthritis (OA) and the main reason for patients to seek medical assistance. OA pain greatly contributes to functional limitations of joints and reduced quality of life. Although several pain-relieving medications are available for OA treatment, the current intervention strategy for OA pain cannot provide satisfactory pain relief, and the chronic use of the drugs for pain management is often associated with significant side effects and toxicities. These observations suggest that the mechanisms of OA-related pain remain undefined. The current review mainly focuses on the characteristics and mechanisms of OA pain. We evaluate pathways associated with OA pain, such as nerve growth factor (NGF)/tropomyosin receptor kinase A (TrkA), calcitonin gene-related peptide (CGRP), C–C motif chemokine ligands 2 (CCL2)/chemokine receptor 2 (CCR2) and tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1β), the NOD-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3) inflammasome, and the Wnt/β-catenin signaling pathway. In addition, animal models currently used for OA pain studies and emerging preclinical studies are discussed. Understanding the multifactorial components contributing to OA pain could provide novel insights into the development of more specific and effective drugs for OA pain management.
Collapse
|
24
|
Kang D, Lee J, Jung J, Carlson BA, Chang MJ, Chang CB, Kang SB, Lee BC, Gladyshev VN, Hatfield DL, Lee BJ, Kim JH. Selenophosphate synthetase 1 deficiency exacerbates osteoarthritis by dysregulating redox homeostasis. Nat Commun 2022; 13:779. [PMID: 35140209 PMCID: PMC8828855 DOI: 10.1038/s41467-022-28385-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 01/24/2022] [Indexed: 12/13/2022] Open
Abstract
Aging and mechanical overload are prominent risk factors for osteoarthritis (OA), which lead to an imbalance in redox homeostasis. The resulting state of oxidative stress drives the pathological transition of chondrocytes during OA development. However, the specific molecular pathways involved in disrupting chondrocyte redox homeostasis remain unclear. Here, we show that selenophosphate synthetase 1 (SEPHS1) expression is downregulated in human and mouse OA cartilage. SEPHS1 downregulation impairs the cellular capacity to synthesize a class of selenoproteins with oxidoreductase functions in chondrocytes, thereby elevating the level of reactive oxygen species (ROS) and facilitating chondrocyte senescence. Cartilage-specific Sephs1 knockout in adult mice causes aging-associated OA, and augments post-traumatic OA, which is rescued by supplementation of N-acetylcysteine (NAC). Selenium-deficient feeding and Sephs1 knockout have synergistic effects in exacerbating OA pathogenesis in mice. Therefore, we propose that SEPHS1 is an essential regulator of selenium metabolism and redox homeostasis, and its dysregulation governs the progression of OA. Osteoarthritis is caused by the gradual accumulation of oxidative stress in cartilage. Here, the authors show that dysregulation of the selenium metabolic pathway underlies a shift in redox homeostasis in chondrocytes, leading to chronic osteoarthritic changes in joints.
Collapse
Affiliation(s)
- Donghyun Kang
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jeeyeon Lee
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea.,Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Jisu Jung
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Bradley A Carlson
- Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Moon Jong Chang
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Boramae Hospital, Seoul, 07061, South Korea
| | - Chong Bum Chang
- Department of Orthopaedic Surgery, Seoul National University Bundang Hospital, Seongnam, 13620, South Korea
| | - Seung-Baik Kang
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Boramae Hospital, Seoul, 07061, South Korea
| | - Byung Cheon Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Dolph L Hatfield
- Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Byeong Jae Lee
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, South Korea.
| | - Jin-Hong Kim
- Center for RNA Research, Institute for Basic Science, Seoul, 08826, South Korea. .,Department of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul, 08826, South Korea. .,Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
25
|
Drevet S, Favier B, Brun E, Gavazzi G, Lardy B. Mouse Models of Osteoarthritis: A Summary of Models and Outcomes Assessment. Comp Med 2022; 72:3-13. [PMID: 34986927 DOI: 10.30802/aalas-cm-21-000043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Osteoarthritis (OA) is a multidimensional health problem and a common chronic disease. It has a substantial impact onpatient quality of life and is a common cause of pain and mobility issues in older adults. The functional limitations, lack of curative treatments, and cost to society all demonstrate the need for translational and clinical research. The use of OA models in mice is important for achieving a better understanding of the disease. Models with clinical relevance are needed to achieve 2 main goals: to assess the impact of the OA disease (pain and function) and to study the efficacy of potential treatments. However, few OA models include practical strategies for functional assessment of the mice. OA signs in mice incorporate complex interrelations between pain and dysfunction. The current review provides a comprehensive compilation of mousemodels of OA and animal evaluations that include static and dynamic clinical assessment of the mice, merging evaluationof pain and function by using automatic and noninvasive techniques. These new techniques allow simultaneous recordingof spontaneous activity from thousands of home cages and also monitor environment conditions. Technologies such as videographyand computational approaches can also be used to improve pain assessment in rodents but these new tools must first be validated experimentally. An example of a new tool is the digital ventilated cage, which is an automated home-cage monitor that records spontaneous activity in the cages.
Collapse
|
26
|
Fouasson-Chailloux A, Dauty M, Bodic B, Masson M, Maugars Y, Metayer B, Veziers J, Lesoeur J, Rannou F, Guicheux J, Vinatier C. Posttraumatic Osteoarthritis Damage in Mice: From Histological and Micro-Computed Tomodensitometric Changes to Gait Disturbance. Cartilage 2021; 13:1478S-1489S. [PMID: 34696628 PMCID: PMC8804860 DOI: 10.1177/19476035211053821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES Osteoarthritis is a painful joint disease responsible for walking impairment. Its quantitative assessment by gait analysis in mice may be a relevant and noninvasive strategy to assess the disease severity. In this study, we aimed to determine the severity of osteoarthritis at the tissular and gait levels in unilateral and bilateral posttraumatic murine osteoarthritis. METHODS Twenty-four C57BL/6 male mice were randomly assigned to 3 groups (n = 8/group): controls, unilateral surgery, and bilateral surgery. Posttraumatic osteoarthritis was induced unilaterally or bilaterally by destabilization of the medial meniscus. Gait analysis was performed weekly with the CatWalkTM XT system until the 16th week after surgery. After animal sacrifices, histological and micro-computed tomographic assessment was performed. RESULTS Operated knees showed a significant increase in the histological score compared with controls (P < 0.001). Calcified anterior medial meniscal bone volume was higher on the ipsilateral side after unilateral destabilization of the medial meniscus (P < 0.001) and on both sides after bilateral intervention (P < 0.01). One week after surgery, the mice mean speed decreased significantly in both operated groups (P < 0.001 and P < 0.05). In the unilateral group, a significant increase in the contralateral hind print area appeared from week 4 to week 16. CONCLUSIONS While bilateral destabilization of the medial meniscus induced no detectable gait modification except 1 week after surgery, unilateral model was responsible for a gait disturbance on the contralateral side. Further studies are needed to better define the place of the CatWalkTM in the evaluation of mouse models of osteoarthritis.
