1
|
Wang D, Liu W, Venkatesan JK, Madry H, Cucchiarini M. Therapeutic Controlled Release Strategies for Human Osteoarthritis. Adv Healthc Mater 2024:e2402737. [PMID: 39506433 DOI: 10.1002/adhm.202402737] [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] [Received: 07/24/2024] [Revised: 10/15/2024] [Indexed: 11/08/2024]
Abstract
Osteoarthritis is a progressive, irreversible debilitating whole joint disease that affects millions of people worldwide. Despite the availability of various options (non-pharmacological and pharmacological treatments and therapy, orthobiologics, and surgical interventions), none of them can definitively cure osteoarthritis in patients. Strategies based on the controlled release of therapeutic compounds via biocompatible materials may provide powerful tools to enhance the spatiotemporal delivery, expression, and activities of the candidate agents as a means to durably manage the pathological progression of osteoarthritis in the affected joints upon convenient intra-articular (injectable) delivery while reducing their clearance, dissemination, or side effects. The goal of this review is to describe the current knowledge and advancements of controlled release to treat osteoarthritis, from basic principles to applications in vivo using therapeutic recombinant molecules and drugs and more innovatively gene sequences, providing a degree of confidence to manage the disease in patients in a close future.
Collapse
Affiliation(s)
- Dan Wang
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Wei Liu
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Kirrbergerstr. Bldg 37, D-66421, Homburg/Saar, Germany
| |
Collapse
|
2
|
Chang H, Liu L, Zhang Q, Xu G, Wang J, Chen P, Li C, Guo X, Yang Z, Zhang F. A comparative metabolomic analysis reveals the metabolic variations among cartilage of Kashin-Beck disease and osteoarthritis. Bone Joint Res 2024; 13:362-371. [PMID: 39013544 PMCID: PMC11251783 DOI: 10.1302/2046-3758.137.bjr-2023-0403.r1] [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: 07/18/2024] Open
Abstract
Aims The metabolic variations between the cartilage of osteoarthritis (OA) and Kashin-Beck disease (KBD) remain largely unknown. Our study aimed to address this by conducting a comparative analysis of the metabolic profiles present in the cartilage of KBD and OA. Methods Cartilage samples from patients with KBD (n = 10) and patients with OA (n = 10) were collected during total knee arthroplasty surgery. An untargeted metabolomics approach using liquid chromatography coupled with mass spectrometry (LC-MS) was conducted to investigate the metabolomics profiles of KBD and OA. LC-MS raw data files were converted into mzXML format and then processed by the XCMS, CAMERA, and metaX toolbox implemented with R software. The online Kyoto Encyclopedia of Genes and Genomes (KEGG) database was used to annotate the metabolites by matching the exact molecular mass data of samples with those from the database. Results A total of 807 ion features were identified for KBD and OA, including 577 positive (240 for upregulated and 337 for downregulated) and 230 negative (107 for upregulated and 123 for downregulated) ions. After annotation, LC-MS identified significant expressions of ten upregulated and eight downregulated second-level metabolites, and 183 upregulated and 162 downregulated first-level metabolites between KBD and OA. We identified differentially expressed second-level metabolites that are highly associated with cartilage damage, including dimethyl sulfoxide, uric acid, and betaine. These metabolites exist in sulphur metabolism, purine metabolism, and glycine, serine, and threonine metabolism. Conclusion This comprehensive comparative analysis of metabolism in OA and KBD cartilage provides new evidence of differences in the pathogenetic mechanisms underlying cartilage damage in these two conditions.
Collapse
Affiliation(s)
- Hong Chang
- Shaanxi Provincial Institute for Endemic Disease Control, Xi'an, China
| | - Li Liu
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Qingping Zhang
- Shaanxi Provincial Institute for Endemic Disease Control, Xi'an, China
| | - Gangyao Xu
- Shaanxi Provincial Institute for Endemic Disease Control, Xi'an, China
| | - Jianpeng Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Ping Chen
- Shaanxi Provincial Institute for Endemic Disease Control, Xi'an, China
| | - Cheng Li
- Shaanxi Provincial Institute for Endemic Disease Control, Xi'an, China
| | - Xianni Guo
- Shaanxi Provincial Institute for Endemic Disease Control, Xi'an, China
| | - Zhengjun Yang
- Shaanxi Provincial Institute for Endemic Disease Control, Xi'an, China
| | - Feng Zhang
- School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
3
|
Montero Furelos LA, De Castro Carrasco A, Cons Lamas S, Sanchez Sierra FB, Caeiro-Rey JR. Rapidly Progressive Osteoarthritis of the Hip: A Prospective Study. J Clin Med 2024; 13:2467. [PMID: 38730996 PMCID: PMC11084505 DOI: 10.3390/jcm13092467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Rapidly progressive osteoarthritis of the hip (RPOH) is a rare syndrome that involves the femoral head and acetabulum. Methods: We analyzed the incidence of RPOH in 2022. The inclusion criteria included a clinical history of pain for 1-6 months and a decrease in joint space of > 2 mm within one year or a decrease in joint space by 50% in that time accompanied by femoral and/or acetabular bone destruction. Exclusion: There are no other destructive arthropathies and no evolutionary radiological image sequence. Results: There were 15 patients, 16 hips, an incidence around 3.17%, a 1:1 laterality ratio, and 1 bilateral affected. The mean average age is 77.35 years. The male/female ratio is 1:2. The average BMI is 31.2. The time of the onset of the symptoms to the patient's diagnosis is 5 months. The functionally modified Harris scale (MHS) had an average score of 30 points. They had surgery hip arthroplasty with a cementless cup in all cases, a revision cup in one of them, and a double mobility cup in the other, with the stem cemented three times. There were no post-surgical complications. Functionally was achieved at 3 months. The average MHS is 70 at 12 months. The average MHS is 85. Conclusions: RPOH is an idiopathic entity characterized by great clinical involvement and rapid radiological evolution. It is most common in women around 77 years of age. The bone quality requires surgical alternatives to implants, and it has good functional recovery post-surgery.
Collapse
Affiliation(s)
- Luis Angel Montero Furelos
- Orthopedic Surgery and Traumatology Service, Hip Unit, Santiago de Compostela University Clinical Hospital, SERGAS, 15706 Santiago de Compostela, Spain; (A.D.C.C.); (S.C.L.); (F.B.S.S.); (J.R.C.-R.)
| | | | | | | | | |
Collapse
|
4
|
Cioroianu GO, Florescu A, Simionescu CE, Sas TN, Tarniţă DN, Rogoveanu OC. The therapeutic benefits of NSAIDs and physical therapy in knee osteoarthritis. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2024; 65:217-224. [PMID: 39020536 PMCID: PMC11384862 DOI: 10.47162/rjme.65.2.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
INTRODUCTION Osteoarthritis (OA) has been established as a progressive wear and tear disease of the synovial joints, which also involves a certain degree of inflammation. Considering there is no disease modifying medication available at the moment, the current guidelines focus on the symptomatic treatment of the affection. Our study aimed to evaluate the therapeutic advantages of the synergistic use of non-steroidal anti-inflammatory drugs (NSAIDs) and physical therapy in the treatment of knee osteoarthritis (KOA). PATIENTS, MATERIALS AND METHODS The study comprised 46 individuals who were diagnosed with KOA and were admitted to the Department of Physical Medicine and Rehabilitation at the Emergency Clinical County Hospital of Craiova, Romania, between January 2021 and April 2022. All the participants received the same combination of pharmacological (Diclofenac 150 mg∕day, no more than 10 days∕month as needed) and non-pharmacological treatment (a 24-week plan of physical therapy). RESULTS The patient group exhibited a statistically significant reduction in both the average Western Ontario and McMaster Universities Osteoarthritis (WOMAC) index (p=0.0142) and the average Visual Analog Scale (VAS) (p=0.0023). Additionally, there was a statistically significant increase in both the average Knee Outcome Survey-Activities of Daily Living (KOS-ADL) (p=0.0128) and the average Oxford Knee Score (OKS) (p=0.0023). The study found a significant positive correlation between higher VAS ratings and cholesterol levels (p=0.0092), but no significant correlation between VAS scores and triglyceride levels (p=0.0986). Patients were evaluated for a further 24 weeks beyond the conclusion of the research to see if surgical intervention was necessary during this time. CONCLUSIONS Our investigation tracked the WOMAC, VAS, KOS-ADL, and OKS measurements in a cohort of patients with KOA. The results demonstrate that the utilization of NSAIDs in conjunction with physical therapy effectively alleviates pain and enhances joint functionality.
Collapse
|
5
|
Kuszel L, Trzeciak T, Begier‐Krasinska B, Richter M, Li J, Czarny‐Ratajczak M. Sex-specific differences in telomere length of patients with primary knee osteoarthritis. J Cell Mol Med 2024; 28:e18107. [PMID: 38235989 PMCID: PMC10844687 DOI: 10.1111/jcmm.18107] [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: 07/31/2023] [Revised: 11/17/2023] [Accepted: 11/30/2023] [Indexed: 01/19/2024] Open
Abstract
Accelerated telomere shortening is associated with age-related diseases, including osteoarthritis (OA). We aimed to determine the relative telomere length (TL) in leukocytes and cartilage of patients with primary knee OA and to investigate factors that may affect TL in OA. Relative TL measurements were performed using qPCR in leukocytes of 612 individuals (310 patients with primary knee OA undergoing total knee arthroplasty (TKA) and 302 unaffected controls). We also analysed cartilage in 57 of the 310 OA patients, measuring relative TL in severely affected and less affected (control) cartilage collected from the same knee. Cartilage TLs were compared to leukocyte TLs in all 57 patients. A significant sex-by-disease-status interaction was found in regard to relative TL. Controlling for age, the average difference of leukocyte TL between female OA patients versus female controls was 0.217 units greater than that between male OA patients versus male controls (95% CI; [0.014, 0.421]). Relative TL comparison of severely and less affected cartilage samples from the same joint showed attrition of telomeres corresponding to disease severity (0.345 mean TL difference with 95% CI of [0.151, 0.539]) in the joint. We also noted that both severely and less affected cartilage had shorter telomeres than leukocytes collected from the same patient. Severe and moderate pain in OA patients was associated with shorter TL in leukocytes, but there was no association with depression or smoking in leukocytes and cartilage. Our study indicates that sex is an important factor in OA contributing to leukocyte and cartilage TL and that pain in OA shows an inverse association only with leukocyte TL.
Collapse
Affiliation(s)
- Lukasz Kuszel
- Department of Medical GeneticsPoznan University of Medical SciencesPoznanPoland
| | - Tomasz Trzeciak
- Department of Orthopedics and TraumatologyPoznan University of Medical SciencesPoznanPoland
| | - Beata Begier‐Krasinska
- Department of Hypertensiology, Angiology and Internal DiseasesPoznan University of Medical SciencesPoznanPoland
| | - Magdalena Richter
- Department of Orthopedics and TraumatologyPoznan University of Medical SciencesPoznanPoland
| | - Jian Li
- Department of Biostatistics & Data ScienceTulane School of Public Health and Tropical MedicineNew OrleansLouisianaUSA
| | - Malwina Czarny‐Ratajczak
- Department of Medicine, Center for AgingTulane University, School of MedicineNew OrleansLouisianaUSA
- Department of Medicine, Center for Biomedical Informatics and GenomicsTulane University, School of MedicineNew OrleansLouisianaUSA
| |
Collapse
|
6
|
Ruan Q, Wang C, Zhang Y, Sun J. Ruscogenin attenuates cartilage destruction in osteoarthritis through suppressing chondrocyte ferroptosis via Nrf2/SLC7A11/GPX4 signaling pathway. Chem Biol Interact 2024; 388:110835. [PMID: 38122922 DOI: 10.1016/j.cbi.2023.110835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/27/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Osteoarthritis (OA) is a common joint degenerative disease, and chondrocyte injury is the main pathological and physiological change. Ruscogenin (Rus), a bioactive compound isolated from Radix Ophiopogon japonicus, exhibits various pharmacological effects. The aim of this research was to test the role and mechanism of Rus on OA both in vivo and in vitro. Destabilized medial meniscus (DMM)-induced OA model was established in vivo and IL-1β-stimulated mouse chondrocytes was used to explore the role of Rus on OA in vitro. In vivo, Rus exhibited protective effects against DMM-induced OA model. Rus could inhibit MMP1 and MMP3 expression in OA mice. In vitro, IL-1β-induced inflammation and degradation of extracellular matrix were inhibited by Rus, as confirmed by the inhibition of PGE2, NO, MMP1, and MMP3 by Rus. Also, IL-1β-induced ferroptosis was suppressed by Rus, as confirmed by the inhibition of MDA, iron, and ROS, as well as the upregulation of GSH, GPX4, Ferritin, Nrf2, and SLC7A11 expression induced by Rus. Furthermore, the suppression of Rus on IL-1β-induced inflammation, MMPs production, and ferroptosis were reversed when Nrf2 was knockdown. In conclusion, Rus attenuated OA progression through inhibiting chondrocyte ferroptosis via Nrf2/SLC7A11/GPX4 signaling pathway.
Collapse
Affiliation(s)
- Qing Ruan
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Cuijie Wang
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Erdao District, 126 Sendai Street, Changchun, Jilin Province, 130033, China
| | - Yunfeng Zhang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Jiayang Sun
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China.
| |
Collapse
|
7
|
Arora D, Taneja Y, Sharma A, Dhingra A, Guarve K. Role of Apoptosis in the Pathogenesis of Osteoarthritis: An Explicative Review. Curr Rheumatol Rev 2024; 20:2-13. [PMID: 37670694 DOI: 10.2174/1573397119666230904150741] [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: 01/14/2023] [Revised: 05/29/2023] [Accepted: 07/20/2023] [Indexed: 09/07/2023]
Abstract
Apoptosis is a complex regulatory, active cell death process that plays a role in cell development, homeostasis, and ageing. Cancer, developmental defects, and degenerative diseases are all pathogenic disorders caused by apoptosis dysregulation. Osteoarthritis (OA) is by far the most frequently diagnosed joint disease in the aged, and it is characterized by the ongoing breakdown of articular cartilage, which causes severe disability. Multiple variables regulate the anabolic and catabolic pathways of the cartilage matrix, which either directly or indirectly contribute to cartilage degeneration in osteoarthritis. Articular cartilage is a highly specialized tissue made up of an extracellular matrix of cells that are tightly packed together. As a result, chondrocyte survival is crucial for the preservation of an optimal cartilage matrix, and chondrocyte characteristics and survival compromise may result in articular cartilage failure. Inflammatory cytokines can either promote or inhibit apoptosis, the process of programmed cell death. Pro-apoptotic cytokines like TNF-α can induce cell death, while anti-apoptotic cytokines like IL-4 and IL-10 protect against apoptosis. The balance between these cytokines plays a critical role in determining cell fate and has implications for tissue damage and disease progression. Similarly, they contribute to the progression of OA by disrupting the metabolic balance in joint tissues by promoting catabolic and anabolic pathways. Their impact on cell joints, as well as the impacts of cell signalling pathways on cytokines and inflammatory substances, determines their function in osteoarthritis development. Apoptosis is evident in osteoarthritic cartilage; however, determining the relative role of chondrocyte apoptosis in the aetiology of OA is difficult, and the rate of apoptotic chondrocytes in osteoarthritic cartilage is inconsistent. The current study summarises the role of apoptosis in the development of osteoarthritis, the mediators, and signalling pathways that trigger the cascade of events, and the other inflammatory features involved.
