1
|
Struijk C, Korpershoek J, Lydon KL, Verdonk P, Michielsen J, Krych AJ, Vonk LA, Saris DBF. Identification and culture of meniscons, meniscus cells with their pericellular matrix. Cytotherapy 2025; 27:98-106. [PMID: 39373674 DOI: 10.1016/j.jcyt.2024.08.006] [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: 05/20/2024] [Revised: 08/11/2024] [Accepted: 08/15/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND AIMS Meniscus injury is highly debilitating and often results in osteoarthritis. Treatment is generally symptomatic; no regenerative treatments are available. "Chondrons," articular chondrocytes with preserved pericellular matrix, produce more hyaline cartilage extracellular matrix and improve cartilage repair. If meniscons exist in the meniscus and have similar therapeutic potential as chondrons, employing these cells has potential for meniscus cell therapy and tissue engineering. In this study, we isolated and cultured "meniscons," meniscus cells surrounded by their native pericellular matrix, and investigated cell behavior in culture compared with chondrons. METHODS Human meniscons were enzymatically isolated from osteoarthritic menisci and cultured up to 28 days in fibrin glue. Freshly isolated meniscons and chondrons were analyzed by histology and transmission electron microscopy. We used 5-([4,6-dichlorotriazin-2-yl]amino)fluorescein hydrochloride labeling and type VI collagen immunohistochemistry to image pericellular matrix after 0 and 28 days of culture. Gene expression was quantified using real-time polymerase chain reaction and DNA content and proteoglycan production were analyzed using biochemical assays. RESULTS Meniscons were successfully isolated from human meniscus tissue. The pericellular matrix of meniscons and chondrons was preserved during 28 days of culture. Meniscons and chondrons had similar cell proliferation and proteoglycan production. Meniscons and chondrons expressed similar levels of collagen type I alpha 1 chain, whereas collagen type II alpha 1 chain and aggrecan expression was lower in the meniscon population. CONCLUSIONS Freshly isolated meniscons and meniscons cultured for 28 days share similarities with chondrons with regard to cell proliferation, morphology and biochemical activity. Rapid isolation of meniscons (45 min) demonstrates potential for one-stage meniscus regeneration and repair, which should be confirmed in vivo.
Collapse
Affiliation(s)
- Caroline Struijk
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA; Department of Orthopedic Surgery, Antwerp University, Antwerp, Belgium
| | - Jasmijn Korpershoek
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA; Department of Orthopaedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Katherine L Lydon
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter Verdonk
- Department of Orthopedic Surgery, Antwerp University, Antwerp, Belgium; Orthoca, Antwerp, Belgium
| | - Jozef Michielsen
- Department of Orthopedic Surgery, Antwerp University, Antwerp, Belgium
| | - Aaron J Krych
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Lucienne A Vonk
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Daniel B F Saris
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA; Department of Orthopaedics, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
2
|
Fei K, Andress BD, Kelly AM, Chasse DAD, McNulty AL. Meniscus gene expression profiling of inner and outer zone meniscus tissue compared to cartilage and passaged monolayer meniscus cells. Sci Rep 2024; 14:27423. [PMID: 39521910 PMCID: PMC11550462 DOI: 10.1038/s41598-024-78580-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Meniscus injuries are common and while surgical strategies have improved, there is a need for alternative therapeutics to improve long-term outcomes and prevent post-traumatic osteoarthritis. Current research efforts in regenerative therapies and tissue engineering are hindered by a lack of understanding of meniscus cell biology and a poorly defined meniscus cell phenotype. This study utilized bulk RNA-sequencing to identify unique and overlapping transcriptomic profiles in cartilage, inner and outer zone meniscus tissue, and passaged inner and outer zone meniscus cells. The greatest transcriptomic differences were identified when comparing meniscus tissue to passaged monolayer cells (> 4,600 differentially expressed genes (DEGs)) and meniscus tissue to cartilage (> 3,100 DEGs). While zonal differences exist within the meniscus tissue (205 DEGs between inner and outer zone meniscus tissue), meniscus resident cells are more similar to each other than to either cartilage or passaged monolayer meniscus cells. Additionally, we identified and validated LUM, PRRX1, and SNTB1 as potential markers for meniscus tissue and ACTA2, TAGLN, SFRP2, and FSTL1 as novel markers for meniscus cell dedifferentiation. Our data contribute significantly to the current characterization of meniscus cells and provide an important foundation for future work in meniscus cell biology, regenerative medicine, and tissue engineering.
Collapse
Affiliation(s)
- Kaileen Fei
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Benjamin D Andress
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
- Department of Pathology, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - A'nna M Kelly
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
- Department of Biomedical Engineering, Duke University, DUMC Box 3093, Durham, NC, 27710, USA
| | - Dawn A D Chasse
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA
| | - Amy L McNulty
- Department of Orthopaedic Surgery, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA.