Collapse
Affiliation(s)
- Alban Fouasson-Chailloux
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Service de Médecine Physique et
Réadaptation Locomotrice et Respiratoire, CHU Nantes, Nantes, France
- Service de Médecine du Sport, CHU
Nantes, Nantes, France
- UFR Odontologie, Université de Nantes,
Nantes, France
| | - Marc Dauty
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Service de Médecine Physique et
Réadaptation Locomotrice et Respiratoire, CHU Nantes, Nantes, France
- Service de Médecine du Sport, CHU
Nantes, Nantes, France
- UFR Odontologie, Université de Nantes,
Nantes, France
| | - Benoit Bodic
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- UFR Odontologie, Université de Nantes,
Nantes, France
| | - Martial Masson
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- CHU Nantes, Université de Nantes,
Nantes, France
| | - Yves Maugars
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- UFR Odontologie, Université de Nantes,
Nantes, France
- Service de Rhumatologie, CHU Nantes,
Nantes, France
| | - Benoit Metayer
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Service de Rhumatologie, CHU Nantes,
Nantes, France
| | - Joëlle Veziers
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- CHU Nantes, Université de Nantes,
Nantes, France
- PHU4 OTONN, CHU Nantes, Nantes,
France
| | - Julie Lesoeur
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- CHU Nantes, Université de Nantes,
Nantes, France
| | - François Rannou
- Service de Rééducation et de
Réadaptation de l’Appareil Locomoteur et des Pathologies du Rachis, Hôpitaux
Universitaires-Paris Centre, Groupe Hospitalier Cochin, Assistance Publique—Hôpitaux
de Paris, Paris, France
- INSERM UMRS 1124, Toxicité
Environnementale, Cibles Thérapeutiques, Signalisation Cellulaire et Biomarqueurs,
UFR Sciences Fondamentales et Biomédicales, Paris, France
- Université de Paris, Paris,
France
| | - Jérôme Guicheux
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- UFR Odontologie, Université de Nantes,
Nantes, France
- PHU4 OTONN, CHU Nantes, Nantes,
France
| | - Claire Vinatier
- Inserm, UMR 1229, Regenerative Medicine
and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- UFR Odontologie, Université de Nantes,
Nantes, France
| |
Collapse
|
27
|
Hsu GCY, Cherief M, Sono T, Wang Y, Negri S, Xu J, Peault B, James AW. Divergent effects of distinct perivascular cell subsets for intra-articular cell therapy in posttraumatic osteoarthritis. J Orthop Res 2021; 39:2388-2397. [PMID: 33512030 PMCID: PMC8319216 DOI: 10.1002/jor.24997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/30/2020] [Accepted: 01/24/2021] [Indexed: 02/04/2023]
Abstract
Intra-articular injection of mesenchymal stem cells has shown benefit for the treatment of osteoarthritis (OA). However, mesenchymal stem/stromal cells at the origin of these clinical results are heterogenous cell populations with limited cellular characterization. Here, two transgenic reporter mice were used to examine the differential effects of two precisely defined perivascular cell populations (Pdgfrα+ and Pdgfrβ+ cells) from white adipose tissue for alleviation of OA. Perivascular mesenchymal cells were isolated from transgenic Pdgfrα-and Pdgfrβ-CreERT2 reporter animals and delivered as a one-time intra-articular dose to C57BL/6J mice after destabilization of the medial meniscus (DMM). Both Pdgfrα+ and Pdgfrβ+ cell preparations improved metrics of cartilage degradation and reduced markers of chondrocyte hypertrophy. While some similarities in cell distribution were identified within the synovial and perivascular spaces, injected Pdgfrα+ cells remained in the superficial layers of articular cartilage, while Pdgfrβ+ cells were more widely dispersed. Pdgfrβ+ cell therapy prevented subchondral sclerosis induced by DMM, while Pdgfrα+ cell therapy had no effect. In summary, while both cell therapies showed beneficial effects in the DMM model, important differences in cell incorporation, persistence, and subchondral sclerosis were identified.
Collapse
Affiliation(s)
- Ginny Ching-Yun Hsu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Takashi Sono
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States;,Department of Orthopedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Stefano Negri
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Jiajia Xu
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| | - Bruno Peault
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, 90095;,Center For Cardiovascular Science and Center for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Ross Research Building, Room 524A, 720 Rutland Avenue, Baltimore, MD, 21205, United States
| |
Collapse
|
28
|
Zaki S, Smith MM, Little CB. Pathology-pain relationships in different osteoarthritis animal model phenotypes: it matters what you measure, when you measure, and how you got there. Osteoarthritis Cartilage 2021; 29:1448-1461. [PMID: 34332049 DOI: 10.1016/j.joca.2021.03.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine whether osteoarthritis (OA) pain characteristics and mechanistic pathways in pre-clinical models are phenotype-specific. DESIGN Male 11-week-old C57BL6 mice had unilateral medial-meniscal-destabilization (DMM) or antigen-induced-arthritis (AIA), vs sham-surgery/immunised-controls (Sham/Im-CT). Pain behaviour (allodynia, mechanical- and thermal-hyperalgesia, hindlimb static weight-bearing, stride-length) and lumbar dorsal root ganglia (DRG) gene-expression were measured at baseline, day-3, week-1/-2/-4/-8/-16, and pain-behaviour:gene-expression:joint-pathology associations investigated. RESULTS DMM and AIA induced structural OA defined by progressively increasing cartilage erosion, subchondral bone sclerosis and osteophyte size and maturation. All pain-behaviours were modified, with model-specific differences in severity and temporal pattern. Tactile allodynia developed acutely in both models and persisted to week-16. During early-OA (wk4-8) there was; reduced right hindlimb weight-bearing in AIA; thermal-hyperalgesia and reduced stride-length in DMM. During chronic-OA (wk12-16); mechanical-hyperalgesia and reduced right hindlimb weight-bearing were observed in DMM only. There were no associations in either model between different pain-behaviour outcomes. A coordinated DRG-expression profile was observed in sham and Im-CT for all 11 genes tested, but not in AIA and DMM. At wk-16 despite equivalent joint pathology, changes in DRG-expression (Calca, Trpa1, Trpv1, Trpv4) were observed only in DMM. In AIA mechanical-hyperalgesia was associated with Trpv1 (r = -0.79) and Il1b (r = 0.53). In DMM stride-length was associated with Calca, Tac1, Trpv1, Trpv2, Trpv4 and Adamts5 (r = 0.4-0.57). DRG gene-expression change was correlated with subchondral-bone sclerosis in DMM, and cartilage damage in AIA. Positive pain-behaviour:joint-pathology associations were only present in AIA - for synovitis, subchondral-bone resorption, chondrocyte-hypertrophy and cartilage damage. CONCLUSION Pain and peripheral sensory neuronal responses are OA-phenotype-specific with distinct pathology:pain-outcome:molecular-mechanism relationships.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, at Royal North Shore Hospital, Australia.
| | - M M Smith
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, at Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute of Medical Research, Faculty of Medicine and Health, The University of Sydney, at Royal North Shore Hospital, Australia.
| |
Collapse
|
29
|
Zhao R, Dong Z, Wei X, Gu X, Han P, Wu H, Yan Y, Huang L, Li H, Zhang C, Li F, Li P. Inflammatory factors are crucial for the pathogenesis of post-traumatic osteoarthritis confirmed by a novel porcine model: "Idealized" anterior cruciate ligament reconstruction" and gait analysis. Int Immunopharmacol 2021; 99:107905. [PMID: 34242997 DOI: 10.1016/j.intimp.2021.107905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine whether idealized anterior cruciate ligament reconstruction (IACL-R) restores normal gait features, and whether inflammatory factors are involved in the pathogenesisof post-traumatic osteoarthritis (PTOA). METHODS Fourteen mature female minipigs were allocated to a sham group (n = 7) or an IACL-R group (n = 7). Load asymmetry during gait was recorded using a pressure-sensing walkway measurement system to evaluate the gait features of the right knee joint before and after surgery. Inflammatory factors (including interleukin [IL]-1α, IL-1β, IL-2, IL-6, IL-8, IL-18, tumor necrosis factor-α, and granulocyte-macrophage colony-stimulating factor) in synovial fluid were measured using Luminex assays before and after surgery. Cartilage integrity and the subchondral bone plate of the right knee were evaluated using histology and imaging at 3 months postoperatively. RESULTS Swing time and stance time returned to their preoperative values on day 31, while maximum force, contact area, peak force ,and impulse returned to their preoperative values on day 45 after the surgery in the IACL-R group (P = 0.073, 0.053, 0.107, 0.052, 0.152, and 0.059, respectively).Thus, IACL-R restored normal gait. Compared with their preoperative concentrations, all tested inflammatory factors showed significantly increased concentrations in the synovial fluid in the IACL-R group, especially at 3, 7, and 15 days postoperatively. X-ray, computed tomography, magnetic resonance imaging, and histological data showed severe cartilage damage in the IACL-R model. CONCLUSION IACL-R restored normal gait features but caused significant cartilage damage, indicating that significantly elevated inflammatory factors maybe crucial for the pathogenesis of PTOA.