Collapse
Affiliation(s)
- Deepshi Arora
- Department of Pharmacy, Guru Gobind Singh College of Pharmacy, Yamuna Nagar, Haryana, 135001, India
| | - Yugam Taneja
- Zeon Lifesciences, Paonta Sahib, Himachal Pradesh, 173025, India
| | - Anjali Sharma
- Department of Pharmacy, Guru Gobind Singh College of Pharmacy, Yamuna Nagar, Haryana, 135001, India
| | - Ashwani Dhingra
- Department of Pharmacy, Guru Gobind Singh College of Pharmacy, Yamuna Nagar, Haryana, 135001, India
| | - Kumar Guarve
- Department of Pharmacy, Guru Gobind Singh College of Pharmacy, Yamuna Nagar, Haryana, 135001, India
| |
Collapse
|
8
|
Wasser JG, Hendershot BD, Acasio JC, Dodd LD, Krupenevich RL, Pruziner AL, Miller RH, Goldman SM, Valerio MS, Senchak LT, Murphey MD, Heltzel DA, Fazio MG, Dearth CL, Hager NA. Exploring relationships among multi-disciplinary assessments for knee joint health in service members with traumatic unilateral lower limb loss: a two-year longitudinal investigation. Sci Rep 2023; 13:21177. [PMID: 38040780 PMCID: PMC10692131 DOI: 10.1038/s41598-023-48662-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 11/29/2023] [Indexed: 12/03/2023] Open
Abstract
Motivated by the complex and multifactorial etiologies of osteoarthritis, here we use a comprehensive approach evaluating knee joint health after unilateral lower limb loss. Thirty-eight male Service members with traumatic, unilateral lower limb loss (mean age = 38 yr) participated in a prospective, two-year longitudinal study comprehensively evaluating contralateral knee joint health (i.e., clinical imaging, gait biomechanics, physiological biomarkers, and patient-reported outcomes); seventeen subsequently returned for a two-year follow-up visit. For this subset with baseline and follow-up data, outcomes were compared between timepoints, and associations evaluated between values at baseline with two-year changes in tri-compartmental joint space. Upon follow-up, knee joint health worsened, particularly among seven Service members who presented at baseline with no joint degeneration (KL = 0) but returned with evidence of degeneration (KL ≥ 1). Joint space narrowing was associated with greater patellar tilt (r[12] = 0.71, p = 0.01), external knee adduction moment (r[13] = 0.64, p = 0.02), knee adduction moment impulse (r[13] = 0.61, p = 0.03), and CTX-1 concentration (r[11] = 0.83, p = 0.001), as well as lesser KOOSSport and VR-36General Health (r[16] = - 0.69, p = 0.01 and r[16] = - 0.69, p = 0.01, respectively). This longitudinal, multi-disciplinary investigation highlights the importance of a comprehensive approach to evaluate the fast-progressing onset of knee osteoarthritis, particularly among relatively young Service members with lower limb loss.
Collapse
Affiliation(s)
- Joseph G Wasser
- Department of Rehabilitation, Walter Reed National Military Medical Center, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
- Department of Physical Medicine and Rehabilitation, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Brad D Hendershot
- Department of Rehabilitation, Walter Reed National Military Medical Center, Bethesda, MD, USA.
- Department of Physical Medicine and Rehabilitation, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
- Research and Surveillance Section, Extremity Trauma and Amputation Center of Excellence, Research and Engineering Directorate, Defense Health Agency, Falls Church, VA, USA.
| | - Julian C Acasio
- Department of Rehabilitation, Walter Reed National Military Medical Center, Bethesda, MD, USA
- Research and Surveillance Section, Extremity Trauma and Amputation Center of Excellence, Research and Engineering Directorate, Defense Health Agency, Falls Church, VA, USA
| | - Lauren D Dodd
- Department of Rehabilitation, Walter Reed National Military Medical Center, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA
| | - Rebecca L Krupenevich
- Department of Rehabilitation, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Alison L Pruziner
- Department of Rehabilitation, Walter Reed National Military Medical Center, Bethesda, MD, USA
- Department of Physical Medicine and Rehabilitation, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Research and Surveillance Section, Extremity Trauma and Amputation Center of Excellence, Research and Engineering Directorate, Defense Health Agency, Falls Church, VA, USA
| | - Ross H Miller
- Department of Kinesiology, University of Maryland, College Park, MD, USA
| | - Stephen M Goldman
- Research and Surveillance Section, Extremity Trauma and Amputation Center of Excellence, Research and Engineering Directorate, Defense Health Agency, Falls Church, VA, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Michael S Valerio
- Research and Surveillance Section, Extremity Trauma and Amputation Center of Excellence, Research and Engineering Directorate, Defense Health Agency, Falls Church, VA, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Lien T Senchak
- Department of Diagnostic Radiology, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Mark D Murphey
- Department of Diagnostic Radiology, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - David A Heltzel
- Department of Diagnostic Radiology, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Michael G Fazio
- Department of Diagnostic Radiology, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Christopher L Dearth
- Department of Rehabilitation, Walter Reed National Military Medical Center, Bethesda, MD, USA
- Research and Surveillance Section, Extremity Trauma and Amputation Center of Excellence, Research and Engineering Directorate, Defense Health Agency, Falls Church, VA, USA
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nelson A Hager
- Department of Physical Medicine and Rehabilitation, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
9
|
Wen ZQ, Lin J, Xie WQ, Shan YH, Zhen GH, Li YS. Insights into the underlying pathogenesis and therapeutic potential of endoplasmic reticulum stress in degenerative musculoskeletal diseases. Mil Med Res 2023; 10:54. [PMID: 37941072 PMCID: PMC10634069 DOI: 10.1186/s40779-023-00485-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Degenerative musculoskeletal diseases are structural and functional failures of the musculoskeletal system, including osteoarthritis, osteoporosis, intervertebral disc degeneration (IVDD), and sarcopenia. As the global population ages, degenerative musculoskeletal diseases are becoming more prevalent. However, the pathogenesis of degenerative musculoskeletal diseases is not fully understood. Previous studies have revealed that endoplasmic reticulum (ER) stress is a stress response that occurs when impairment of the protein folding capacity of the ER leads to the accumulation of misfolded or unfolded proteins in the ER, contributing to degenerative musculoskeletal diseases. By affecting cartilage degeneration, synovitis, meniscal lesion, subchondral bone remodeling of osteoarthritis, bone remodeling and angiogenesis of osteoporosis, nucleus pulposus degeneration, annulus fibrosus rupture, cartilaginous endplate degeneration of IVDD, and sarcopenia, ER stress is involved in the pathogenesis of degenerative musculoskeletal diseases. Preclinical studies have found that regulation of ER stress can delay the progression of multiple degenerative musculoskeletal diseases. These pilot studies provide foundations for further evaluation of the feasibility, efficacy, and safety of ER stress modulators in the treatment of musculoskeletal degenerative diseases in clinical trials. In this review, we have integrated up-to-date research findings of ER stress into the pathogenesis of degenerative musculoskeletal diseases. In a future perspective, we have also discussed possible directions of ER stress in the investigation of degenerative musculoskeletal disease, potential therapeutic strategies for degenerative musculoskeletal diseases using ER stress modulators, as well as underlying challenges and obstacles in bench-to-beside research.
Collapse
Affiliation(s)
- Ze-Qin Wen
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Jun Lin
- Department of Orthopaedics, Suzhou Dushu Lake Hospital, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, 215001, China
| | - Wen-Qing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yun-Han Shan
- Xiangya School of Medicine, Central South University, Changsha, 410013, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ge-Hua Zhen
- Department of Orthopaedic Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | - Yu-Sheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
10
|
Zhang Y, Niu Y, Peng Y, Pan X, Wang F. COL3A1, COL5A1 and COL6A2 serve as potential molecular biomarkers for osteoarthritis based on weighted gene co‑expression network analysis bioinformatics analysis. Exp Ther Med 2023; 26:540. [PMID: 37869636 PMCID: PMC10587888 DOI: 10.3892/etm.2023.12239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/13/2023] [Indexed: 10/24/2023] Open
Abstract
Osteoarthritis (OA) is a non-inflammatory degenerative joint disease, characterized by joint pain and stiffness. The prevalence of OA increases with age. However, the relationship between biomarkers [collagen type III α1 (COL3A1), COL5A1, COL6A2, COL12A1] and OA remains unclear. The OA subchondral bone dataset GSE51588 was downloaded from the GEO database, and the differentially expressed genes (DEGs) were screened. Weighted gene co-expression network analysis was performed, and a protein-protein interaction network was constructed and further analyzed using Cytoscape and STRING. Functional enrichment analysis was performed using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and then Gene Set Enrichment Analysis (GSEA) was used to formulate the molecular functions and pathways based on the results of GO and KEGG analyses. Comparative Toxicogenomics Database and TargetScan were used to identify the hub-gene-related diseases and the microRNAs that regulated the central hub genes. Immunohistochemical staining was performed to confirm the expression of related proteins in OA and non-OA tissue samples. A total of 1,679 DEGs were identified. GO analysis showed that the DEGs were primarily enriched in the process of 'immune system', 'extracellular region', 'secretory granule', 'collagen-containing extracellular matrix', 'ECM-receptor, glycosaminoglycan binding' and 'systemic lupus erythematosus'. The results of GSEA were similar to those of GO and KEGG enrichment terms for DEGs. A total of 25 important modules were generated, and two core gene clusters and seven core genes were obtained (COL6A2, COL5A2, COL12A1, COL5A1, COL6A1, LUM and COL3A1). Core genes were expressed differentially between OA subchondral bone and normal tissue samples. The expression levels of COL3A1, COL5A1 and COL6A2 in OA subchondral bone tissue were higher compared with those in normal tissues, but COL12A1 expression was not significantly increased; all stained markers were highly expressed in surrounding tissues of immunohistochemical staining. In conclusion, COL3A1, COL5A1 and COL6A2 may be potential molecular biomarkers for OA.
Collapse
Affiliation(s)
- Yufeng Zhang
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Yingzhen Niu
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Yonggang Peng
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Xueyang Pan
- Department of Tactical Medical Service, Special Medical Service Teaching and Research Section, Army Medical University Non-Commissioned Officer School, Shijiazhuang, Hebei 050051, P.R. China
| | - Fei Wang
- Department of Orthopedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
11
|
Kehayova YS, Wilkinson JM, Rice SJ, Loughlin J. Mediation of the Same Epigenetic and Transcriptional Effect by Independent Osteoarthritis Risk-Conferring Alleles on a Shared Target Gene, COLGALT2. Arthritis Rheumatol 2023; 75:910-922. [PMID: 36538011 PMCID: PMC10952352 DOI: 10.1002/art.42427] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Over 100 DNA variants have been associated with osteoarthritis (OA), including rs1046934, located within a linkage disequilibrium block encompassing part of COLGALT2 and TSEN15. The present study was undertaken to determine the target gene(s) and the mechanism of action of the OA locus using human fetal cartilage, cartilage from OA and femoral neck fracture arthroplasty patients, and a chondrocyte cell model. METHODS Genotyping and methylation array data of DNA from human OA cartilage samples (n = 87) were used to determine whether the rs1046934 genotype is associated with differential DNA methylation at proximal CpGs. Results were replicated in DNA from human arthroplasty (n = 132) and fetal (n = 77) cartilage samples using pyrosequencing. Allelic expression imbalance (AEI) measured the effects of genotype on COLGALT2 and TSEN15 expression. Reporter gene assays and epigenetic editing determined the functional role of regions harboring differentially methylated CpGs. In silico analyses complemented these experiments. RESULTS Three differentially methylated CpGs residing within regulatory regions were detected in the human OA cartilage array data, and 2 of these were replicated in human arthroplasty and fetal cartilage. AEI was detected for COLGALT2 and TSEN15, with associations between expression and methylation for COLGALT2. Reporter gene assays confirmed that the CpGs are in chondrocyte enhancers, with epigenetic editing results directly linking methylation with COLGALT2 expression. CONCLUSION COLGALT2 is a target of this OA locus. We previously characterized another OA locus, marked by rs11583641, that independently targets COLGALT2. The genotype of rs1046934, like rs11583641, mediates its effect by modulating expression of COLGALT2 via methylation changes to CpGs located in enhancers. Although the single-nucleotide polymorphisms, CpGs, and enhancers are distinct between the 2 independent OA risk loci, their effect on COLGALT2 is the same. COLGALT2 is the target of independent OA risk loci sharing a common mechanism of action.
Collapse
Affiliation(s)
| | - J. Mark Wilkinson
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Sarah J. Rice
- Biosciences Institute, Newcastle UniversityNewcastle upon TyneUK
| | - John Loughlin
- Biosciences Institute, Newcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
12
|
Liu Z, Liu H, Li D, Ma L, Lu T, Sun H, Zhang Y, Yang H. Comprehensive analysis of m6A RNA methylation modification patterns and the immune microenvironment in osteoarthritis. Front Immunol 2023; 14:1128459. [PMID: 37006311 PMCID: PMC10062708 DOI: 10.3389/fimmu.2023.1128459] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
Abstract
BackgroundOsteoarthritis (OA) is the most common joint degenerative disease, and so far, there is no effective therapy to prevent or delay its development. Considerable attention is now being given to the impact of m6A RNA methylation modification on the disease immune regulation. However, much remains unknown about the function of m6A modification in OA.MethodsA total of 63 OA and 59 healthy samples were applied to comprehensively examine the m6A regulators mediated RNA methylation modification pattern in OA, and evaluate the impacts of distinct patterns on the characteristics of OA immune microenvironment, including immune infiltration cells, immune responses and human leukocyte antigen (HLAs) genes expression. In addition, we screened out the m6A phenotype-related genes and further explored their potential biological functions. At last, we verified the expression of key m6A regulators and their associations with immune cells, in vitro.ResultsMost of m6A regulators was differentially expressed in OA samples compared to the normal tissues. Based on six hub-m6A regulators identified as abnormally expressed in OA samples, we developed a classifier to distinguish OA patients from healthy individuals. We noted that immune characteristics of OA were correlated with m6A regulators. For instance, YTHDF2 had a strongest significantly positive correlation with regulatory T cells (Tregs) and IGFBP2 was strongest negatively associated with dendritic cells (DCs), which were confirmed by the immunohistochemistry (IHC) staining. Two distinct m6A modification patterns were determined: pattern B had higher infiltrating immunocytes and more active immune responses than pattern A, and two patterns differed in the expression of HLA genes. We also identified 1,592 m6A phenotype-related genes that could mediate the OA synovitis and cartilage degradation by the PI3K-Akt signaling pathway. Quantitative real-time polymerase chain reaction (qRT-PCR) results indicated that IGFBP2 was significantly overexpressed, while YTHDF2 mRNA expression was decreased in OA samples, which was consistent with our findings.ConclusionOur research proves the essential impact of m6A RNA methylation modification on the OA immune microenvironment, and helps to explain the regulatory mechanism behind it, which may open up a new direction for more precise immunotherapy of osteoarthritis.