Collapse
Affiliation(s)
- Ruipeng Zhao
- Department of orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Zhengquan Dong
- Department of orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Xiaochun Wei
- Department of orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Xiaodong Gu
- Department of Orthopaedics, Bethune Hospital, Shanxi Medical University, Taiyuan 030032, PR China.
| | - Pengfei Han
- Department of Orthopedics, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi Province 046000, PR China.
| | - Hongru Wu
- Shanxi Institute of Sports Science, Taiyuan 030000, PR China.
| | - Yanxia Yan
- Department of orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Lingan Huang
- Department of orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Haoqian Li
- Department of orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Chengming Zhang
- Department of orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Fei Li
- Department of orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| | - Pengcui Li
- Department of orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, PR China.
| |
Collapse
|
30
|
Motherwell JM, Hendershot BD, Goldman SM, Dearth CL. Gait biomechanics: A clinically relevant outcome measure for preclinical research of musculoskeletal trauma. J Orthop Res 2021; 39:1139-1151. [PMID: 33458856 DOI: 10.1002/jor.24990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/01/2020] [Accepted: 01/11/2021] [Indexed: 02/04/2023]
Abstract
Traumatic injuries to the musculoskeletal system are the most prevalent of those suffered by United States Military Service members and accounts for two-thirds of initial hospital costs to the Department of Defense. These combat-related wounds often leave survivors with life-long disability and represent a significant impediment to the readiness of the fighting force. There are immense opportunities for the field of tissue engineering and regenerative medicine (TE/RM) to address these musculoskeletal injuries through regeneration of damaged tissues as a means to restore limb functionality and improve quality of life for affected individuals. Indeed, investigators have made promising advancements in the treatment for these injuries by utilizing small and large preclinical animal models to validate therapeutic efficacy of next-generation TE/RM-based technologies. Importantly, utilization of a comprehensive suite of functional outcome measures, particularly those designed to mimic data collected within the clinical setting, is critical for successful translation and implementation of these therapeutics. To that end, the objective of this review is to emphasize the clinical relevance and application of gait biomechanics as a functional outcome measure for preclinical research studies evaluating the efficacy of TE/RM therapies to treat traumatic musculoskeletal injuries. Specifically, common musculoskeletal injuries sustained by service members-including volumetric muscle loss, post-traumatic osteoarthritis, and composite tissue injuries-are examined as case examples to highlight the use of gait biomechanics as an outcome measure using small and large preclinical animal models.
Collapse
Affiliation(s)
- Jessica M Motherwell
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, Maryland, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Brad D Hendershot
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, Maryland, USA.,Department of Rehabilitation, Walter Reed National Military Medical Center, Bethesda, Maryland, USA.,Department of Rehabilitation Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Stephen M Goldman
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, Maryland, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Christopher L Dearth
- DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, Maryland, USA.,Department of Surgery, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Grol MW, Haelterman NA, Lim J, Munivez EM, Archer M, Hudson DM, Tufa SF, Keene DR, Lei K, Park D, Kuzawa CD, Ambrose CG, Eyre DR, Lee BH. Tendon and motor phenotypes in the Crtap-/- mouse model of recessive osteogenesis imperfecta. eLife 2021; 10:e63488. [PMID: 34036937 PMCID: PMC8186905 DOI: 10.7554/elife.63488] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 05/24/2021] [Indexed: 01/22/2023] Open
Abstract
Osteogenesis imperfecta (OI) is characterized by short stature, skeletal deformities, low bone mass, and motor deficits. A subset of OI patients also present with joint hypermobility; however, the role of tendon dysfunction in OI pathogenesis is largely unknown. Using the Crtap-/- mouse model of severe, recessive OI, we found that mutant Achilles and patellar tendons were thinner and weaker with increased collagen cross-links and reduced collagen fibril size at 1- and 4-months compared to wildtype. Patellar tendons from Crtap-/- mice also had altered numbers of CD146+CD200+ and CD146-CD200+ progenitor-like cells at skeletal maturity. RNA-seq analysis of Achilles and patellar tendons from 1-month Crtap-/- mice revealed dysregulation in matrix and tendon marker gene expression concomitant with predicted alterations in TGF-β, inflammatory, and metabolic signaling. At 4-months, Crtap-/- mice showed increased αSMA, MMP2, and phospho-NFκB staining in the patellar tendon consistent with excess matrix remodeling and tissue inflammation. Finally, a series of behavioral tests showed severe motor impairments and reduced grip strength in 4-month Crtap-/- mice - a phenotype that correlates with the tendon pathology.
Collapse
Affiliation(s)
- Matthew William Grol
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Nele A Haelterman
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Joohyun Lim
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Elda M Munivez
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Marilyn Archer
- Department of Orthopaedics and Sports Medicine, University of WashingtonSeattleUnited States
| | - David M Hudson
- Department of Orthopaedics and Sports Medicine, University of WashingtonSeattleUnited States
| | - Sara F Tufa
- Shriners Hospital for ChildrenPortlandUnited States
| | | | - Kevin Lei
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Dongsu Park
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Cole D Kuzawa
- Department of Orthopaedic Surgery, UT Health Sciences CenterHoustonUnited States
| | - Catherine G Ambrose
- Department of Orthopaedic Surgery, UT Health Sciences CenterHoustonUnited States
| | - David R Eyre
- Department of Orthopaedics and Sports Medicine, University of WashingtonSeattleUnited States
| | - Brendan H Lee
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
32
|
Andrographolide attenuates synovial inflammation of osteoarthritis by interacting with tumor necrosis factor receptor 2 trafficking in a rat model. J Orthop Translat 2021; 29:89-99. [PMID: 34094861 PMCID: PMC8144533 DOI: 10.1016/j.jot.2021.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 02/08/2023] Open
Abstract
Background Synovial inflammation plays a major role in the pathogenesis of osteoarthritis (OA). This study investigated the effect of andrographolide (Andro) on synovial inflammation mediated by tumor necrosis factor-alpha receptor 2 (TNFR2) trafficking and its utility in attenuating OA progression. Methods Knee joints were harvested from rats subjected to radial transection of the medial collateral ligament (MCLT) and medial meniscus (MMT) to examine the effect of Andro on synovial inflammation and OA progression. Quantitative real-time polymerase chain reaction was used to evaluate the expression of inflammatory factors in primary fibroblast-like synoviocytes (FLSs) after Andro treatment in vitro. The mechanism underlying Andro-mediated regulation of TNFR2 distribution and nuclear factor-κB (NF-κB) expression was verified using endosome maturation inhibitor hydroxychloroquine (HCQ) through flow cytometry, immunofluorescence, and western blot analysis. Results Andro treatment was found to reduce synovial inflammation and OA progression in vivo. Furthermore, a decrease in pain hypersensitivity and dorsal horn neuron activation was observed after treatment. Andro also downregulated the expression of inflammatory mediators and TNFR2 in FLSs. TNFR2 is crucial for the activation of the NF-κB signaling pathway, and Andro-induced degradation of TNFR2 was associated with lysosomal function, which in turn, reduced the downstream phosphorylation of p65 in the NF-κB signaling pathway. Conclusions Andro could suppress synovial inflammation via regulation of TNFR2 trafficking and degradation. This also suggests it could be a potential treatment for the prevention of synovial inflammation and OA progression. The translational potential of this article This study provides strong evidence that Andro reduces NF-κB activation and inflammatory responses in OA FLSs via regulation of TNFR2 trafficking. The inhibition of TNFR2 and Andro could be a novel therapeutic approach for OA and pain management.
Collapse
|
33
|
Berke IM, Jain E, Yavuz B, McGrath T, Chen L, Silva MJ, Mbalaviele G, Guilak F, Kaplan DL, Setton LA. NF-κB-mediated effects on behavior and cartilage pathology in a non-invasive loading model of post-traumatic osteoarthritis. Osteoarthritis Cartilage 2021; 29:248-256. [PMID: 33246158 PMCID: PMC8023431 DOI: 10.1016/j.joca.2020.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/25/2020] [Accepted: 10/13/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This study aimed to examine the temporal activation of NF-κB and its relationship to the development of pain-related sensitivity and behavioral changes in a non-invasive murine knee loading model of PTOA. METHOD Following knee injury NF-κB activity was assessed longitudinally via in vivo imaging in FVB. Cg-Tg (HIV-EGFP,luc)8Tsb/J mice. Measures of pain-related sensitivity and behavior were also assessed longitudinally for 16 weeks. Additionally, we antagonized NF-κB signaling via intra-articular delivery of an IκB kinase two antagonist to understand how local NF-κB inhibition might alter disease progression. RESULTS Following joint injury NF-κB signaling within the knee joint was transiently increased and peaked on day 3 with an estimated 1.35 p/s/cm2/sr (95% CI 0.913.1.792 p/s/cm2/sr) fold increase in signaling when compared to control joints. Furthermore, injury resulted in the long-term development of hindpaw allodynia. Hyperalgesia withdrawal thresholds were reduced at injured knee joints, with the largest reduction occurring 2 days following injury (estimate of between group difference 129.1 g with 95% CI 60.9,197.4 g), static weight bearing on injured limbs was also reduced. Local delivery of an NF-κB inhibitor following joint injury reduced chondrocyte death and influenced the development of pain-related sensitivity but did not reduce long-term cartilage degeneration. CONCLUSION These findings underscore the development of behavioral changes in this non-invasive loading model of PTOA and their relationships to NF-κB activation and pathology. They also highlight the potential chondroprotective effects of NF-κB inhibition shortly following joint injury despite limitations in preventing the long-term development of joint degeneration in this model of PTOA.
Collapse
Affiliation(s)
- I M Berke
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - E Jain
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - B Yavuz
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA, 02155, USA
| | - T McGrath
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - L Chen
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - M J Silva
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA; Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA; Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - G Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA; Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - F Guilak
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA; Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA; Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO, 63110, USA
| | - D L Kaplan
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA, 02155, USA
| | - L A Setton
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA; Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA; Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
34
|
Faust HJ, Zhang H, Han J, Wolf MT, Jeon OH, Sadtler K, Peña AN, Chung L, Maestas DR, Tam AJ, Pardoll DM, Campisi J, Housseau F, Zhou D, Bingham CO, Elisseeff JH. IL-17 and immunologically induced senescence regulate response to injury in osteoarthritis. J Clin Invest 2020; 130:5493-5507. [PMID: 32955487 PMCID: PMC7524483 DOI: 10.1172/jci134091] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 07/09/2020] [Indexed: 12/17/2022] Open
Abstract
Senescent cells (SnCs) are implicated in the pathogenesis of age-related diseases including osteoarthritis (OA), in part via expression of a senescence-associated secretory phenotype (SASP) that includes immunologically relevant factors and cytokines. In a model of posttraumatic OA (PTOA), anterior cruciate ligament transection (ACLT) induced a type 17 immune response in the articular compartment and draining inguinal lymph nodes (LNs) that paralleled expression of the senescence marker p16INK4a (Cdkn2a) and p21 (Cdkn1a). Innate lymphoid cells, γδ+ T cells, and CD4+ T cells contributed to IL-17 expression. Intra-articular injection of IL-17-neutralizing antibody reduced joint degeneration and decreased expression of the senescence marker Cdkn1a. Local and systemic senolysis was required to attenuate tissue damage in aged animals and was associated with decreased IL-17 and increased IL-4 expression in the articular joint and draining LNs. In vitro, we found that Th17 cells induced senescence in fibroblasts and that SnCs skewed naive T cells toward Th17 or Th1, depending on the presence of TGF-β. The SASP profile of the inflammation-induced SnCs included altered Wnt signaling, tissue remodeling, and cell-cycle pathways not previously implicated in senescence. These findings provide molecular targets and mechanisms for senescence induction and therapeutic strategies to support tissue healing in an aged environment.
Collapse
Affiliation(s)
- Heather J. Faust
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hong Zhang
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Matthew T. Wolf
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ok Hee Jeon
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, South Korea
| | - Kaitlyn Sadtler
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexis N. Peña
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liam Chung
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David R. Maestas
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ada J. Tam
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and
| | - Drew M. Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Daohong Zhou
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Clifton O. Bingham
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer H. Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and
| |
Collapse
|
35
|
Effectiveness of Led Photobiomodulation Therapy on Treatment With Knee Osteoarthritis: A Rat Study. Am J Phys Med Rehabil 2020; 99:725-732. [PMID: 32167952 DOI: 10.1097/phm.0000000000001408] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The present study aimed to evaluate the effectiveness of photobiomodulation therapy by light-emitting diode on osteoarthritis treatment in the knees of rats. DESIGN Twenty male Wistar rats were randomly assigned into two experimental groups: OAC: animals subjected to induction of osteoarthritis, without therapeutic intervention and the group OAL: animals subjected to induction of osteoarthritis treated with light-emitting diode photobiomodulation therapy (850 nm, 200 mW, 6 J). RESULTS The results of gait analysis showed no statistical difference between the groups. The histological findings showed that the OAL group presented abnormal chondrocyte orientation, yet with less irregularities along fibrillation and the joint tissue. Thus, it presented a lower degenerative process when evaluated by the Osteoarthritis Research Society International. Likewise, in the immunohistochemical analysis, the OAL group showed higher collagen 2 and transforming growth factor β immunoexpression when compared with the OAC group. CONCLUSIONS Given the above, it is possible to suggest that the photobiomodulation therapy by light-emitting diode had positive effects on the expression of extracellular matrix proteins responsible for synthesis of articular tissue.