Collapse
Affiliation(s)
- Zhixin Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Heng Liu
- NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Deqiang Li
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Liang Ma
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tongxin Lu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hao Sun
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yuankai Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- *Correspondence: Yuankai Zhang, ; Hui Yang,
| | - Hui Yang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- *Correspondence: Yuankai Zhang, ; Hui Yang,
| |
Collapse
|
13
|
Lagneau N, Tournier P, Nativel F, Maugars Y, Guicheux J, Le Visage C, Delplace V. Harnessing cell-material interactions to control stem cell secretion for osteoarthritis treatment. Biomaterials 2023; 296:122091. [PMID: 36947892 DOI: 10.1016/j.biomaterials.2023.122091] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 03/16/2023]
Abstract
Osteoarthritis (OA) is the most common debilitating joint disease, yet there is no curative treatment for OA to date. Delivering mesenchymal stromal cells (MSCs) as therapeutic cells to mitigate the inflammatory symptoms associated with OA is attracting increasing attention. In principle, MSCs could respond to the pro-inflammatory microenvironment of an OA joint by the secretion of anti-inflammatory, anti-apoptotic, immunomodulatory and pro-regenerative factors, therefore limiting pain, as well as the disease development. However, the microenvironment of MSCs is known to greatly affect their survival and bioactivity, and using tailored biomaterial scaffolds could be key to the success of intra-articular MSC-based therapies. The aim of this review is to identify and discuss essential characteristics of biomaterial scaffolds to best promote MSC secretory functions in the context of OA. First, a brief introduction to the OA physiopathology is provided, followed by an overview of the MSC secretory functions, as well as the current limitations of MSC-based therapy. Then, we review the current knowledge on the effects of cell-material interactions on MSC secretion. These considerations allow us to define rational guidelines for next-generation biomaterial design to improve the MSC-based therapy of OA.
Collapse
Affiliation(s)
- Nathan Lagneau
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| | - Pierre Tournier
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| | - Fabien Nativel
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France; Nantes Université, UFR Sciences Biologiques et Pharmaceutiques, Nantes, F-44035, France
| | - Yves Maugars
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France.
| | - Catherine Le Visage
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| | - Vianney Delplace
- Nantes Université, Oniris, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000, France
| |
Collapse
|
14
|
Zhang ZJ, Hou YK, Chen MW, Yu XZ, Chen SY, Yue YR, Guo XT, Chen JX, Zhou Q. A pH-responsive metal-organic framework for the co-delivery of HIF-2α siRNA and curcumin for enhanced therapy of osteoarthritis. J Nanobiotechnology 2023; 21:18. [PMID: 36650517 PMCID: PMC9847079 DOI: 10.1186/s12951-022-01758-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
The occurrence of osteoarthritis (OA) is highly correlated with the reduction of joint lubrication performance, in which persistent excessive inflammation and irreversible destruction of cartilage dominate the mechanism. The inadequate response to monotherapy methods, suboptimal efficacy caused by undesirable bioavailability, short retention, and lack of stimulus-responsiveness, are few unresolved issues. Herein, we report a pH-responsive metal-organic framework (MOF), namely, MIL-101-NH2, for the co-delivery of anti-inflammatory drug curcumin (CCM) and small interfering RNA (siRNA) for hypoxia inducible factor (HIF-2α). CCM and siRNA were loaded via encapsulation and surface coordination ability of MIL-101-NH2. Our vitro tests showed that MIL-101-NH2 protected siRNA from nuclease degradation by lysosomal escape. The pH-responsive MIL-101-NH2 gradually collapsed in an acidic OA microenvironment to release the CCM payloads to down-regulate the level of pro-inflammatory cytokines, and to release the siRNA payloads to cleave the target HIF-2α mRNA for gene-silencing therapy, ultimately exhibiting the synergetic therapeutic efficacy by silencing HIF-2α genes accompanied by inhibiting the inflammation response and cartilage degeneration of OA. The hybrid material reported herein exhibited promising potential performance for OA therapy as supported by both in vitro and in vivo studies and may offer an efficacious therapeutic strategy for OA utilizing MOFs as host materials.
Collapse
Affiliation(s)
- Zi-Jian Zhang
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Ying-Ke Hou
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Ming-Wa Chen
- grid.284723.80000 0000 8877 7471NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515 People’s Republic of China
| | - Xue-Zhao Yu
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Si-Yu Chen
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Ya-Ru Yue
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Xiong-Tian Guo
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| | - Jin-Xiang Chen
- grid.284723.80000 0000 8877 7471NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515 People’s Republic of China
| | - Quan Zhou
- grid.284723.80000 0000 8877 7471Department of Medical Imaging, Third Affiliated Hospital of Southern Medical University (Academy of Orthopedics Guangdong Province), Southern Medical University, Guangzhou, 510630 Guangdong People’s Republic of China
| |
Collapse
|
15
|
Guo J, Huang X, Dou L, Yan M, Shen T, Tang W, Li J. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther 2022; 7:391. [PMID: 36522308 PMCID: PMC9755275 DOI: 10.1038/s41392-022-01251-0] [Citation(s) in RCA: 305] [Impact Index Per Article: 152.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 12/23/2022] Open
Abstract
Aging is a gradual and irreversible pathophysiological process. It presents with declines in tissue and cell functions and significant increases in the risks of various aging-related diseases, including neurodegenerative diseases, cardiovascular diseases, metabolic diseases, musculoskeletal diseases, and immune system diseases. Although the development of modern medicine has promoted human health and greatly extended life expectancy, with the aging of society, a variety of chronic diseases have gradually become the most important causes of disability and death in elderly individuals. Current research on aging focuses on elucidating how various endogenous and exogenous stresses (such as genomic instability, telomere dysfunction, epigenetic alterations, loss of proteostasis, compromise of autophagy, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, deregulated nutrient sensing) participate in the regulation of aging. Furthermore, thorough research on the pathogenesis of aging to identify interventions that promote health and longevity (such as caloric restriction, microbiota transplantation, and nutritional intervention) and clinical treatment methods for aging-related diseases (depletion of senescent cells, stem cell therapy, antioxidative and anti-inflammatory treatments, and hormone replacement therapy) could decrease the incidence and development of aging-related diseases and in turn promote healthy aging and longevity.
Collapse
Affiliation(s)
- Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Lin Dou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Mingjing Yan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| |
Collapse
|
16
|
Shirogane Y, Homma Y, Yanagisawa N, Higano M, Hirasawa Y, Nakamura S, Baba T, Kaneko K, Taneda H, Ishijima M. Relationship between labral length and symptoms in patients with acetabular dysplasia before rotational acetabular osteotomy. J Hip Preserv Surg 2022; 9:240-251. [PMID: 36908550 PMCID: PMC9993447 DOI: 10.1093/jhps/hnac045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/03/2022] [Accepted: 09/06/2022] [Indexed: 03/14/2023] Open
Abstract
The aim of this study was to investigate the relationship between acetabular labral length and symptoms in patients with acetabular dysplasia. In a retrospective medical record review, 218 patients with acetabular dysplasia who had undergone rotational acetabular osteotomy were identified. After implementing the inclusion and exclusion criteria, 53 patients were analyzed for preoperative symptoms measured by the Japanese Orthopaedic Association Hip Disease Evaluation Questionnaire (JHEQ), acetabular bone morphology parameters by anteroposterior pelvic radiographs and labral parameters by radial magnetic resonance imaging. Spearman's correlation coefficients were calculated among JHEQ scores, bone morphologic parameters and labral parameters. Multiple linear regression models to determine the predictive variables of JHEQ score and labral length were obtained. There was no correlation between bone morphologic parameters and JHEQ scores. Labral length measured anteriorly correlated with JHEQ pain {r [95% confidence interval (CI)] = -0.335 (-0.555, -0.071), P = 0.014}, movement subscale [r (95% CI) = -0.398 (-0.603, -0.143), P = 0.003], mental subscale [r (95% CI) = -0.436 (-0.632, -0.188), P = 0.001] and total JHEQ score [r (95% CI) = -0.451 (-0.642, -0.204), P = 0.001]. The multiple linear regression results showed that anterior labral length was independently associated with JHEQ subscales in some models. Meanwhile, age, acetabular head index and total JHEQ score were independently associated with anterior labral length in all models. Labral length, notably in anterosuperior area, in patients with symptomatic acetabular dysplasia was related to patient's symptom. Labral length may be an important objective image finding that can be used to assess the severity of cumulative hip instability.
Collapse
Affiliation(s)
- Yuichi Shirogane
- Department of Orthopaedic Surgery, Nishitokyo Chuo General Hospital, 2-4-19 Shibakubocho, Nishitokyo-shi, Tokyo 188-0014,Japan.,Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 133-8421, Japan.,Department of Orthopaedic, Faculty of Medicine, Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo 133-8431, Japan
| | - Yasuhiro Homma
- Department of Orthopaedic Surgery, Nishitokyo Chuo General Hospital, 2-4-19 Shibakubocho, Nishitokyo-shi, Tokyo 188-0014,Japan.,Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 133-8421, Japan.,Department of Orthopaedic, Faculty of Medicine, Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo 133-8431, Japan
| | - Naotake Yanagisawa
- Clinical Research and Trial Center, Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo 133-8431, Japan
| | - Masanori Higano
- Department of Orthopaedic Surgery, Nishitokyo Chuo General Hospital, 2-4-19 Shibakubocho, Nishitokyo-shi, Tokyo 188-0014,Japan
| | - Yoichiro Hirasawa
- Department of Orthopaedic Surgery, Nishitokyo Chuo General Hospital, 2-4-19 Shibakubocho, Nishitokyo-shi, Tokyo 188-0014,Japan
| | - Shigeru Nakamura
- Department of Orthopaedic Surgery, Nishitokyo Chuo General Hospital, 2-4-19 Shibakubocho, Nishitokyo-shi, Tokyo 188-0014,Japan
| | - Tomonori Baba
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 133-8421, Japan.,Department of Orthopaedic, Faculty of Medicine, Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo 133-8431, Japan
| | - Kazuo Kaneko
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 133-8421, Japan.,Department of Orthopaedic, Faculty of Medicine, Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo 133-8431, Japan
| | - Hitoshi Taneda
- Department of Orthopaedic Surgery, Nishitokyo Chuo General Hospital, 2-4-19 Shibakubocho, Nishitokyo-shi, Tokyo 188-0014,Japan
| | - Muneaki Ishijima
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 133-8421, Japan.,Department of Orthopaedic, Faculty of Medicine, Juntendo University, 3-1-3 Hongo, Bunkyo-ku, Tokyo 133-8431, Japan
| |
Collapse
|
17
|
Liu L, Luo P, Yang M, Wang J, Hou W, Xu P. The role of oxidative stress in the development of knee osteoarthritis: A comprehensive research review. Front Mol Biosci 2022; 9:1001212. [PMID: 36203877 PMCID: PMC9532006 DOI: 10.3389/fmolb.2022.1001212] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Knee osteoarthritis (KOA) is one of the most common degenerative diseases, and its core feature is the degeneration and damage of articular cartilage. The cartilage degeneration of KOA is due to the destruction of dynamic balance caused by the activation of chondrocytes by various factors, with oxidative stress playing an important role in the pathogenesis of KOA. The overproduction of reactive oxygen species (ROS) is a result of oxidative stress, which is caused by a redox process that goes awry in the inherent antioxidant defence system of the human body. Superoxide dismutase (SOD) inside and outside chondrocytes plays a key role in regulating ROS in cartilage. Additionally, synovitis is a key factor in the development of KOA. In an inflammatory environment, hypoxia in synovial cells leads to mitochondrial damage, which leads to an increase in ROS levels, which further aggravates synovitis. In addition, oxidative stress significantly accelerates the telomere shortening and ageing of chondrocytes, while ageing promotes the development of KOA, damages the regulation of redox of mitochondria in cartilage, and stimulates ROS production to further aggravate KOA. At present, there are many drugs to regulate the level of ROS, but these drugs still need to be developed and verified in animal models of KOA. We discuss mainly how oxidative stress plays a part in the development of KOA. Although the current research has achieved some results, more research is needed.
Collapse
|
18
|
Ruiz-Fernández C, González-Rodríguez M, Abella V, Francisco V, Cordero-Barreal A, Ait Eldjoudi D, Farrag Y, Pino J, Conde-Aranda J, González-Gay MÁ, Mera A, Mobasheri A, García-Caballero L, Gándara-Cortés M, Lago F, Scotece M, Gualillo O. WISP-2 modulates the induction of inflammatory mediators and cartilage catabolism in chondrocytes. J Transl Med 2022; 102:989-999. [PMID: 36775427 DOI: 10.1038/s41374-022-00793-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 04/12/2022] [Accepted: 04/12/2022] [Indexed: 11/08/2022] Open
Abstract
Wnt-1 inducible signaling pathway protein 2 (WISP-2/CCN5) is a recently identified adipokine that has been described as an important mediator of canonical Wnt activation in adipogenic precursor cells. In osteoarthritis (OA), the most common form of arthritis, chondrocytes exhibit aberrant and increased production of pro-inflammatory mediators and matrix degrading enzymes such as IL-1β and MMP-13. Although recent evidence suggests a role for Wnt signaling in OA physiopathology, little is known about the involvement of WISP-2 in cartilage degradation. In the present study, we determined the expression of WISP-2 in healthy and OA human chondrocytes. WISP-2 expression is modulated along chondrocyte differentiation and downregulated at the onset of hypertrophy by inflammatory mediators. We also investigated the effect of WISP-2 on cartilage catabolism and performed WISP-2 loss-of-function experiments using RNA interference technology in human T/C-28a2 immortalized chondrocytes. We demonstrated that recombinant human WISP-2 protein reduced IL-1β-mediated chondrocyte catabolism, that IL-1β and WNT/b-catenin signaling pathways are involved in rhWISP-2 protein and IL-1β effects in human chondrocytes, and that WISP-2 has a regulatory role in attenuating the catabolic effects of IL-1β in chondrocytes. Gene silencing of WISP-2 increased the induction of the catabolic markers MMP-13 and ADAMTS-5 and the inflammatory mediators IL-6 and IL-8 triggered by IL-1β in human primary OA chondrocytes in a Wnt/β-catenin dependent manner. In conclusion, here we have shown for the first time that WISP-2 may have relevant roles in modulating the turnover of extracellular matrix in the cartilage and that its downregulation may detrimentally alter the inflammatory environment in OA cartilage. We also proved the participation of Wnt/β-catenin signaling pathway in these processes. Thus, targeting WISP-2 might represent a potential therapeutical approach for degenerative and/or inflammatory diseases of musculoskeletal system, such as osteoarthritis.