Collapse
|
36
|
Barbosa GM, Cunha JE, Russo TL, Cunha TM, Castro PATS, Oliveira FFB, Cunha FQ, Ramalho FS, Salvini TF. Thirty days after anterior cruciate ligament transection is sufficient to induce signs of knee osteoarthritis in rats: pain, functional impairment, and synovial inflammation. Inflamm Res 2020; 69:279-288. [PMID: 32006078 DOI: 10.1007/s00011-020-01317-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/15/2019] [Accepted: 01/08/2020] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE To compare the unilateral signs of knee osteoarthritis (KOA) 30 and 60 days after anterior cruciate ligament transection (ACLT). Pain, gait function, synovial fluid inflammation, and histopathological changes in the synovial membrane were analyzed, as well as the interaction between the variables. MATERIALS AND METHODS Male Wistar rats (n = 32; 219.2 ± 18.6 g) were randomly distributed into four groups of eight animals each. Two groups were submitted to unilateral ACLT surgery to induce KOA and analyzed after 30 (KOA30) and 60 days (KOA60). Two control groups (without surgery) were also assessed after the same time periods (C30 and C60). All the groups were evaluated before ACLT from the least to most stressful tests (skin temperature, mechanical response threshold, gait test, thermal response threshold, and joint swelling), as well as 30 and 60 days after surgery. After euthanasia, the synovial fluid and synovial membrane were collected. RESULTS Thirty days after ACLT, KOA30 showed decrease paw print area and mechanical response threshold, higher joint swelling, skin temperature, leukocyte count, cytokine levels, and synovitis score. No differences were found between KOA30 and KOA60. CONCLUSION Our data showed that 30 days after ACLT is sufficient to induce signs of KOA in rats, such as pain, functional impairment, and synovial inflammation, suggesting that a shorter time period can be used as an experimental model.
Collapse
Affiliation(s)
- Germanna M Barbosa
- Department of Physical Therapy, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Jonathan E Cunha
- Department of Physical Therapy, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Thiago L Russo
- Department of Physical Therapy, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, University of Sao Paulo, Ribeirão Preto, SP, Brazil
| | - Paula A T S Castro
- Department of Physical Therapy, Federal University of São Carlos, São Carlos, SP, Brazil
| | | | - Fernando Q Cunha
- Department of Pharmacology, University of Sao Paulo, Ribeirão Preto, SP, Brazil
| | - Fernando S Ramalho
- Department of Pathology and Forensic Medicine, University of Sao Paulo, Ribeirão Preto, SP, Brazil
| | - Tania F Salvini
- Department of Physical Therapy, Federal University of São Carlos, São Carlos, SP, Brazil. .,Laboratório de Plasticidade Muscular, Departamento de Fisioterapia, Universidade Federal de São Carlos, Rodovia Washington Luiz, Km 235, São Carlos, SP, 13565-905, Brazil.
| |
Collapse
|
37
|
Understanding the Molecular Mechanisms Underlying the Pathogenesis of Arthritis Pain Using Animal Models. Int J Mol Sci 2020; 21:ijms21020533. [PMID: 31947680 PMCID: PMC7013391 DOI: 10.3390/ijms21020533] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/27/2019] [Accepted: 01/09/2020] [Indexed: 12/22/2022] Open
Abstract
Arthritis, including osteoarthritis (OA) and rheumatoid arthritis (RA), is the leading cause of years lived with disability (YLD) worldwide. Although pain is the cardinal symptom of arthritis, which is directly related to function and quality of life, the elucidation of the mechanism underlying the pathogenesis of pain in arthritis has lagged behind other areas, such as inflammation control and regulation of autoimmunity. The lack of therapeutics for optimal pain management is partially responsible for the current epidemic of opioid and narcotic abuse. Recent advances in animal experimentation and molecular biology have led to significant progress in our understanding of arthritis pain. Despite the inherent problems in the extrapolation of data gained from animal pain studies to arthritis in human patients, the critical assessment of molecular mediators and translational studies would help to define the relevance of novel therapeutic targets for the treatment of arthritis pain. This review discusses biological and molecular mechanisms underlying the pathogenesis of arthritis pain determined in animal models of OA and RA, along with the methodologies used.
Collapse
|
38
|
Barbosa GM, Cunha JE, Cunha TM, Martinho LB, Castro PATS, Oliveira FFB, Cunha FQ, Ramalho FS, Salvini TF. Clinical-like cryotherapy improves footprint patterns and reduces synovial inflammation in a rat model of post-traumatic knee osteoarthritis. Sci Rep 2019; 9:14518. [PMID: 31601862 PMCID: PMC6787208 DOI: 10.1038/s41598-019-50958-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 09/17/2019] [Indexed: 12/16/2022] Open
Abstract
Cryotherapy is a non-pharmacological treatment commonly used to control inflammation and improve function after acute traumas. However, there are no definitive findings about its effects on chronic joint diseases such as knee osteoarthritis (KOA). The aim of this study was to investigate the effects of clinical-like cryotherapy on functional impairment and synovial inflammation in a rat model of KOA generated by anterior cruciate ligament transection (ACLT). Thirty-two male Wistar rats were randomly divided into four groups (n = 8/group): Control, KOA, KOA + Cryotherapy and KOA + Placebo. The last two groups were submitted to the relevant interventions twice a day for five days (61 to 65), with each session lasting 20 min. Gait test, skin temperature, thermal response threshold and joint swelling were assessed in all groups before ACLT surgery, and pre (60th day) and post (66th day) intervention protocols. On day 66, the animals were euthanized and exsanguinated to remove the synovial membrane for histopathological examination and synovial fluid to determine the leukocyte count and cytokine concentration. After the intervention period (66th day), footprint area only increased in the KOA + Cryotherapy group (P = 0.004; 14%) when compared to KOA and KOA + Placebo, but did not differ from controls. Cryotherapy lowered the synovial fluid leukocyte count (P < 0.0001; ≥95.0%) and cytokine concentration (P < 0.0001; ≥55%) when compared to the KOA and Placebo groups. Synovial score and synovial fibrosis did not differ in the KOA groups. In conclusion, footprint patterns improved in rats with ACLT-induced KOA as a result of clinical-like cryotherapy, which also lowered the synovial fluid leukocyte count and inflammatory cytokine concentration in these rats.
Collapse
Affiliation(s)
| | - Jonathan Emanuel Cunha
- Department of Physical Therapy, Federal University of São Carlos, São Carlos, SP, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | | | | | - Fernando Silva Ramalho
- Department of Pathology and Forensic Medicine, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Tania Fátima Salvini
- Department of Physical Therapy, Federal University of São Carlos, São Carlos, SP, Brazil.
| |
Collapse
|
39
|
Cunha JE, Barbosa GM, Castro PATDS, Luiz BLF, Silva ACA, Russo TL, Vasilceac FA, Cunha TM, Cunha FQ, Salvini TF. Knee osteoarthritis induces atrophy and neuromuscular junction remodeling in the quadriceps and tibialis anterior muscles of rats. Sci Rep 2019; 9:6366. [PMID: 31019213 PMCID: PMC6482306 DOI: 10.1038/s41598-019-42546-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 04/01/2019] [Indexed: 12/21/2022] Open
Abstract
Knee osteoarthritis (KOA) is associated with muscle weakness, but it is unclear which structures are involved in the muscle changes. This study assessed morphological alterations and the expression of genes and proteins linked to muscular atrophy and neuromuscular junctions (NMJs) in KOA, induced by anterior cruciate ligament transection (ACLT) in rats. Two groups of rats were assessed: control (without intervention) and KOA (ACLT surgery in the right knee). After 8 weeks, quadriceps, tibialis anterior (TA) and gastrocnemius muscles were analyzed (area of muscle fibers, NMJ, gene and protein expression). KOA group showed atrophy in quadriceps (15.7%) and TA (33%), with an increase in atrogin-1 and muscle RING-finger protein-1 (MuRF-1). KOA group showed quadriceps NMJ remodeling (reduction area and perimeter) and decrease in NMJ diameter in TA muscle. The expression of nicotinic acetylcholine receptor (nAChR) γ-nAChR increased and that of α-nAChR and muscle specific tyrosine kinase (MuSK) declined in the quadriceps, with a decrease in ε-nAChR in TA. MuRF-1 protein expression increased in quadriceps and TA, with no changes in neural cell adhesion molecule (NCAM). In conclusion, ACLT-induced KOA promotes NMJ remodeling and atrophy in quadriceps and TA muscles, associated with inflammatory signs and changes in muscle gene and protein expression.
Collapse
Affiliation(s)
| | | | | | | | | | - Thiago Luiz Russo
- Physical Therapy Department, Federal University of São Carlos, São Carlos, SP, Brazil
| | | | - Thiago Mattar Cunha
- Pharmacology Department, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Tania Fátima Salvini
- Physical Therapy Department, Federal University of São Carlos, São Carlos, SP, Brazil.
| |
Collapse
|
40
|
Chavez RD, Sohn P, Serra R. Prg4 prevents osteoarthritis induced by dominant-negative interference of TGF-ß signaling in mice. PLoS One 2019; 14:e0210601. [PMID: 30629676 PMCID: PMC6328116 DOI: 10.1371/journal.pone.0210601] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 12/28/2018] [Indexed: 11/20/2022] Open
Abstract
Objective Prg4, also known as Lubricin, acts as a joint/boundary lubricant. Prg4 has been used to prevent surgically induced osteoarthritis (OA) in mice. Surgically induced OA serves as a good model for post-traumatic OA but is not ideal for recapitulating age-related OA. Reduced expression of the TGF-β type II receptor (TGFβR2) is associated with age-related OA in clinical samples, so we previously characterized a mouse model that exhibits OA due to expression of a mutated dominant-negative form of TGFβR2 (DNIIR). Prg4 expression was significantly reduced in DNIIR mice. Furthermore, we showed that Prg4 was a transcriptional target of TGF-ß via activation of Smad3, the main signal transducing protein for TGF-ß. The objective of the present study was to determine whether maintenance of Prg4, a down-stream transcriptional target of TGF-ß, prevents OA associated with attenuated TGF-ß signaling in mice. Design Wild-type, DNIIR, and bitransgenic mice that express both DNIIR and Prg4, were compared. Mice were assessed with a foot misplacement behavioral test, μCT, histology, and Western blot. Results Compared to DNIIR mice, bitransgenic DNIIR+Prg4 mice missed 1.3 (0.4, 2.1) fewer steps while walking (mean difference (95% confidence interval)), exhibited a cartilage fibrillation score that was 1.8 (0.4, 3.1) points lower, exhibited cartilage that was 28.2 (0.5, 55.9) μm thicker, and exhibited an OARSI score that was 6.8 (-0.9, 14.5) points lower. However, maintenance of Prg4 expression did not restore levels of phosphorylated Smad3 in DNIIR mice, indicating Prg4 does not simply stimulate TGF-ß signaling. Conclusions Our results indicate that maintenance of Prg4 expression prevents OA progression associated with reduced TGF-β signaling in mice. Since there was no evidence that Prg4 acts by stimulating the TGF-ß signaling cascade, we propose that Prg4, a transcriptional target of TGF-ß, attenuates OA progression through its joint lubrication function.
Collapse
Affiliation(s)
- Robert Dalton Chavez
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Philip Sohn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Rosa Serra
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
41
|
Thompson AL, Largent-Milnes TM, Vanderah TW. Animal Models for the Study of Bone-Derived Pain. Methods Mol Biol 2019; 1914:391-407. [PMID: 30729479 DOI: 10.1007/978-1-4939-8997-3_23] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bone pain is a prevalent issue in society today and also is one of the hardest types of pain to control. Pain originating in the bone can be caused by many different entities including metastatic and primary neoplasm, fracture, osteoarthritis as well as numerous other metabolic disorders. In this chapter we describe the methods and protocols that currently are accepted and validated for the study of bone pain in models of metastatic cancer, bicortical fracture and osteoarthritis. These animal models provide invaluable information as to the nature of bone pain and give rise to potential new targets for its treatment and management.
Collapse
Affiliation(s)
- Austen L Thompson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | | | - Todd W Vanderah
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
42
|
Chou YJ, Chuu JJ, Peng YJ, Cheng YH, Chang CH, Chang CM, Liu HW. The potent anti-inflammatory effect of Guilu Erxian Glue extracts remedy joint pain and ameliorate the progression of osteoarthritis in mice. J Orthop Surg Res 2018; 13:259. [PMID: 30340603 PMCID: PMC6194592 DOI: 10.1186/s13018-018-0967-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 10/05/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a slow progressing, degenerative disorder of the synovial joints. Guilu Erxian Glue (GEG) is a multi-component Chinese herbal remedy with long-lasting favorable effects on several conditions, including articular pain and muscle strength in elderly men with knee osteoarthritis. The present study aimed to identify the effects of Guilu Erxian Paste (GE-P) and Liquid (GE-L) extracted from Guilu Erxian Glue in anterior cruciate ligament transection (ACLT)-induced osteoarthritis mice, and to compare the effectiveness of different preparations on knee cartilage degeneration during the progression of osteoarthritis. METHODS Male C57BL/6J mice underwent anterior cruciate ligament transection to induce mechanically destabilized osteoarthritis in the right knee. 4 weeks later, the mice were orally treated with PBS, celecoxib (10 mg/kg/day), Guilu Erxian Paste (100 or 300 mg/kg/day), and Guilu Erxian Liquid (100 or 300 mg/kg/day) for 28 consecutive days. Von Frey and open-field tests (OFT) were used to evaluate pain behaviors (mechanical hypersensitivity and locomotor performance). Narrowing of the joint space and osteophyte formation were examined radiographically. Inflammatory cytokine (IL-1β, IL-6, and TNF-α) levels in the articular cartilage were determined by quantitative real-time PCR. Histopathological examinations were conducted to evaluate the severity and extent of the cartilage lesions. RESULTS Guilu Erxian Paste and Guilu Erxian Liquid (300 mg/kg/day) were significantly more effective (p < 0.01) than celecoxib (10 mg/kg/day) in decreasing secondary allodynia when compared to the saline-treated group (#p < 0.05). Open-field tests revealed no significant motor dysfunction between the Guilu Erxian Paste- and Guilu Erxian Liquid-treated mice compared to the saline-treated mice. Radiographic findings also confirmed that the administration of Guilu Erxian Paste and Guilu Erxian Liquid (100 and 300 mg/kg/day) significantly and dose-dependently reduced osteolytic lesions and bone spur formation in the anterior cruciate ligament transection-induced osteoarthritis mice when compared to the saline-treated group. Notably, Guilu Erxian Liquid (100 mg/kg/day) treatment significantly reduced the mRNA levels of IL-1β, IL-6, and TNF-α as well as relative the protein expression of IL-1β and TNF-α to the effect of celecoxib. Guilu Erxian Paste and Guilu Erxian Liquid (300 mg/kg/day) markedly attenuated cartilage destruction, surface unevenness, proteoglycan loss, chondrocyte degeneration, and cartilage erosion in the superficial layers (##p < 0.01 and ###p < 0.001 respectively). CONCLUSIONS As expected, our findings suggest that the anti-inflammatory effects of Guilu Erxian Liquid (GE-L), following marked decrease on both IL-1β and TNF-α during the early course of post-traumatic osteoarthrosis (OA), may be of potential value in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Yen-Jung Chou
- Department of Traditional Chinese Medicine, MacKay Memorial Hospital, Taipei City, Taiwan
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Applied Science and Engineering, Fu Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City, 24205 Taiwan
| | - Jiunn-Jye Chuu
- Department of Biotechnology, College of Engineering, Southern Taiwan University, Tainan City, Taiwan
| | - Yi-Jen Peng
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei City, Taiwan
| | - Yu-Hsuan Cheng
- Department of Biotechnology, College of Engineering, Southern Taiwan University, Tainan City, Taiwan
| | - Chin-Hsien Chang
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Tao-Yuan City, Taiwan
- Department of Traditional Chinese Medicine, En Chu Kong Hospital, New Taipei City, 237 Taiwan
| | - Chieh-Min Chang
- Department of Traditional Chinese Medicine, En Chu Kong Hospital, New Taipei City, 237 Taiwan