Collapse
Affiliation(s)
- Clara Ruiz-Fernández
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
- International PhD School of the University of Santiago de Compostela (EDIUS), Doctoral Programme in Medicine Clinical Research, Santiago de Compostela, Spain
| | - María González-Rodríguez
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
- International PhD School of the University of Santiago de Compostela (EDIUS), Doctoral Programme in Drug Research and Development, Santiago de Compostela, Spain
| | - Vanessa Abella
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Vera Francisco
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Alfonso Cordero-Barreal
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Djedjiga Ait Eldjoudi
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Yousof Farrag
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Jesús Pino
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Javier Conde-Aranda
- Molecular and Cellular Gastroenterology Group, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Miguel Ángel González-Gay
- Hospital Universitario Marqués de Valdecilla, Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, IDIVAL, University of Cantabria, Avenida de Valdecilla s/n, Santander, Cantabria, Spain
| | - Antonio Mera
- SERGAS, Santiago University Clinical Hospital, Division of Rheumatology, Santiago de Compostela, Spain
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics, and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- University Medical Center Utrecht, Departments of Orthopedics, Rheumatology and Clinical Immunology, Utrecht, The Netherlands
- Department of Joint Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lucía García-Caballero
- Department of Morphological Sciences. School of Medicine and Dentistry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Marina Gándara-Cortés
- Department of Morphological Sciences. School of Medicine and Dentistry, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Francisca Lago
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Molecular and Cellular Cardiology Lab, Research Laboratory 7, Santiago University Clinical Hospital, Santiago de Compostela, Spain
| | - Morena Scotece
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain.
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, IDIS (Instituto de Investigación Sanitaria de Santiago), Santiago University Clinical Hospital, Santiago de Compostela, Spain.
| |
Collapse
|
19
|
Godivier J, Lawrence EA, Wang M, Hammond CL, Nowlan NC. Growth orientations, rather than heterogeneous growth rates, dominate jaw joint morphogenesis in the larval zebrafish. J Anat 2022; 241:358-371. [PMID: 35510779 PMCID: PMC9296026 DOI: 10.1111/joa.13680] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022] Open
Abstract
In early limb embryogenesis, synovial joints acquire specific shapes which determine joint motion and function. The process by which the opposing cartilaginous joint surfaces are moulded into reciprocal and interlocking shapes, called joint morphogenesis, is one of the least understood aspects of joint formation and the cell-level dynamics underlying it are yet to be unravelled. In this research, we quantified key cellular dynamics involved in growth and morphogenesis of the zebrafish jaw joint and synthesised them in a predictive computational simulation of joint development. Cells in larval zebrafish jaw joints labelled with cartilage markers were tracked over a 48-h time window using confocal imaging. Changes in distance and angle between adjacent cell centroids resulting from cell rearrangement, volume expansion and extracellular matrix (ECM) deposition were measured and used to calculate the rate and direction of local tissue deformations. We observed spatially and temporally heterogeneous growth patterns with marked anisotropy over the developmental period assessed. There was notably elevated growth at the level of the retroarticular process of the Meckel's cartilage, a feature known to undergo pronounced shape changes during zebrafish development. Analysis of cell dynamics indicated a dominant role for cell volume expansion in growth, with minor influences from ECM volume increases and cell intercalation. Cell proliferation in the joint was minimal over the timeframe of interest. Synthesising the dynamic cell data into a finite element model of jaw joint development resulted in accurate shape predictions. Our biofidelic computational simulation demonstrated that zebrafish jaw joint growth can be reasonably approximated based on cell positional information over time, where cell positional information derives mainly from cell orientation and cell volume expansion. By modifying the input parameters of the simulation, we were able to assess the relative contributions of heterogeneous growth rates and of growth orientation. The use of uniform rather than heterogeneous growth rates only minorly impacted the shape predictions, whereas isotropic growth fields resulted in altered shape predictions. The simulation results suggest that growth anisotropy is the dominant influence on joint growth and morphogenesis. This study addresses the gap of the cellular processes underlying joint morphogenesis, with implications for understanding the aetiology of developmental joint disorders such as developmental dysplasia of the hip and arthrogryposis.
Collapse
Affiliation(s)
| | | | | | | | - Niamh C. Nowlan
- Imperial College LondonLondonUnited Kingdom,University College DublinDublinIreland
| |
Collapse
|
20
|
Pei W, Deng J, Wang P, Wang X, Zheng L, Zhang Y, Huang C. Sustainable lignin and lignin-derived compounds as potential therapeutic agents for degenerative orthopaedic diseases: A systemic review. Int J Biol Macromol 2022; 212:547-560. [PMID: 35643155 DOI: 10.1016/j.ijbiomac.2022.05.152] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/13/2022] [Accepted: 05/22/2022] [Indexed: 12/12/2022]
Abstract
Lignin, the most abundant natural and sustainable phenolic compound in biomass, has exhibited medicinal values due to its biological activities decided by physicochemical properties. Recently, the lignin and its derivatives (such as lignosulfonates and lignosulfonate) have been proven efficient in regulating cellular process and the extracellular microenvironment, which has been regarded as the key factor in disease progression. In orthopaedic diseases, especially the degenerative diseases represented by osteoarthritis and osteoporosis, excessive activated inflammation has been proven as a key stage in the pathological process. Due to the excellent biocompatibility, antibacterial and antioxidative activities of lignin and its derivatives, they have been applied to stimulate cells and restore the uncoupling bone remodeling in the degenerative orthopaedic diseases. However, there is a lack of a systemic review to state the current research actuality of lignin and lignin-derived compounds in treating degenerative orthopaedic diseases. Herein, we summarized the current application of lignin and lignin-derived compounds in orthopaedic diseases and proposed their possible therapeutic mechanism in treating degenerative orthopaedic diseases. It is hoped this work could guide the future preparation of lignin/lignin-derived drugs and implants as available therapeutic strategies for clinically degenerative orthopaedic diseases.
Collapse
Affiliation(s)
- Wenhui Pei
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China
| | - Junping Deng
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China
| | - Peng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xucai Wang
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China
| | - Liming Zheng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Yangheng Zhang
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, 210008, China.
| | - Caoxing Huang
- Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
21
|
Kim M, Koyama E, Saunders CM, Querido W, Pleshko N, Pacifici M. Synovial joint cavitation initiates with microcavities in interzone and is coupled to skeletal flexion and elongation in developing mouse embryo limbs. Biol Open 2022; 11:bio059381. [PMID: 35608281 PMCID: PMC9212078 DOI: 10.1242/bio.059381] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/16/2022] [Indexed: 11/20/2022] Open
Abstract
The synovial cavity and its fluid are essential for joint function and lubrication, but their developmental biology remains largely obscure. Here, we analyzed E12.5 to E18.5 mouse embryo hindlimbs and discovered that cavitation initiates around E15.0 with emergence of multiple, discrete, µm-wide tissue discontinuities we term microcavities in interzone, evolving into a single joint-wide cavity within 12 h in knees and within 72-84 h in interphalangeal joints. The microcavities were circumscribed by cells as revealed by mTmG imaging and exhibited a carbohydrate and protein content based on infrared spectral imaging at micro and nanoscale. Accounting for differing cavitation kinetics, we found that the growing femur and tibia anlagen progressively flexed at the knee over time, with peak angulation around E15.5 exactly when the full knee cavity consolidated; however, interphalangeal joint geometry changed minimally over time. Indeed, cavitating knee interzone cells were elongated along the flexion angle axis and displayed oblong nuclei, but these traits were marginal in interphalangeal cells. Conditional Gdf5Cre-driven ablation of Has2 - responsible for production of the joint fluid component hyaluronic acid (HA) - delayed the cavitation process. Our data reveal that cavitation is a stepwise process, brought about by sequential action of microcavities, skeletal flexion and elongation, and HA accumulation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Minwook Kim
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Cheri M. Saunders
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - William Querido
- Department of Bioengineering, Temple University, Philadelphia, PA 19122, USA
| | - Nancy Pleshko
- Department of Bioengineering, Temple University, Philadelphia, PA 19122, USA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, Department of Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
22
|
Olufade O, Negron G, Berrigan W, Sirutis B, Whitley J, Easley K, Chen Y, Mautner K. Amniotic dehydrated cell and protein concentrate versus corticosteroid in knee osteoarthritis: preliminary findings. Regen Med 2022; 17:431-443. [PMID: 35586982 DOI: 10.2217/rme-2022-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Objectives: The purpose is to report preliminary data on clinical response to dehydrated cell and protein concentrate (dCPC) versus corticosteroid (CSI). Design: A single-site prospective, randomized controlled single-blinded trial of patients with knee osteoarthritis. Methods: Pain and function were assessed using a visual analog scale (VAS), the Knee Injury and Osteoarthritis Outcome Score (KOOS) and the Emory Quality of Life (EQOL) measure at 1, 2, 3, 6, 9 and 12 months. Results: 51 patients were enrolled at the time of analysis (27 dCPC, 24 CSI). Both groups demonstrated improvement on the VAS, KOOS and EQOL. Largest differences were observed at 2 (p = 0.05), 3 (p = 0.012) and 6 months (p < 0.001) with a decrease of 1.66 in VAS at 6 months for dCPC (95% CI: -2.67 to -0.65) and 1.34 (95% CI: -2.37 to -0.3) for CSI. Time-averaged measures showed no difference between groups (p = 0.20). Limited data at 9 and 12 months trended toward improvement in the dCPC group. Conclusion: dCPC products may be used as a treatment for knee osteoarthritis. Larger trials are warranted. Clinical Trial Registration: NCT03710005 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Oluseun Olufade
- Department of Orthopedics, Emory University, Atlanta, GA 30329, USA.,Department of Physical Medicine & Rehabilitation, Emory University, Atlanta, GA 30307, USA
| | - Giorgio Negron
- Department of Physical Medicine & Rehabilitation, Emory University, Atlanta, GA 30307, USA
| | - William Berrigan
- Department of Orthopedics, Emory University, Atlanta, GA 30329, USA
| | - Benjamin Sirutis
- Department of Physical Medicine & Rehabilitation, Emory University, Atlanta, GA 30307, USA
| | - Jeremy Whitley
- Department of Orthopedics, Emory University, Atlanta, GA 30329, USA
| | - Kirk Easley
- Department of Biostatistics & Bioinformatics, Rollins School of Public Health Emory University, Atlanta, GA 30329, USA
| | - Yunyun Chen
- Department of Biostatistics & Bioinformatics, Rollins School of Public Health Emory University, Atlanta, GA 30329, USA
| | - Kenneth Mautner
- Department of Orthopedics, Emory University, Atlanta, GA 30329, USA.,Department of Physical Medicine & Rehabilitation, Emory University, Atlanta, GA 30307, USA
| |
Collapse
|
23
|
Aubourg G, Rice SJ, Bruce-Wootton P, Loughlin J. Genetics of osteoarthritis. Osteoarthritis Cartilage 2022; 30:636-649. [PMID: 33722698 PMCID: PMC9067452 DOI: 10.1016/j.joca.2021.03.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/17/2021] [Accepted: 03/06/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis genetics has been transformed in the past decade through the application of large-scale genome-wide association scans. So far, over 100 polymorphic DNA variants have been associated with this common and complex disease. These genetic risk variants account for over 20% of osteoarthritis heritability and the vast majority map to non-protein coding regions of the genome where they are presumed to act by regulating the expression of target genes. Statistical fine mapping, in silico analyses of genomics data, and laboratory-based functional studies have enabled the identification of some of these targets, which encode proteins with diverse roles, including extracellular signaling molecules, intracellular enzymes, transcription factors, and cytoskeletal proteins. A large number of the risk variants correlate with epigenetic factors, in particular cartilage DNA methylation changes in cis, implying that epigenetics may be a conduit through which genetic effects on gene expression are mediated. Some of the variants also appear to have been selected as humans adapted to bipedalism, suggesting that a proportion of osteoarthritis genetic susceptibility results from antagonistic pleiotropy, with risk variants having a positive role in joint formation but a negative role in the long-term health of the joint. Although data from an osteoarthritis genetic study has not yet directly led to a novel treatment, some of the osteoarthritis associated genes code for proteins that have available therapeutics. Genetic investigations are therefore revealing fascinating fundamental insights into osteoarthritis and can expose options for translational intervention.
Collapse
Affiliation(s)
- G Aubourg
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - S J Rice
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - P Bruce-Wootton
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - J Loughlin
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
24
|
Dunn CM, Jeffries MA. The Microbiome in Osteoarthritis: a Narrative Review of Recent Human and Animal Model Literature. Curr Rheumatol Rep 2022; 24:139-148. [PMID: 35389162 DOI: 10.1007/s11926-022-01066-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE OF THE REVIEW The microbiome has recently emerged as a powerful contributor to health and illness in chronic, systemic disorders. Furthermore, new microbiome niches beyond traditional gut locations are frequently being described. Over the past 5 years, numerous pivotal studies have demonstrated associations between changes in various microbiome niches and the development of osteoarthritis (OA). Herein, we review the most impactful recent literature, including microbiome associations with disease and the potential therapeutic value of microbiome manipulation. RECENT FINDINGS The gut microbiome of human OA patients is enriched in specific bacterial clades, most notably Streptococcus, which correlates with OA pain, Firmicutes, and others. Most studies have focused on knee OA, although one publication demonstrated positive associations with 3 gut microbiome clades in hand OA. OA can be easily distinguished from RA by evaluating differences in oral microbiome composition. Most studies have also demonstrated a reduction in richness of the gut microbiome (alpha diversity) associated with OA. Several studies have identified bacterial signatures within human knee and hip cartilage, synovial fluid, and synovial tissue and have described changes in these patterns occurring with the development of OA. In animal models of OA, high-fat diet-induced obesity has been the most well-studied OA risk factor associated with changes in the microbiome, with numerous bacterial clades changed within the gut microbiome and associated with OA. Also in animal models, various oral supplementations, including dietary fiber, probiotics including Lactobacillus species, and cecal microbiome transplantation have all shown improvements in OA histopathology or cartilage healing. Microbiome changes are strongly associated with the OA disease process and with individual OA risk factors related to both the gut microbiome and the microbial DNA patterns in the joint. Microbiome-directed interventions have the potential to prevent or reduce the progression of OA. Future studies should investigate the mechanistic underpinnings of these microbiome associations and further define the therapeutic potential of microbiome augmentation.
Collapse
Affiliation(s)
- Christopher M Dunn
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Laboratory MC400, Oklahoma City, OK, 73104, USA
- Department of Internal Medicine, Division of Rheumatology, Immunology and Allergy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matlock A Jeffries
- Arthritis & Clinical Immunology Program, Oklahoma Medical Research Foundation, 825 NE 13th Street, Laboratory MC400, Oklahoma City, OK, 73104, USA.
- Department of Internal Medicine, Division of Rheumatology, Immunology and Allergy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
25
|
Liu Y, Xuan R, He Y, Ren F, Gu Y. Computation of Fetal Kicking in Various Fetal Health Examinations: A Systematic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:4366. [PMID: 35410056 PMCID: PMC8998667 DOI: 10.3390/ijerph19074366] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/28/2022] [Accepted: 04/02/2022] [Indexed: 11/16/2022]
Abstract
Fetal movement has always been considered an essential indicator to evaluate the health of the unborn fetus. Many factors affect fetal movement. The frequency of fetal kicking is an important measurement of whether fetal development is progressing and healthy. Various instruments and methods of detecting fetal movement have been used and each method has its advantages and disadvantages. Although limited by the fetal environment in utero, the finite element method and musculoskeletal model can be used to calculate fetal lower limb movement. This review aims to summarize the current detection techniques for fetal movement, especially in the lower limbs. These will be outlined by describing the different measurements of fetal movement, and the related biomechanical analyses of fetal lower limb skeletogenesis and the associated muscular development to better evaluate and calculate the movements of the fetus in the womb.
Collapse
Affiliation(s)
- Yuwei Liu
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China; (Y.L.); (Y.H.); (Y.G.)
| | - Rongrong Xuan
- Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China
| | - Yuhuan He
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China; (Y.L.); (Y.H.); (Y.G.)
- Department of Physical and Health Education, Udon Thani Rajabhat University, Udon Thani 41000, Thailand
| | - Feng Ren
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China; (Y.L.); (Y.H.); (Y.G.)
| | - Yaodong Gu
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China; (Y.L.); (Y.H.); (Y.G.)
- Faculty of Informatics, Eötvös Loránd University, H-9700 Szombathely, Hungary
| |
Collapse
|
26
|
Muenzebrock KA, Kersten V, Alblas J, Garcia JP, Creemers LB. The Added Value of the “Co” in Co-Culture Systems in Research on Osteoarthritis Pathology and Treatment Development. Front Bioeng Biotechnol 2022; 10:843056. [PMID: 35309991 PMCID: PMC8927651 DOI: 10.3389/fbioe.2022.843056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a highly prevalent disease and a major health burden. Its development and progression are influenced by factors such as age, obesity or joint overuse. As a whole organ disease OA affects not only cartilage, bone and synovium but also ligaments, fatty or nervous tissue surrounding the joint. These joint tissues interact with each other and understanding this interaction is important in developing novel treatments. To incorporate and study these interactions in OA research, several co-culture models have evolved. They combine two or more cell types or tissues and investigate the influence of amongst others inflammatory or degenerative stimuli seen in OA. This review focuses on co-cultures and the differential processes occurring in a given tissue or cell as a consequence of being combined with another joint cell type or tissue, and/or the extent to which a co-culture mimics the in vivo processes. Most co-culture models depart from synovial lining and cartilage culture, but also fat pad and bone have been included. Not all of the models appear to reflect the postulated in vivo OA pathophysiology, although some of the discrepancies may indicate current assumptions on this process are not entirely valid. Systematic analysis of the mutual influence the separate compartments in a given model exert on each other and validation against in vivo or ex vivo observation is still largely lacking and would increase their added value as in vitro OA models.
Collapse
|
27
|
Loughlin J. Translating osteoarthritis genetics research: challenging times ahead. Trends Mol Med 2022; 28:176-182. [PMID: 35033441 DOI: 10.1016/j.molmed.2021.12.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 12/15/2022]
Abstract
The ultimate goal of molecular genetic studies of human diseases is to translate the discoveries for patient benefit. For diseases that lack licensed disease-modifying therapeutics, such as osteoarthritis (OA), the need is acute. OA is polygenic and affects older individuals, with a recent genome-wide study of over 800 000 individuals adding 52 novel association signals to those already reported on for this common arthritis. Many of the predicted effector genes of these signals encode proteins that are targets of drugs for other indications, highlighting repurposing opportunities. Here, the potential for OA genetic data to translate is discussed, including whether the developmental origin of OA will limit the application of genetic risk data for disease-modification purposes.
Collapse
Affiliation(s)
- John Loughlin
- Newcastle University, Biosciences Institute, International Centre for Life, Newcastle upon Tyne, NE1 3BZ, UK.
| |
Collapse
|
28
|
Wang Y, Meng F, Wu J, Long H, Li J, Wu Z, He H, Wang H, Wang N, Xie D. Associations between adipokines gene polymorphisms and knee osteoarthritis: a meta-analysis. BMC Musculoskelet Disord 2022; 23:166. [PMID: 35193537 PMCID: PMC8864815 DOI: 10.1186/s12891-022-05111-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Background Adipokines gene polymorphisms are speculated to be associated with the risk of knee osteoarthritis (OA), but evidence remains conflicting. This study therefore aimed to examine whether associations exist between adipokines gene polymorphisms and knee OA by considering the evidence collected from eligible studies through a meta-analysis. Methods A systematic search was performed on PubMed, Embase, Web of Science, China National Knowledge Infrastructure (CNKI), and Wanfang up to March 31, 2020. Meta-analysis was carried out by focusing on the associations between adipokines gene polymorphisms and knee OA with the allele model, dominant model, and recessive model. Results The present meta-analysis included 5 eligible studies for ADIPOQ rs1501299 with 1,021 cases and 1,097 controls, 3 eligible studies for ADIPOQ rs2241766 with 549 cases and 544 controls, 3 eligible studies for LEPR rs1137101 with 808 cases and 856 controls, 2 eligible studies for VISFATIN rs4730153 with 339 cases and 680 controls and 2 eligible studies for VISFATIN rs16872158 with 339 cases and 680 controls. Significant association was observed between LEPR rs1137101 and knee OA in the overall population (recessive: OR = 0.40, 95% CI 0.21–0.79). Limited data revealed that associations may exist between ADIPOQ rs2241766 and knee OA in Asians (dominant: OR = 1.35, 95% CI 1.03–1.78), between VISFATIN rs4730153 and knee OA in Asians (allele: OR = 0.58, 95% CI 0.41–0.83; dominant: OR = 0.57, 95% CI 0.39–0.83), and between VISFATIN rs16872158 and knee OA in Asians (allele: OR = 1.84, 95% CI 1.26–2.68; dominant: OR = 1.94, 95% CI 1.31–2.89). Conclusions Adipokines gene polymorphisms may be associated with knee OA. The association was observed in LEPR rs1137101 in the present study. In addition, limited data revealed that associations may also exist in ADIPOQ rs2241766, VISFATIN rs4730153 and VISFATIN rs16872158. Prospero registration CRD42020187664. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-05111-4.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fanqiang Meng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Wu
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Huizhong Long
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiatian Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziying Wu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongyi He
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haochen Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ning Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dongxing Xie
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China. .,Hunan Engineering Research Center for Osteoarthritis, Changsha, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
29
|
Fazeli MS, McIntyre L, Huang Y, Chevalier X. Intra-articular placebo effect in the treatment of knee osteoarthritis: a survey of the current clinical evidence. Ther Adv Musculoskelet Dis 2022; 14:1759720X211066689. [PMID: 35126683 PMCID: PMC8808023 DOI: 10.1177/1759720x211066689] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/23/2021] [Indexed: 01/06/2023] Open
Abstract
Knee osteoarthritis (KOA) is a debilitating disease characterized by chronic pain, stiffness, and decreased mobility. Intra-articular injectable therapies show good clinical efficacy in improving symptoms; however, these therapies and their comparators (intra-articular saline) have been associated with a large underlying placebo effect. We aimed to describe the existing evidence on the challenges, hypotheses, and potential solutions to mitigate the intra-articular placebo effect in clinical trials in KOA. A targeted literature review was conducted by searching Embase, MEDLINE®, and CENTRAL using predefined study selection criteria. All eligible studies identified were extracted for relevant data, and results were narratively summarized. Forty-three studies were included following screening. Challenges associated with the intra-articular placebo effect included its ability to mask the comparative efficacy of active treatments in trials (n = 7 studies), long-lasting effects (up to 6 months; n = 3), and substantial variation of placebo effect sizes across populations (n = 3). Hypotheses for the mechanism of the placebo effect included aspiration of synovial fluid during administration (n = 6) and dilution of inflammatory mediators (n = 2). Factors affecting the placebo effect size were more invasive routes of administration (e.g., injection versus oral; n = 4) and patient expectations (n = 2). Proposed solutions included the suggestion for readers to weigh the relevance of clinical trial evidence against the presence of large underlying placebo effects (n = 9), discontinuation of intra-articular saline as an appropriate placebo (n = 5), and inclusion of 'no treatment' or sham injection as a control (n = 4). The intra-articular placebo effect is a well-documented occurrence in KOA clinical trials, and it is suggested that it be accounted for when designing randomized controlled trials. Awareness and understanding of the intra-articular placebo effect in KOA are required for fair interpretation of clinical trial evidence.
Collapse
Affiliation(s)
- Mir Sohail Fazeli
- Evidinno Outcomes Research Inc., 1750 Davie Street, Suites 601 & 602, Vancouver, BC V6G 1W3, Canada
| | | | - Yili Huang
- Northwell Health, New Hyde Park, NY, USA
| | - Xavier Chevalier
- Hôpital Henri Mondor, Université Paris XII, UPEC, Créteil, France
| |
Collapse
|
30
|
Fatima S, Khan B, Khan OY, Amjad M, Zehra S, Azhar A. Tetra-primers ARMS-PCR Based Association Analyses of Synonymous and Intronic Variants in the ADAM12 Gene with Susceptibility to Knee Osteoarthritis: A Case-Control Study. Biochem Genet 2022; 60:1695-1715. [PMID: 35083608 DOI: 10.1007/s10528-022-10189-5] [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: 06/25/2021] [Accepted: 01/05/2022] [Indexed: 11/28/2022]
Abstract
Genetic variations in a disintegrin and metalloprotease 12 (ADAM12) gene may contribute to develop Osteoarthritis (OA) that is characterized by cartilage matrix degradation and osteophytes formation. Therefore, the aim of present study was to analyze the association between the ADAM12 gene variants and knee OA predisposition. Tetra-primers ARMS-PCR was employed, to genotype the ADAM12 gene polymorphisms (rs1044122 and rs1871054) in 400 knee OA patients and equal number of age-matched controls. The association between ADAM12 gene variants and OA susceptibility was estimated using the Chi-square, logistic regression, haplotypes and linkage analyses. A significant association of rs1044122 (genotype: χ2 = 18.94; P < 0.001, allele: χ2 = 19.10; P < 0.001) and rs1871054 (genotype: χ2 = 10.04; P = 0.007, allele: χ2 = 10.57; P = 0.001) was observed with increased OA susceptibility. The variant genotype of rs1044122 increased OA risk more than twice [odds ratio (OR) 2.20; P = 0.001] and the risk was higher in females (OR 2.43; P = 0.001). The variant genotype of rs1871054 was perceived to almost double the risk in females (OR 1.97; P = 0.003). Moreover, a significant association of rs1044122 and rs1871054 under the additive genetic model (P < 0.001 and P = 0.002, respectively) was observed. The targeted ADAM12 gene polymorphisms, showed significant association with knee OA susceptibility. Females harboring the polymorphisms might be at risk. Besides, the haplotype CC of rs1044122 and rs1871054 in the ADAM12 gene may double knee OA risk. These findings may help in determining the etiology of OA and recognizing the people at risk of developing knee OA.
Collapse
Affiliation(s)
- Sehrish Fatima
- The Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), Faculty of Science, University of Karachi, Karachi, Pakistan.
| | - Bushra Khan
- The Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), Faculty of Science, University of Karachi, Karachi, Pakistan
| | - Obaid Yusuf Khan
- Department of Genetics, Faculty of Science, University of Karachi, Karachi, Sindh, Pakistan
| | - Maryam Amjad
- The Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), Faculty of Science, University of Karachi, Karachi, Pakistan
| | - Sitwat Zehra
- The Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), Faculty of Science, University of Karachi, Karachi, Pakistan
| | - Abid Azhar
- The Karachi Institute of Biotechnology and Genetic Engineering (KIBGE), Faculty of Science, University of Karachi, Karachi, Pakistan
| |
Collapse
|
31
|
Yurube T, Iguchi T, Kinoshita K, Sadamitsu T, Kakutani K. Upper Cervical Compression Myelopathy Caused by the Retro-Odontoid Pseudotumor With Degenerative Osteoarthritis and Calcium Pyrophosphate Dihydrate Disease: A Case Report and Literature Review. Neurospine 2022; 18:903-913. [PMID: 35000348 PMCID: PMC8752696 DOI: 10.14245/ns.2142112.056] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 06/29/2021] [Indexed: 02/05/2023] Open
Abstract
The retro-odontoid pseudotumor is often concurrent with atlantoaxial subluxation (AAS). Therefore, the pseudotumor is relatively common in rheumatoid arthritis (RA) but rare in primary osteoarthritis (OA). This is a case report of an elderly male patient suffering from neck pain and compression myelopathy caused by the craniocervical pseudotumor with OA but without atlantoaxial instability. He had long-lasting peripheral and spinal pain treated by nonsteroidal anti-inflammatory drugs. Imaging found upper cervical spondylosis without AAS or dynamic instability but with periodontoid calcifications and ossifications, suggesting calcium pyrophosphate dihydrate (CPPD) crystal deposition. Based on a comprehensive literature search and review, CPPD disease around the atlantodental joint is a possible contributor to secondary OA development and retro-odontoid pannus formation through chronic inflammation, which can be enough severe to induce compression myelopathy in non-RA patients without AAS. The global increase in the aged population advises caution regarding more prevalent upper cervical spine disorders associated with OA and CPPD.
Collapse
Affiliation(s)
- Takashi Yurube
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tetsuhiro Iguchi
- Department of Orthopedic Surgery, Saiseikai Hyogo Prefecture Hospital, Kobe, Japan
| | - Keisuke Kinoshita
- Department of Orthopedic Surgery, Saiseikai Hyogo Prefecture Hospital, Kobe, Japan
| | - Takashi Sadamitsu
- Department of Orthopedic Surgery, Saiseikai Hyogo Prefecture Hospital, Kobe, Japan
| | - Kenichiro Kakutani
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
32
|
Ge Y, Zhou C, Xiao X, Jin Z, Zhou L, Chen Z, Liu F, Yuan Q, Zhang G, Shan L, Tong P. A Novel Mutation of the KLK6 Gene in a Family With Knee Osteoarthritis. Front Genet 2021; 12:784176. [PMID: 34858488 PMCID: PMC8631809 DOI: 10.3389/fgene.2021.784176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
To investigate the correlation between gene mutation and knee osteoarthritis (KOA), a whole-exome sequencing (WES) was applied to analyze blood samples of four KOA patients and two normal subjects in a family. Gene mutations were identified by gene-trapping and high-throughput sequencing analysis across the differences between the patients and normal subjects. The interactive gene network analysis on the retrieval of interacting genes (STRING) database and the KOA-related genes expression data sets was performed. A possibly detrimental and nonsynonymous mutation at the kallikrein-related peptidase 6 (KLK6) gene (rs201586262, c. C80A, P27H) was identified and attracted our attention. KLK6 belongs to the kallikrein family of serine proteases and its serum level is known as a prevalent biomarker in inflammatory and malignant diseases. KLK6 expresses in the extracellular compartment for matrix degradation, highlighting that KLK6 plays a role in the pathogenesis of KOA. By using the gene databases, the KOA-related genes were mined after de-duplication and IL6 was selected as the most relevant gene through interactive analysis of protein-protein interaction (PPI) network. The data suggested that KLK6 gene mutation and the related expression alteration of IL6 gene might determine the occurrence of hereditary KOA. The is the first study discovering the gene mutation of KLK6 as a factor of pathogenesis of KOA, especially the hereditary KOA.