| | - Hsia-Wei Liu
- Department of Life Science, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Applied Science and Engineering, Fu Jen Catholic University, No. 510, Zhongzheng Rd., Xinzhuang Dist., New Taipei City, 24205 Taiwan
| |
Collapse
|
43
|
Nixon AJ, Grol MW, Lang HM, Ruan MZC, Stone A, Begum L, Chen Y, Dawson B, Gannon F, Plutizki S, Lee BHL, Guse K. Disease-Modifying Osteoarthritis Treatment With Interleukin-1 Receptor Antagonist Gene Therapy in Small and Large Animal Models. Arthritis Rheumatol 2018; 70:1757-1768. [PMID: 30044894 DOI: 10.1002/art.40668] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 07/10/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Gene therapy holds great promise for the treatment of osteoarthritis (OA) because a single intraarticular injection can lead to long-term expression of therapeutic proteins within the joint. This study was undertaken to investigate the use of a helper-dependent adenovirus (HDAd)-mediated intraarticular gene therapy approach for long-term expression of interleukin-1 receptor antagonist (IL-1Ra) as sustained symptomatic and disease-modifying therapy for OA. METHODS In mouse models of OA, efficacy of HDAd-IL-1Ra was evaluated by histologic analysis, micro-computed tomography (micro-CT), and hot plate analysis. In a horse OA model, safety and efficacy of HDAd-IL-1Ra were evaluated by blood chemistry, analyses of synovial fluid, synovial membrane, and cartilage, and gross pathology and lameness assessments. RESULTS In skeletally immature mice, HDAd-IL-1Ra prevented development of cartilage damage, osteophytes, and synovitis. In skeletally immature and mature mice, treatment with HDAd-interleukin-1 receptor antagonist post-OA induction resulted in improved-albeit not significantly-cartilage status assessed histologically and significantly increased cartilage volume, cartilage surface, and bone surface covered by cartilage as assessed by micro-CT. Fewer osteophytes were observed in HDAd-IL-1Ra-treated skeletally immature mice. Synovitis was not affected in skeletally immature or mature mice. HDAd-IL-1Ra protected against disease-induced thermal hyperalgesia in skeletally mature mice. In the horse OA model, HDAd-IL-1Ra therapy significantly improved lameness parameters, indicating efficient symptomatic treatment. Moreover, macroscopically and histologically assessed cartilage and synovial membrane parameters were significantly improved, suggesting disease-modifying efficacy. CONCLUSION These data from OA models in small and large animals demonstrated safe symptomatic and disease-modifying treatment with an HDAd-expressing IL-1Ra. Furthermore, this study establishes HDAd as a vector for joint gene therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Kilian Guse
- Baylor College of Medicine, Houston, Texas, and GeneQuine Biotherapeutics GmbH, Hamburg, Germany
| |
Collapse
|
44
|
Faust HJ, Sommerfeld SD, Rathod S, Rittenbach A, Ray Banerjee S, Tsui BMW, Pomper M, Amzel ML, Singh A, Elisseeff JH. A hyaluronic acid binding peptide-polymer system for treating osteoarthritis. Biomaterials 2018; 183:93-101. [PMID: 30149233 DOI: 10.1016/j.biomaterials.2018.08.045] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/13/2018] [Accepted: 08/20/2018] [Indexed: 01/20/2023]
Abstract
Hyaluronic acid (HA) is found naturally in synovial fluid and is utilized therapeutically to treat osteoarthritis (OA). Here, we employed a peptide-polymer cartilage coating platform to localize HA to the cartilage surface for the purpose of treating post traumatic osteoarthritis. The objective of this study was to increase efficacy of the peptide-polymer platform in reducing OA progression in a mouse model of post-traumatic OA without exogenous HA supplementation. The peptide-polymer is composed of an HA-binding peptide (HABP) conjugated to a heterobifunctional poly (ethylene glycol) (PEG) chain and a collagen binding peptide (COLBP). We created a library of different peptide-polymers and characterized their HA binding properties in vitro using quartz crystal microbalance (QCM-D) and isothermal calorimetry (ITC). The peptide polymers were further tested in vivo in an anterior cruciate ligament transection (ACLT) murine model of post traumatic OA. The peptide-polymer with the highest affinity to HA as tested by QCM-D (∼4-fold greater binding compared to other peptides tested) and by ITC (∼3.8-fold) was HABP2-8-arm PEG-COLBP. Biotin tagging demonstrated that HABP2-8-arm PEG-COLBP localizes to both cartilage defects and synovium. In vivo, HABP2-8-arm PEG-COLBP treatment and the clinical HA comparator Orthovisc lowered levels of inflammatory genes including IL-6, IL-1B, and MMP13 compared to saline treated animals and increased aggrecan expression in young mice. HABP2-8-arm PEG-COLBP and Orthovisc also reduced pain as measured by incapacitance and hotplate testing. Cartilage degeneration as measured by OARSI scoring was also reduced by HABP2-8-arm PEG-COLBP and Orthovisc. In aged mice, HABP2-8-arm PEG-COLBP therapeutic efficacy was similar to its efficacy in young mice, but Orthovisc was less efficacious and did not significantly improve OARSI scoring. These results demonstrate that HABP2-8-arm PEG-COLBP is effective at reducing PTOA progression.
Collapse
Affiliation(s)
- Heather J Faust
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Sven D Sommerfeld
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Sona Rathod
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Andrew Rittenbach
- Information Sciences Institute, University of Southern California, Arlington, VA 22203, USA
| | | | - Benjamin M W Tsui
- Department of Radiology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Martin Pomper
- Department of Radiology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Mario L Amzel
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Anirudha Singh
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21287, USA; Department of Urology, The James Buchanan Brady Urological Institute, The Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21287, USA.
| |
Collapse
|
45
|
Muscle wasting in osteoarthritis model induced by anterior cruciate ligament transection. PLoS One 2018; 13:e0196682. [PMID: 29709011 PMCID: PMC5927423 DOI: 10.1371/journal.pone.0196682] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 04/17/2018] [Indexed: 12/02/2022] Open
Abstract
This study aimed to investigate the molecular pathways involved in muscle wasting in an animal model of osteoarthritis (OA) induced by anterior cruciate ligament transection (ACLT) in rats. Reduction of protein syntheses, increased proteolysis and impaired muscle regeneration are important pathways related to muscle wasting, and myogenin, MyoD, myostatin and MuRF-1 are some of their markers. Female Wistar rats were allocated into two groups: OA (submitted to the ACLT) and SHAM (submitted to surgery without ACLT). Nociception, spontaneous exploratory locomotion and body weight of animals were evaluated weekly. Twelve weeks after the disease induction, animals were euthanized, and the right knee joints were collected. Gastrocnemius muscle of the right hind paw were dissected and weighed. Gastrocnemius was used for evaluation of muscle atrophy and expression of IL-1β, TNF-α, Pax7, myogenin, MyoD, myostatin and MuRF-1. Histopathology of the knee confirmed the development of the disease in animals of OA group. Gastrocnemius of OA animals showed a reduction of about 10% in area and an increased IL-1β expression compared to animals of SHAM group. Expression of myostatin was increased in OA group, while myogenin expression was decreased. TNF-α, Pax7, MuRF-1 and MyoD expression was similar in both OA and SHAM groups. Nociception was significantly elevated in OA animals in the last two weeks of experimental period. Spontaneous exploratory locomotion, body weight and weight of gastrocnemius showed no difference between OA and SHAM groups. Gastrocnemius atrophy in OA induced by ACLT involves elevated expression of IL-1β within the muscle, as well as increased expression of myostatin and decreased expression of myogenin. Therefore, muscle wasting may be linked to impaired muscle regeneration.