Collapse
Affiliation(s)
- Yanzhi Ge
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenfen Zhou
- National Genomics Data Center, CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiujuan Xiao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijiang Jin
- Department of Orthopaedics, The 9th People's Hospital of Hangzhou, Hangzhou, China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zuxiang Chen
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fucun Liu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Qiang Yuan
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guoqing Zhang
- National Genomics Data Center, CAS Key Laboratory of Computational Biology, Bio-Med Big Data Center, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Peijian Tong
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
33
|
Mobasheri A, Kapoor M, Ali SA, Lang A, Madry H. The future of deep phenotyping in osteoarthritis: How can high throughput omics technologies advance our understanding of the cellular and molecular taxonomy of the disease? OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100144. [PMID: 36474763 PMCID: PMC9718223 DOI: 10.1016/j.ocarto.2021.100144] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is the most common form of musculoskeletal disease with significant healthcare costs and unmet needs in terms of early diagnosis and treatment. Many of the drugs that have been developed to treat OA failed in phase 2 and phase 3 clinical trials or produced inconclusive and ambiguous results. High throughput omics technologies are a powerful tool to better understand the mechanisms of the development of OA and other arthritic diseases. In this paper we outline the strategic reasons for increasingly applying deep phenotyping in OA for the benefit of gaining a better understanding of disease mechanisms and developing targeted treatments. This editorial is intended to launch a special themed issue of Osteoarthritis and Cartilage Open addressing the timely topic of "Advances in omics technologies for deep phenotyping in osteoarthritis". High throughput omics technologies are increasingly being applied in mechanistic studies of OA and other arthritic diseases. Applying multi-omics approaches in OA is a high priority and will allow us to gather new information on disease pathogenesis at the cellular level, and integrate data from diverse omics technology platforms to enable deep phenotyping. We anticipate that new knowledge in this area will allow us to harness the power of Big Data Analytics and resolve the extremely complex and overlapping clinical phenotypes into molecular endotypes, revealing new information about the cellular taxonomy of OA and "druggable pathways", thus facilitating future drug development.
Collapse
Affiliation(s)
- Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Departments of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Mohit Kapoor
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Shabana Amanda Ali
- Bone and Joint Center, Henry Ford Health System, Detroit, MI, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Annemarie Lang
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany
- German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, Homburg, Germany
| |
Collapse
|
34
|
Cheng X, Xu P. The articular cartilage preservative effects of genistein in an experimental model of knees osteoarthritis. Appl Physiol Nutr Metab 2021; 46:1331-1336. [PMID: 33989507 DOI: 10.1139/apnm-2020-0958] [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] [Indexed: 11/22/2022]
Abstract
The study aimed to investigate the preservative effects of genistein on articular cartilage in an experimental model of knee osteoarthritis in rats. Thirty male Wistar rats were assigned to 3 equal groups: sham group, osteoarthritis control group (OAG), and genistein-treated osteoarthritis group (GTG). Intra-articular injections of monosodium iodoacetate were used for osteoarthritis induction. After 2 weeks of rest for the induction of the inflammatory process, genistein (30 mg/kg/day) vs. saline gavage was administered for 8 weeks. The expression of matrix metalloproteinase (MMP)-8 and MMP-13, Sox5/Sox6, Indian hedgehog (IHH), and Col2 were evaluated in medial femoral condyle sections by immunohistochemical staining. The number of chondrocytes and cartilage thicknesses were also measured and compared among the groups. No significant change in cartilage thickness was observed in GTG compared with OAG (p = 0.188). Chondrocyte count was significantly higher in the articular cartilage of GTG compared with OAG (p = 0.006). Induction of osteoarthritis significantly increased the expression of MMP-8, MMP-13, and IHH, but decreased Col2, Sox5, and Sox6 expression (p < 0.001); these were partially prevented in the GTG. Our findings support the effectiveness of genistein treatment in the prevention of articular cartilage damage in the experimental model of knee osteoarthritis. The proposed mechanism of action is through the suppression of the MMP, IHH, and Col2 pathways, besides the induction of Sox5 and Sox6 expression. Novelty: Genistein prevents articular cartilage damage in the experimental model of knee osteoarthritis. The osteoprotective effect is manifested by the modulation of expression of MMP, Sox, IHH, and Col2 proteins.
Collapse
Affiliation(s)
- Xiangjun Cheng
- Department of Orthopaedics, Second Affiliated Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing 400010, China
| | - Peilian Xu
- Department One of Orthopedics, Ninth People's Hospital of Chongqing, Chongqing 400700, China
| |
Collapse
|
35
|
Wallace IJ, Riew GJ, Landau R, Bendele AM, Holowka NB, Hedrick TL, Konow N, Brooks DJ, Lieberman DE. Experimental evidence that physical activity inhibits osteoarthritis: Implications for inferring activity patterns from osteoarthritis in archeological human skeletons. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2021. [DOI: 10.1002/ajpa.24429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Ian J. Wallace
- Department of Human Evolutionary Biology Harvard University Cambridge Massachusetts USA
- Department of Anthropology University of New Mexico Albuquerque New Mexico USA
| | - Grant J. Riew
- Department of Human Evolutionary Biology Harvard University Cambridge Massachusetts USA
- Harvard Medical School Boston Massachusetts USA
| | - Rebecca Landau
- Department of Human Evolutionary Biology Harvard University Cambridge Massachusetts USA
| | | | - Nicholas B. Holowka
- Department of Human Evolutionary Biology Harvard University Cambridge Massachusetts USA
- Department of Anthropology University at Buffalo Buffalo New York USA
| | - Tyson L. Hedrick
- Department of Biology University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Nicolai Konow
- Department of Biological Sciences University of Massachusetts Lowell Lowell Massachusetts USA
| | - Daniel J. Brooks
- Center for Advanced Orthopaedic Studies Beth Israel Deaconess Medical Center, Harvard Medical School Boston Massachusetts USA
| | - Daniel E. Lieberman
- Department of Human Evolutionary Biology Harvard University Cambridge Massachusetts USA
| |
Collapse
|
36
|
Rochoux Q, Sopkova-de Oliveira Santos J, Marcelli C, Rovelet-Lecrux A, Chevallier V, Dutheil JJ, Leclercq S, Boumédiene K, Baugé C, Aury-Landas J. Description of Joint Alterations Observed in a Family Carrying p.Asn453Ser COMP Variant: Clinical Phenotypes, In Silico Prediction of Functional Impact on COMP Protein and Stability, and Review of the Literature. Biomolecules 2021; 11:biom11101460. [PMID: 34680093 PMCID: PMC8533395 DOI: 10.3390/biom11101460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/20/2021] [Accepted: 09/30/2021] [Indexed: 12/26/2022] Open
Abstract
The role of genetics in the development of osteoarthritis is well established but the molecular bases are not fully understood. Here, we describe a family carrying a germline mutation in COMP (Cartilage Oligomeric Matrix Protein) associated with three distinct phenotypes. The index case was enrolled for a familial form of idiopathic early-onset osteoarthritis. By screening potential causal genes for osteoarthritis, we identified a heterozygous missense mutation of COMP (c.1358C>T, p.Asn453Ser), absent from genome databases, located on a highly conserved residue and predicted to be deleterious. Molecular dynamics simulation suggests that the mutation destabilizes the overall COMP protein structure and consequently the calcium releases from neighboring calcium binding sites. This mutation was once reported in the literature as causal for severe multiple epiphyseal dysplasia (MED). However, no sign of dysplasia was present in the index case. The mutation was also identified in one of her brothers diagnosed with MED and secondary osteoarthritis, and in her sister affected by an atypical syndrome including peripheral inflammatory arthritis of unknown cause, without osteoarthritis nor dysplasia. This article suggests that this mutation of COMP is not only causal for idiopathic early-onset osteoarthritis or severe MED, but can also be associated to a broad phenotypic variability with always joint alterations.
Collapse
Affiliation(s)
- Quitterie Rochoux
- EA7451 BioConnecT, Normandie Université, UNICAEN, 14032 Caen, France; (Q.R.); (S.L.); (K.B.); (C.B.)
- Service de Rhumatologie, CHU de Caen, 14000 Caen, France;
| | | | | | - Anne Rovelet-Lecrux
- Department of Genetics and CNR-MAJ, Normandy Center for Genomic and Personalized Medicine, Normandie Université, UNIROUEN, Inserm U1245 and Rouen University Hospital, 76000 Rouen, France;
| | - Virginie Chevallier
- Délégation de la Recherche Clinique et de l’Innovation, CHU de Caen, 14000 Caen, France; (V.C.); (J.-J.D.)
| | - Jean-Jacques Dutheil
- Délégation de la Recherche Clinique et de l’Innovation, CHU de Caen, 14000 Caen, France; (V.C.); (J.-J.D.)
| | - Sylvain Leclercq
- EA7451 BioConnecT, Normandie Université, UNICAEN, 14032 Caen, France; (Q.R.); (S.L.); (K.B.); (C.B.)
- Service de Chirurgie Orthopédique, Clinique Saint-Martin, 14000 Caen, France
| | - Karim Boumédiene
- EA7451 BioConnecT, Normandie Université, UNICAEN, 14032 Caen, France; (Q.R.); (S.L.); (K.B.); (C.B.)
| | - Catherine Baugé
- EA7451 BioConnecT, Normandie Université, UNICAEN, 14032 Caen, France; (Q.R.); (S.L.); (K.B.); (C.B.)
| | - Juliette Aury-Landas
- EA7451 BioConnecT, Normandie Université, UNICAEN, 14032 Caen, France; (Q.R.); (S.L.); (K.B.); (C.B.)
- Correspondence: ; Tel.: +33-(0)2-31-56-82-18
| |
Collapse
|
37
|
Chang L, Yao H, Yao Z, Ho KKW, Ong MTY, Dai B, Tong W, Xu J, Qin L. Comprehensive Analysis of Key Genes, Signaling Pathways and miRNAs in Human Knee Osteoarthritis: Based on Bioinformatics. Front Pharmacol 2021; 12:730587. [PMID: 34497524 PMCID: PMC8419250 DOI: 10.3389/fphar.2021.730587] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/12/2021] [Indexed: 01/13/2023] Open
Abstract
Background: Osteoarthritis (OA) is one of the main causes of disability in the elderly population, accompanied by a series of underlying pathologic changes, such as cartilage degradation, synovitis, subchondral bone sclerosis, and meniscus injury. The present study aimed to identify key genes, signaling pathways, and miRNAs in knee OA associated with the entire joint components, and to explain the potential mechanisms using computational analysis. Methods: The differentially expressed genes (DEGs) in cartilage, synovium, subchondral bone, and meniscus were identified using the Gene Expression Omnibus 2R (GEO2R) analysis based on dataset from GSE43923, GSE12021, GSE98918, and GSE51588, respectively and visualized in Volcano Plot. Venn diagram analyses were performed to identify the overlapping DEGs (overlapping DEGs) that expressed in at least two types of tissues mentioned above. Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, protein-protein interaction (PPI) analysis, and module analysis were conducted. Furthermore, qRT-PCR was performed to validate above results using our clinical specimens. Results: As a result, a total of 236 overlapping DEGs were identified, of which 160 were upregulated and 76 were downregulated. Through enrichment analysis and constructing the PPI network and miRNA-mRNA network, knee OA-related key genes, such as HEY1, AHR, VEGFA, MYC, and CXCL12 were identified. Clinical validation by qRT-PCR experiments further supported above computational results. In addition, knee OA-related key miRNAs such as miR-101, miR-181a, miR-29, miR-9, and miR-221, and pathways such as Wnt signaling, HIF-1 signaling, PI3K-Akt signaling, and axon guidance pathways were also identified. Among above identified knee OA-related key genes, pathways and miRNAs, genes such as AHR, HEY1, MYC, GAP43, and PTN, pathways like axon guidance, and miRNAs such as miR-17, miR-21, miR-155, miR-185, and miR-1 are lack of research and worthy for future investigation. Conclusion: The present informatic study for the first time provides insight to the potential therapeutic targets of knee OA by comprehensively analyzing the overlapping genes differentially expressed in multiple joint components and their relevant signaling pathways and interactive miRNAs.
Collapse
Affiliation(s)
- Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China
| | - Zhi Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China
| | - Kevin Ki-Wai Ho
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China
| | - Michael Tim-Yun Ong
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, SAR China
| |
Collapse
|
38
|
Boer CG, Hatzikotoulas K, Southam L, Stefánsdóttir L, Zhang Y, Coutinho de Almeida R, Wu TT, Zheng J, Hartley A, Teder-Laving M, Skogholt AH, Terao C, Zengini E, Alexiadis G, Barysenka A, Bjornsdottir G, Gabrielsen ME, Gilly A, Ingvarsson T, Johnsen MB, Jonsson H, Kloppenburg M, Luetge A, Lund SH, Mägi R, Mangino M, Nelissen RRGHH, Shivakumar M, Steinberg J, Takuwa H, Thomas LF, Tuerlings M, Babis GC, Cheung JPY, Kang JH, Kraft P, Lietman SA, Samartzis D, Slagboom PE, Stefansson K, Thorsteinsdottir U, Tobias JH, Uitterlinden AG, Winsvold B, Zwart JA, Davey Smith G, Sham PC, Thorleifsson G, Gaunt TR, Morris AP, Valdes AM, Tsezou A, Cheah KSE, Ikegawa S, Hveem K, Esko T, Wilkinson JM, Meulenbelt I, Lee MTM, van Meurs JBJ, Styrkársdóttir U, Zeggini E. Deciphering osteoarthritis genetics across 826,690 individuals from 9 populations. Cell 2021; 184:4784-4818.e17. [PMID: 34450027 PMCID: PMC8459317 DOI: 10.1016/j.cell.2021.07.038] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/26/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022]
Abstract
Osteoarthritis affects over 300 million people worldwide. Here, we conduct a genome-wide association study meta-analysis across 826,690 individuals (177,517 with osteoarthritis) and identify 100 independently associated risk variants across 11 osteoarthritis phenotypes, 52 of which have not been associated with the disease before. We report thumb and spine osteoarthritis risk variants and identify differences in genetic effects between weight-bearing and non-weight-bearing joints. We identify sex-specific and early age-at-onset osteoarthritis risk loci. We integrate functional genomics data from primary patient tissues (including articular cartilage, subchondral bone, and osteophytic cartilage) and identify high-confidence effector genes. We provide evidence for genetic correlation with phenotypes related to pain, the main disease symptom, and identify likely causal genes linked to neuronal processes. Our results provide insights into key molecular players in disease processes and highlight attractive drug targets to accelerate translation.
Collapse
Affiliation(s)
- Cindy G Boer
- Department of Internal Medicine, Erasmus MC, Medical Center, 3015CN Rotterdam, the Netherlands
| | - Konstantinos Hatzikotoulas
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Lorraine Southam
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger Health System, Danville, PA 17822, USA
| | - Rodrigo Coutinho de Almeida
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - Tian T Wu
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jie Zheng
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - April Hartley
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; Musculoskeletal Research Unit, Translation Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK
| | - Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Anne Heidi Skogholt
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa 230-0045, Japan
| | - Eleni Zengini
- 4(th) Psychiatric Department, Dromokaiteio Psychiatric Hospital, 12461 Athens, Greece
| | - George Alexiadis
- 1(st) Department of Orthopaedics, KAT General Hospital, 14561 Athens, Greece
| | - Andrei Barysenka
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Maiken E Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Thorvaldur Ingvarsson
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland; Department of Orthopedic Surgery, Akureyri Hospital, 600 Akureyri, Iceland
| | - Marianne B Johnsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0316 Oslo, Norway; Research and Communication Unit for Musculoskeletal Health (FORMI), Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, 0424 Oslo, Norway
| | - Helgi Jonsson
- Department of Medicine, Landspitali The National University Hospital of Iceland, 108 Reykjavik, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Margreet Kloppenburg
- Departments of Rheumatology and Clinical Epidemiology, Leiden University Medical Center, 9600, 23OORC Leiden, the Netherlands
| | - Almut Luetge
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | | | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Rob R G H H Nelissen
- Department of Orthopaedics, Leiden University Medical Center, 9600, 23OORC Leiden, the Netherlands
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julia Steinberg
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW 1340, Australia
| | - Hiroshi Takuwa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo 108-8639, Japan; Department of Orthopedic Surgery, Shimane University, Shimane 693-8501, Japan
| | - Laurent F Thomas
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway; BioCore-Bioinformatics Core Facility, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Margo Tuerlings
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - George C Babis
- 2(nd) Department of Orthopaedics, National and Kapodistrian University of Athens, Medical School, Nea Ionia General Hospital Konstantopouleio, 14233 Athens, Greece
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jae Hee Kang
- Department of Medicine, Brigham and Women's Hospital, 181 Longwood Ave, Boston, MA 02115, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Steven A Lietman
- Musculoskeletal Institute, Geisinger Health System, Danville, PA 17822, USA
| | - Dino Samartzis
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - Kari Stefansson
- deCODE Genetics/Amgen Inc., 102 Reykjavik, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen Inc., 102 Reykjavik, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Jonathan H Tobias
- Musculoskeletal Research Unit, Translation Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, Medical Center, 3015CN Rotterdam, the Netherlands
| | - Bendik Winsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | - John-Anker Zwart
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Pak Chung Sham
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | - Tom R Gaunt
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester M13 9LJ, UK
| | - Ana M Valdes
- Faculty of Medicine and Health Sciences, School of Medicine, University of Nottingham, Nottingham, Nottinghamshire NG5 1PB, UK
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa 411 10, Greece
| | - Kathryn S E Cheah
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo 108-8639, Japan
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; HUNT Research Center, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - J Mark Wilkinson
- Department of Oncology and Metabolism and Healthy Lifespan Institute, University of Sheffield, Sheffield S10 2RX, UK
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - Ming Ta Michael Lee
- Genomic Medicine Institute, Geisinger Health System, Danville, PA 17822, USA; Institute of Biomedical Sciences, Academia Sinica, 115 Taipei, Taiwan
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus MC, Medical Center, 3015CN Rotterdam, the Netherlands
| | | | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; TUM School of Medicine, Technical University of Munich and Klinikum Rechts der Isar, 81675 Munich, Germany.
| |
Collapse
|
39
|
Park S, Min S, Park SH, Yoo J, Jee YS. Influence of Isometric Exercise Combined With Electromyostimulation on Inflammatory Cytokine Levels, Muscle Strength, and Knee Joint Function in Elderly Women With Early Knee Osteoarthritis. Front Physiol 2021; 12:688260. [PMID: 34326779 PMCID: PMC8313868 DOI: 10.3389/fphys.2021.688260] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022] Open
Abstract
Background and Objectives: Muscle strengthening exercise is suggested to beneficial for patients with knee osteoarthritis (OA) and electrical muscular stimulation is reported to be effective in improvement of muscle strength. This study examined whether isometric exercise combined with whole body-electromyostimulation (WB-EMS) can improve serum cytokine levels, muscle strength, and knee function in elderly women with early knee OA. Materials and Methods: This randomized controlled study included 75 participants assigned into three groups: the control group (CON), isometric exercise group (ISOM), and isometric exercise and electromyostimulation group (ISOM + EMS). The two exercise groups performed their respective programs for 8 weeks, 3 days a week, 30 min a day. The main exercises for both groups were performed continuously during the 20 min in an alternation of a 6-s contraction with a 4-s break. At pre- and post-intervention, anthropometric variables, muscle strength, Knee Injury and Osteoarthritis Outcome Score (KOOS), and blood sampling for biomarkers including interleukin-6, tumor necrosis factor-α, C-reactive protein, and resistin were performed. Results: All variables at pre-intervention showed no significant differences among the three groups. However, there were significant differences between groups for body composition, muscle strength, KOOS subscale scores, and biomarkers. ISOM + EMS group resulted in a significant reduction in body weight, fat mass, fat percentage, inflammatory cytokine levels, and increased muscle strength. An ISOM + EMS group had the best KOOS score among all groups. Conclusion: Isometric exercise combined with WB-EMS resulted in the best overall improvements in knee function and alleviating the pain and symptoms of patients with early knee OA. Further, reduced levels of inflammatory cytokines were observed. These non-pharmacologic, non-invasive interventions should be considered by healthcare specialists for elderly patients with early knee OA.
Collapse
Affiliation(s)
- Sunhee Park
- Research Institute of Sports and Industry Science, Hanseo University, Seosan, South Korea
| | - Sukyung Min
- Research Institute of Sports and Industry Science, Hanseo University, Seosan, South Korea
| | - Si-Hwa Park
- Research Institute of Sports and Industry Science, Hanseo University, Seosan, South Korea
| | - Jaehyun Yoo
- Department of Physical Education, Sahmyook University, Seoul, South Korea
| | - Yong-Seok Jee
- Research Institute of Sports and Industry Science, Hanseo University, Seosan, South Korea
| |
Collapse
|
40
|
A Current Update on Pelvifemoral Conditions That Should be in the Differential Diagnosis for Patients With Lower Extremity Radiculopathy. Clin Spine Surg 2021; 34:206-215. [PMID: 34121075 DOI: 10.1097/bsd.0000000000001180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 02/24/2021] [Indexed: 11/27/2022]
Abstract
Patients presenting to an outpatient spine clinic frequently report symptoms of low back pain with associated buttock, groin, and lower extremity pain. While many of these individuals suffer from lumbar spine radiculopathy, a number of different orthopedic pathologies can mimic these symptoms. Management depends substantially on a detailed history and physical examination, in addition to working from a broad list of differential diagnoses when evaluating these patients. It is imperative that spine practitioners have a comprehensive understanding of the differential diagnoses that may mimic those originating from the lumbar spine, especially when a patient's symptoms are atypical from classic radicular pain. Misdiagnosis can lead to unnecessary testing and treatment, while delaying an accurate clinical assessment and treatment plan. This review highlights common orthopedic diagnoses that may present similar to lumbar spine pathologies and the evidence-based evaluation of these conditions.
Collapse
|
41
|
Alonso-Llamazares C, Blanco Márquez B, Lopez B, Pardiñas AF. Assessing individual and population variability in degenerative joint disease prevalence using generalized linear mixed models. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2021; 175:611-625. [PMID: 33336804 DOI: 10.1002/ajpa.24195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/30/2020] [Accepted: 11/19/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVES In this paper, we introduce the use of generalized linear mixed models (GLMM) as a better alternative to traditional statistical methods for studying factors associated to the prevalence of degenerative joint disease (DJD) in bioarchaeological contexts. MATERIALS AND METHODS DJD prevalence was assessed for the appendicular joints and the spine of a Spanish population dated from the 15th to the 18th century. Data were analyzed using contingency tables, logistic regression models, and logistic GLMM. RESULTS In general, results from GLMMs find agreement in other methods. However, by being able to analyze the data at the level of individual bones instead of aggregated joints or limbs, GLMMs are capable of revealing associations that are not evident in other frameworks. DISCUSSION Currently widely available in statistical analysis software, GLMMs can accommodate a wide array of data distributions, account for hierarchical correlations, and return estimates of DJD prevalence within individuals and skeletal locations that are unbiased by the effect of covariates. This gives clear advantages for the analysis of bioarchaeological datasets which can lead to more robust and comparable analyses across populations.
Collapse
Affiliation(s)
| | | | - Belen Lopez
- Department of Biology of Organisms and Systems, University of Oviedo, Asturias, Spain
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
42
|
van Buuren MMA, Arden NK, Bierma-Zeinstra SMA, Bramer WM, Casartelli NC, Felson DT, Jones G, Lane NE, Lindner C, Maffiuletti NA, van Meurs JBJ, Nelson AE, Nevitt MC, Valenzuela PL, Verhaar JAN, Weinans H, Agricola R. Statistical shape modeling of the hip and the association with hip osteoarthritis: a systematic review. Osteoarthritis Cartilage 2021; 29:607-618. [PMID: 33338641 DOI: 10.1016/j.joca.2020.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/30/2020] [Accepted: 12/08/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To summarize available evidence on the association between hip shape as quantified by statistical shape modeling (SSM) and the incidence or progression of hip osteoarthritis. DESIGN We conducted a systematic search of five electronic databases, based on a registered protocol (available: PROSPERO CRD42020145411). Articles presenting original data on the longitudinal relationship between radiographic hip shape (quantified by SSM) and hip OA were eligible. Quantitative meta-analysis was precluded because of the use of different SSM models across studies. We used the Newcastle-Ottawa Scale (NOS) for risk of bias assessment. RESULTS Nine studies (6,483 hips analyzed with SSM) were included in this review. The SSM models used to describe hip shape ranged from 16 points on the femoral head to 85 points on the proximal femur and hemipelvis. Multiple hip shape features and combinations thereof were associated with incident or progressive hip OA. Shape variants that seemed to be consistently associated with hip OA across studies were acetabular dysplasia, cam morphology, and deviations in acetabular version (either excessive anteversion or retroversion). CONCLUSIONS Various radiographic, SSM-defined hip shape features are associated with hip OA. Some hip shape features only seem to increase the risk for hip OA when combined together. The heterogeneity of the used SSM models across studies precludes the estimation of pooled effect sizes. Further studies using the same SSM model and definition of hip OA are needed to allow for the comparison of outcomes across studies, and to validate the found associations.
Collapse
Affiliation(s)
- M M A van Buuren
- Department of Orthopedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - N K Arden
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK; NIHR Musculoskeletal Biomedical Research Unit, Arthritis Research UK Centre for Sport, Exercise, and Osteoarthritis, University of Oxford, Oxford, UK
| | - S M A Bierma-Zeinstra
- Department of Orthopedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of General Practice and Department of Orthopedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - W M Bramer
- Medical Library, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - N C Casartelli
- Human Performance Lab, Schulthess Clinic, Zürich, Switzerland; Laboratory of Exercise and Health, ETH Zürich, Schwerzenbach, Switzerland
| | - D T Felson
- Centre for Epidemiology Versus Arthritis, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK; Department of Rheumatology, Boston University School of Medicine, Boston, MA, USA
| | - G Jones
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - N E Lane
- Department of Medicine, University of California, Davis, CA, USA
| | - C Lindner
- Division of Informatics, Imaging & Data Sciences, University of Manchester, Manchester, UK
| | - N A Maffiuletti
- Human Performance Lab, Schulthess Clinic, Zürich, Switzerland
| | - J B J van Meurs
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - A E Nelson
- Thurston Arthritis Research Center and Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - M C Nevitt
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - P L Valenzuela
- Department of Systems Biology, University of Alcalá, Madrid, Spain
| | - J A N Verhaar
- Department of Orthopedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - H Weinans
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Biomechanical Engineering, Delft University of Technology, Delft, the Netherlands
| | - R Agricola
- Department of Orthopedics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
43
|
Grässel S, Zaucke F, Madry H. Osteoarthritis: Novel Molecular Mechanisms Increase Our Understanding of the Disease Pathology. J Clin Med 2021; 10:jcm10091938. [PMID: 33946429 PMCID: PMC8125020 DOI: 10.3390/jcm10091938] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Although osteoarthritis (OA) is the most common musculoskeletal condition that causes significant health and social problems worldwide, its exact etiology is still unclear. With an aging and increasingly obese population, OA is becoming even more prevalent than in previous decades. Up to 35% of the world’s population over 60 years of age suffers from symptomatic (painful, disabling) OA. The disease poses a tremendous economic burden on the health-care system and society for diagnosis, treatment, sick leave, rehabilitation, and early retirement. Most patients also experience sleep disturbances, reduced capability for exercising, lifting, and walking and are less capable of working, and maintaining an independent lifestyle. For patients, the major problem is disability, resulting from joint tissue destruction and pain. So far, there is no therapy available that effectively arrests structural deterioration of cartilage and bone or is able to successfully reverse any of the existing structural defects. Here, we elucidate novel concepts and hypotheses regarding disease progression and pathology, which are relevant for understanding underlying the molecular mechanisms as a prerequisite for future therapeutic approaches. Emphasis is placed on topographical modeling of the disease, the role of proteases and cytokines in OA, and the impact of the peripheral nervous system and its neuropeptides.