Collapse
|
46
|
Grol MW, Lee BH. Gene therapy for repair and regeneration of bone and cartilage. Curr Opin Pharmacol 2018; 40:59-66. [PMID: 29621661 DOI: 10.1016/j.coph.2018.03.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 03/12/2018] [Indexed: 12/28/2022]
Abstract
Gene therapy refers to the use of viral and non-viral vectors to deliver nucleic acids to tissues of interest using direct (in vivo) or transduced cell-mediated (ex vivo) approaches. Over the past few decades, strategies have been adopted to express therapeutic transgenes at sites of injury to promote or facilitate repair of bone and cartilage. Targets of interest have typically included secreted proteins such as growth factors and anti-inflammatory mediators; however, work has also begun to focus intracellularly on signaling components, transcription factors and small, regulatory nucleic acids such as microRNAs (miRNAs). In recent years, a number of single therapeutic gene approaches (termed 'monotherapies') have proven effective in preclinical models of disease, and several are being evaluated in clinical trials. In particular, an ex vivo TGF-β1 gene therapy was approved in Korea in 2017 for treatment of moderate-to-severe osteoarthritis (OA). The ability to utilize viral vectors for context-specific and combinatorial gene therapy is also being investigated, and these strategies are likely to be important in more robustly addressing the complexities of tissue repair and regeneration in skeletal disease. In this review, we provide an overview of viral gene therapies being developed for treatment of bone and cartilage pathologies, with an emphasis on emerging combinatorial strategies as well as those targeting intracellular mediators such as miRNAs.
Collapse
Affiliation(s)
- Matthew W Grol
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brendan H Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
47
|
Tsai HC, Chen TL, Chen YP, Chen RM. Traumatic osteoarthritis-induced persistent mechanical hyperalgesia in a rat model of anterior cruciate ligament transection plus a medial meniscectomy. J Pain Res 2017; 11:41-50. [PMID: 29317848 PMCID: PMC5743113 DOI: 10.2147/jpr.s154038] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Osteoarthritis (OA) is a degenerative joint disease characterized by progressive cartilage degeneration, subchondral bone changes, osteophyte formation, and synovitis. A major symptom is pain that is triggered by peripheral and central changes within the pain pathways. Some surgery-induced joint instability rat models of OA were described to mimic traumatic OA. Several behavioral tests were developed to access OA-induced pain. However, follow-up in most studies usually only occurred for about 4 weeks. Since traumatic OA is a chronic disease which gradually develops after trauma, the pattern of pain might differ between early and late stages after the trauma. Purpose To observe the time-dependent development of hypersensitivity after traumatic OA and to determine the best timing and methods to investigate traumatic OA-induced pain. Methods Anterior cruciate ligament transection plus medial meniscectomy was used to induce traumatic OA in Sprague-Dawley rats. Traumatic OA-induced pain was evaluated using four different behavioral tests for 15 weeks. Results A significant difference in mechanical hypersensitivity developed throughout the observational period. It was worst in the first 3 weeks after the operation, then became less significant after 5 weeks but persisted. There were no differences in thermal hyperalgesia or motor coordination. Conclusion Traumatic OA induced mechanical hyperalgesia but did not cause thermal hyperalgesia or influence motor coordination. Furthermore, to investigate chronic pain induced by OA, the observational period should be at least 5 weeks after the intervention. These findings may help in further research and improve our understanding of traumatic OA-induced pain mechanisms.
Collapse
Affiliation(s)
- Hsiao-Chien Tsai
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Anesthesiology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ta-Liang Chen
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu-Pin Chen
- Department of Orthopedic Surgery, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ruei-Ming Chen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Comprehensive Cancer Center, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
48
|
Pharmacological validation of voluntary gait and mechanical sensitivity assays associated with inflammatory and neuropathic pain in mice. Neuropharmacology 2017; 130:18-29. [PMID: 29191755 DOI: 10.1016/j.neuropharm.2017.11.036] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/09/2017] [Accepted: 11/22/2017] [Indexed: 01/01/2023]
Abstract
The urgent need for more effective analgesic treatment options has prompted a re-evaluation of the behavioral tests used to assess pain in pre-clinical research, with an emphasis on inclusion of more voluntary, un-evoked behavioral assessments of pain. In order to validate voluntary gait analysis and a voluntary mechanical conflict-avoidance assay, we tested mouse models of neuropathy (spared nerve injury) and inflammation (complete Freund's adjuvant) alongside reflexive measures of mechanical and thermal hypersensitivity. To establish whether the observed changes in behavioral responses were pain-related, known analgesics (buprenorphine, gabapentin, carprofen) were also administered. Spared nerve injury persistently altered several gait indices, whereas complete Freund's adjuvant caused only transient changes. Furthermore, known analgesics could not reverse these gait changes, despite demonstrating their previously established efficacy in reflexive measures of mechanical and thermal hypersensitivity. In contrast, the mechanical conflict-avoidance assay demonstrated aversion in mice with neuropathy and inflammation-induced hypersensitivity, which could both be reversed by analgesics. We conclude that voluntary gait changes in rodent neuropathic and inflammatory pain models are not necessarily indicative of pain-related adaptations. On the other hand, mechanical conflict-avoidance represents a valid operant assay for quantifying pain-related behaviors in mice that can be reversed by known analgesics.
Collapse
|
49
|
King W, Bendele A, Marohl T, Woodell-May J. Human blood-based anti-inflammatory solution inhibits osteoarthritis progression in a meniscal-tear rat study. J Orthop Res 2017; 35:2260-2268. [PMID: 28139016 DOI: 10.1002/jor.23528] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 01/06/2017] [Indexed: 02/04/2023]
Abstract
Current osteoarthritis (OA) research searches for treatments that modify the course of disease progression. Autologous Protein Solution (APS) is derived from whole blood and is a unique autologous therapy that contains high concentrations of white blood cells, platelets, and concentrated plasma, providing cytokines that can target the underlying mechanisms of disease progression. The APS Kit is currently under investigation for clinical use in the USA (NCT02262364). The aim of this study was to determine if APS has disease-modifying properties in the well-characterized rat meniscal tear-induced model of OA. Thirty male athymic rats underwent surgery to induce OA by a complete tear of the medial meniscus of the right knee. Seven days later, rats were administered 50 μl intra-articular (IA) APS from a human donor or phosphate buffered saline (PBS) control. Rats were euthanized 3 weeks following treatment and knee joints were processed for histological analysis. Collagen and cartilage degeneration were decreased by APS treatment, resulting in a significantly improved total joint score in APS-treated rats compared to those treated with the PBS control. No significant variations in gait analysis, weight gain, osteophyte score, or synovitis score were observed between APS- and PBS-treated animals. There were no adverse effects of APS reported in the study. Treatment with a single IA injection of APS reduced the cartilage degeneration that characterizes the progression of OA. Further studies are necessary to determine if APS can modify OA disease progression in humans. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2260-2268, 2017.
Collapse
|
50
|
|