Collapse
Affiliation(s)
- Susanne Grässel
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), Bio Park 1, University of Regensburg, 93053 Regensburg, Germany
- Correspondence:
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital Friedrichsheim, 60528 Frankfurt am Main, Germany;
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, 66421 Homburg, Germany;
| |
Collapse
|
44
|
Baer K, Kieser S, Schon B, Rajendran K, Ten Harkel T, Ramyar M, Löbker C, Bateman C, Butler A, Raja A, Hooper G, Anderson N, Woodfield T. Spectral CT imaging of human osteoarthritic cartilage via quantitative assessment of glycosaminoglycan content using multiple contrast agents. APL Bioeng 2021; 5:026101. [PMID: 33834156 PMCID: PMC8018795 DOI: 10.1063/5.0035312] [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: 10/27/2020] [Accepted: 02/22/2021] [Indexed: 01/06/2023] Open
Abstract
Detection of early osteoarthritis to stabilize or reverse the damage to articular cartilage would improve patient function, reduce disability, and limit the need for joint replacement. In this study, we investigated nondestructive photon-processing spectral computed tomography (CT) for the quantitative measurement of the glycosaminoglycan (GAG) content compared to destructive histological and biochemical assay techniques in normal and osteoarthritic tissues. Cartilage-bone cores from healthy bovine stifles were incubated in 50% ioxaglate (Hexabrix®) or 100% gadobenate dimeglumine (MultiHance®). A photon-processing spectral CT (MARS) scanner with a CdTe-Medipix3RX detector imaged samples. Calibration phantoms of ioxaglate and gadobenate dimeglumine were used to determine iodine and gadolinium concentrations from photon-processing spectral CT images to correlate with the GAG content measured using a dimethylmethylene blue assay. The zonal distribution of GAG was compared between photon-processing spectral CT images and histological sections. Furthermore, discrimination and quantification of GAG in osteoarthritic human tibial plateau tissue using the same contrast agents were demonstrated. Contrast agent concentrations were inversely related to the GAG content. The GAG concentration increased from 25 μg/ml (85 mg/ml iodine or 43 mg/ml gadolinium) in the superficial layer to 75 μg/ml (65 mg/ml iodine or 37 mg/ml gadolinium) in the deep layer of healthy bovine cartilage. Deep zone articular cartilage could be distinguished from subchondral bone by utilizing the material decomposition technique. Photon-processing spectral CT images correlated with histological sections in healthy and osteoarthritic tissues. Post-imaging material decomposition was able to quantify the GAG content and distribution throughout healthy and osteoarthritic cartilage using Hexabrix® and MultiHance® while differentiating the underlying subchondral bone.
Collapse
Affiliation(s)
| | - Sandra Kieser
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE), Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch 8011, New Zealand
| | | | | | | | - Mohsen Ramyar
- Department of Radiology, University of Otago Christchurch, Christchurch 8011, New Zealand
| | | | - Christopher Bateman
- Department of Radiology, University of Otago Christchurch, Christchurch 8011, New Zealand
| | | | | | | | - Nigel Anderson
- Department of Radiology, University of Otago Christchurch, Christchurch 8011, New Zealand
| | | |
Collapse
|
45
|
Rezende MU, Brito NLR, Farias FES, Silva CAC, Cernigoy CHA, Rodrigues da Silva JM, Moreira MM, Santana OFN, Hissadomi MI, Frucchi R, Pasqualin T, Campos GC, Pailo AF, Ocampos GP, Camargo OP. Improved function and strength in patients with knee osteoarthritis as a result of adding a two-day educational program to usual care. Prospective randomized trial. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100137. [DOI: 10.1016/j.ocarto.2020.100137] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 11/28/2022] Open
|
46
|
Kraeutler MJ, Houck DA, Garabekyan T, Miller SL, Dragoo JL, Mei-Dan O. Comparing Intra-articular Injections of Leukocyte-Poor Platelet-Rich Plasma Versus Low-Molecular Weight Hyaluronic Acid for the Treatment of Symptomatic Osteoarthritis of the Hip: A Double-Blind, Randomized Pilot Study. Orthop J Sports Med 2021; 9:2325967120969210. [PMID: 33786329 PMCID: PMC7934058 DOI: 10.1177/2325967120969210] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 07/02/2020] [Indexed: 11/16/2022] Open
Abstract
Background: Hyaluronic acid (HA) and leukocyte-poor platelet-rich plasma (LP-PRP) are 2 nonoperative treatment options that have been studied in patients with hip osteoarthritis (OA). Purpose: To compare the efficacy of intra-articular injections of low–molecular weight (LMW) HA and LP-PRP in patients with hip OA. Study Design: Randomized controlled trial; Level of evidence, 1. Methods: A total of 34 patients (36 hips) presenting with signs of hip OA were randomized to receive 3 blinded, weekly intra-articular injections of either LP-PRP or LMW-HA. Patients were prospectively evaluated before injections and at 6 weeks and then at 3, 6, 12, and 24 months. The primary outcome, conversion to total hip arthroplasty (THA) or a hip resurfacing procedure, was analyzed along with secondary outcomes including the Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) score and hip range of motion. Results: The final analysis included 33 hips (mean Kellgren-Lawrence grade, 2.73) (LMW-HA: n = 14; LP-PRP: n = 19) in 31 patients (18 male; mean age, 53.8 years). Significantly more patients converted to THA or a hip resurfacing procedure in the LMW-HA group (7/14; 50.0%) (mean, 1.3 years after first injection) than the LP-PRP group (3/19; 15.8%) (mean, 0.73 years after first injection) (P = .035). There was no significant improvement or decline in any outcome scores within the LMW-HA group from before injections to 6 weeks or 3, 6, and 12 months. For the LP-PRP group, WOMAC overall (P = .032), joint (P = .030), and function scores (P = .025) significantly improved from before injections to 6 weeks, and WOMAC joint scores significantly improved from before injections to 6 months (P = .036). When comparing the difference between groups in internal rotation at 90° of hip flexion from before injections to 6 months, the LP-PRP group demonstrated a mean 5.0° improvement, while the LMW-HA group showed a mean 1.5° decrease (P = .028). Conclusion: Intra-articular hip injections of LP-PRP in patients with hip OA resulted in an improvement in WOMAC scores and hip internal rotation at 6 months and delayed the need for THA or a hip resurfacing procedure compared with treatment with LMW-HA. A longer follow-up is necessary to further compare the effects of LP-PRP and LMW-HA injections in patients with hip OA. Registration: NCT01920152 (ClinicalTrials.gov identifier).
Collapse
Affiliation(s)
- Matthew J Kraeutler
- Department of Orthopedic Surgery, St Joseph's University Medical Center, Paterson, New Jersey, USA
| | - Darby A Houck
- Department of Orthopedics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Shannon L Miller
- Department of Orthopedics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jason L Dragoo
- Department of Orthopedics, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Omer Mei-Dan
- Department of Orthopedics, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
47
|
Gielis WP, de Jong PA, Bartstra JW, Foppen W, Spiering W, den Harder AM. Osteoarthritis in Pseudoxanthoma Elasticum Patients: An Explorative Imaging Study. J Clin Med 2020; 9:E3898. [PMID: 33271791 PMCID: PMC7760162 DOI: 10.3390/jcm9123898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 11/16/2022] Open
Abstract
Pseudoxanthoma elasticum (PXE) is a systemic disease affecting the skin, eyes, and cardiovascular system of patients. Cardiovascular disease is associated with osteoarthritis (OA), which is the most common cause of joint pain. There is a lack of systematic investigations on joint manifestations in PXE in the literature. In this explorative study, we aimed to investigate whether patients with PXE are more at risk for developing osseous signs of OA. Patients with PXE and hospital controls with whole-body low-dose CT examinations available were included. OA was assessed using the OsteoArthritis Computed Tomography (OACT)-score, which is a 4-point Likert scale, in the acromioclavicular (AC), glenohumeral (GH), facet, hip, knee, and ankle joints. Additionally, intervertebral disc degeneration was scored. Data were analyzed using ordinal logistic regression adjusted for age, body mass index (BMI), and smoking status. In total, 106 PXE patients (age 56 (48-64), 42% males, BMI 25.3 (22.7-28.2)) and 87 hospital controls (age 55 (43-67), 46% males, BMI 26.0 (22.5-29.2)) were included. PXE patients were more likely to have a higher OA score for the AC joints (OR 2.00 (1.12-3.61)), tibiofemoral joint (OR 2.63 (1.40-5.07)), and patellofemoral joint (2.22 (1.18-4.24)). For the other joints, the prevalence and severity of OA did not differ significantly. This study suggests that patients with PXE are more likely to have structural OA of the knee and AC joints, which needs clinical confirmation in larger groups and further investigation into the mechanism.
Collapse
Affiliation(s)
- Willem Paul Gielis
- University Medical Center Utrecht, Department of Orthopaedics, Utrecht University, 3584 CX Utrecht, The Netherlands
- University Medical Center Utrecht, Department of Radiology, Utrecht University, 3584 CX Utrecht, The Netherlands; (P.A.d.J.); (J.W.B.); (W.F.); (A.M.d.H.)
| | - Pim A. de Jong
- University Medical Center Utrecht, Department of Radiology, Utrecht University, 3584 CX Utrecht, The Netherlands; (P.A.d.J.); (J.W.B.); (W.F.); (A.M.d.H.)
| | - Jonas W. Bartstra
- University Medical Center Utrecht, Department of Radiology, Utrecht University, 3584 CX Utrecht, The Netherlands; (P.A.d.J.); (J.W.B.); (W.F.); (A.M.d.H.)
| | - Wouter Foppen
- University Medical Center Utrecht, Department of Radiology, Utrecht University, 3584 CX Utrecht, The Netherlands; (P.A.d.J.); (J.W.B.); (W.F.); (A.M.d.H.)
| | - Wilko Spiering
- University Medical Center Utrecht, Department of Vascular Medicine, Utrecht University, 3584 CX Utrecht, The Netherlands;
| | - Annemarie M. den Harder
- University Medical Center Utrecht, Department of Radiology, Utrecht University, 3584 CX Utrecht, The Netherlands; (P.A.d.J.); (J.W.B.); (W.F.); (A.M.d.H.)
| |
Collapse
|
48
|
Yang H, Zhou X, Xu D, Chen G. The IL-6 rs12700386 polymorphism is associated with an increased risk of developing osteoarthritis in the knee in the Chinese Han population: a case-control study. BMC MEDICAL GENETICS 2020; 21:199. [PMID: 33036557 PMCID: PMC7547410 DOI: 10.1186/s12881-020-01139-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/04/2020] [Indexed: 11/10/2022]
Abstract
Background This case-control study aims to examine the association between the Interleukin-6 (IL-6) rs12700386 polymorphism and the increased risk of developing osteoarthritis (OA) in the knee in the Chinese Han population. Methods We extracted DNA from 763 subjects (352 OA patients and 411 healthy controls). The relative expression levels of IL-6 in blood samples of patients with knee OA was determined by quantitative reverse transcription PCR (qRT-PCR) and polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) was used for genotyping the IL-6 gene polymorphism. Results We found that the IL-6 polymorphism rs12700386 enhanced patient susceptibility to developing knee OA. Based on a subgroup analysis, the risk of developing knee OA was elevated in smokers, drinkers, and subjects ≥55 years old or with BMI ≥ 25 kg/m2. The combination of smoking, drinking, and having the rs12700386 genotype led to an increase in the risk of developing knee OA, indicating that an underlying interaction between gene and environment exists. The rs12700386 genotype was found to be correlated with an increase in IL-6 expression. We also found that IL-6 levels were significantly higher in the CC genotype compared to the GG genotype carriers in OA patients. Conclusion These data suggest that the rs12700386 polymorphism in the IL-6 gene leads to an increase in the risk of knee OA in Chinese Han individuals.
Collapse
Affiliation(s)
- Hui Yang
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Xindie Zhou
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China
| | - Dongmei Xu
- Department of Orthopedics, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, 213000, China.
| | - Gang Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China.
| |
Collapse
|
49
|
Shah K, Furniss D, Collins GS, Peirce N, Arden NK, Filbay SR. Cricket related hand injury is associated with increased odds of hand pain and osteoarthritis. Sci Rep 2020; 10:16775. [PMID: 33033307 PMCID: PMC7545096 DOI: 10.1038/s41598-020-73586-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 09/10/2020] [Indexed: 11/09/2022] Open
Abstract
Radiographic osteoarthritis (OA) is most prevalent in the hand. The association of hand injury with pain or OA is unclear. The objective was to describe the relationship between hand injury and ipsilateral pain and OA in cricketers. Data from former and current cricketers aged ≥ 30 years was used. Data included history of cricket-related hand/finger injury leading to > 4 weeks of reduced exercise, hand/finger joint pain on most days of the last month, self-reported history of physician-diagnosed hand/finger OA. Logistic regression assessed the relationship between injury with hand pain (in former cricketers) and with OA (in all cricketers), adjusted for age, seasons played, playing standard. Of 1893 participants (844 former cricketers), 16.9% reported hand pain, 4.3% reported OA. A history of hand injury increased the odds of hand pain (OR (95% CI) 2.2, 1.4 to 3.6). A history of hand injury also had increased odds of hand OA (3.1, 2.1 to 4.7). Cricket-related hand injury was related to an increased odds of hand pain and OA. This highlights the importance of hand injury prevention strategies within cricket. The high prevalence of hand pain is concerning, and further research is needed to determine the impacts of hand pain.
Collapse
Affiliation(s)
- Karishma Shah
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Old Road, Oxford, OX3 7LD, UK
| | - Dominic Furniss
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Old Road, Oxford, OX3 7LD, UK
| | - Gary S Collins
- Centre for Statistics in Medicine, University of Oxford, Oxford, UK
| | - Nick Peirce
- England and Wales Cricket Board, and National Centre for Sports and Exercise Medicine and National Cricket Performance Centre, Loughborough University, Loughborough, UK
| | - Nigel K Arden
- Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Stephanie R Filbay
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Old Road, Oxford, OX3 7LD, UK.
- Centre for Sport, Exercise and Osteoarthritis Research Versus Arthritis, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK.
- Centre for Health Exercise and Sports Medicine, Department of Physiotherapy, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
50
|
Götze M, Hagmann S. [Hip joint contractures, leg length discrepancies, and spinopelvic alignment: impact on the development of osteoarthritis of the hip?]. DER ORTHOPADE 2020; 49:877-882. [PMID: 32902656 DOI: 10.1007/s00132-020-03977-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hip joint problems are common in every-day orthopedic practice. At the age of 60, approximately 5% of the German population suffer from symptomatic osteoarthritis of the hip. One of the main trigger is the morphological abnormality of the joint. Causes may also be overloading of the joint, developmental dysplasia of the hip with reduced coverage, as well as femoral or acetabular malpositioning, for example. The impact of hip joint contractures, leg length discrepancies, and the spinopelvic alignment and their prophylactic treatment in childhood, adolescence and early adulthood are discussed here.
Collapse
Affiliation(s)
- Marco Götze
- Zentrum für Orthopädie, Unfallchirurgie und Paraplegiologie, Sektion Kinderorthopädie, Neuroorthopädie & Fußchirurgie, Universitätsklinikum Heidelberg, Schlierbacher Landstr. 200a, 69118, Heidelberg, Deutschland.
| | - Sébastien Hagmann
- Zentrum für Orthopädie, Unfallchirurgie und Paraplegiologie, Sektion Kinderorthopädie, Neuroorthopädie & Fußchirurgie, Universitätsklinikum Heidelberg, Schlierbacher Landstr. 200a, 69118, Heidelberg, Deutschland
| |
Collapse
|