- Department of Pathology, Duke University School of Medicine, DUMC Box 3093, Durham, NC, 27710, USA.
- Department of Biomedical Engineering, Duke University, DUMC Box 3093, Durham, NC, 27710, USA.
| |
Collapse
|
3
|
Iacobescu GL, Iacobescu L, Popa MIG, Covache-Busuioc RA, Corlatescu AD, Cirstoiu C. Genomic Determinants of Knee Joint Biomechanics: An Exploration into the Molecular Basis of Locomotor Function, a Narrative Review. Curr Issues Mol Biol 2024; 46:1237-1258. [PMID: 38392197 PMCID: PMC10888373 DOI: 10.3390/cimb46020079] [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: 12/30/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
In recent years, the nexus between genetics and biomechanics has garnered significant attention, elucidating the role of genomic determinants in shaping the biomechanical attributes of human joints, specifically the knee. This review seeks to provide a comprehensive exploration of the molecular basis underlying knee joint locomotor function. Leveraging advancements in genomic sequencing, we identified specific genetic markers and polymorphisms tied to key biomechanical features of the knee, such as ligament elasticity, meniscal resilience, and cartilage health. Particular attention was devoted to collagen genes like COL1A1 and COL5A1 and their influence on ligamentous strength and injury susceptibility. We further investigated the genetic underpinnings of knee osteoarthritis onset and progression, as well as the potential for personalized rehabilitation strategies tailored to an individual's genetic profile. We reviewed the impact of genetic factors on knee biomechanics and highlighted the importance of personalized orthopedic interventions. The results hold significant implications for injury prevention, treatment optimization, and the future of regenerative medicine, targeting not only knee joint health but joint health in general.
Collapse
Affiliation(s)
- Georgian-Longin Iacobescu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Loredana Iacobescu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Mihnea Ioan Gabriel Popa
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| | - Razvan-Adrian Covache-Busuioc
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Antonio-Daniel Corlatescu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Catalin Cirstoiu
- Orthopaedics and Traumatology Department, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- University Emergency Hospital, 050098 Bucharest, Romania
| |
Collapse
|
4
|
Yan W, Wu Y, Zhao F, Dai R, Zhou Y, Liu D, Cheng J, Hu X, Ao Y. Anti-Apoptosis Therapy for Meniscal Avascular Zone Repair: A Proof-of-Concept Study in a Lapine Model. Bioengineering (Basel) 2023; 10:1422. [PMID: 38136013 PMCID: PMC10740472 DOI: 10.3390/bioengineering10121422] [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: 10/18/2023] [Revised: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
In the present study, 24 rabbits were firstly used to evaluate the apoptosis index and matrix degeneration after untreated adult meniscal tears. Vertical tears (0.25 cm in length) were prepared in the avascular zone of the anterior horn. Specimens were harvested at 1, 3, 6, 12 weeks postoperatively. The apoptosis index around tear sites stayed at a high level throughout the whole follow-up period. The depletion of glycosaminoglycans (GAG) and aggrecan at the tear site was observed, while the deposition of COL I and COL II was not affected, even at the last follow-up of 12 weeks after operation. The expression of SOX9 decreased significantly; no cellularity was observed at the wound interface at all timepoints. Secondly, another 20 rabbits were included to evaluate the effects of anti-apoptosis therapy on rescuing meniscal cells and enhancing meniscus repair. Longitudinal vertical tears (0.5 cm in length) were made in the meniscal avascular body. Tears were repaired by the inside-out suture technique, or repaired with sutures in addition to fibrin gel and blank silica nanoparticles, or silica nanoparticles encapsulating apoptosis inhibitors (z-vad-fmk). Samples were harvested at 12 months postoperatively. We found the locally administered z-vad-fmk agent at the wound interface significantly alleviated meniscal cell apoptosis and matrix degradation, and enhanced meniscal repair in the avascular zone at 12 months after operation. Thus, local administration of caspase inhibitors (z-vad-fmk) is a promising therapeutic strategy for alleviating meniscal cell loss and enhancing meniscal repair after adult meniscal tears in the avascular zone.
Collapse
Affiliation(s)
- Wenqiang Yan
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (W.Y.); (Y.W.); (F.Z.); (R.D.); (Y.Z.); (D.L.); (J.C.)
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Yue Wu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (W.Y.); (Y.W.); (F.Z.); (R.D.); (Y.Z.); (D.L.); (J.C.)
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Fengyuan Zhao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (W.Y.); (Y.W.); (F.Z.); (R.D.); (Y.Z.); (D.L.); (J.C.)
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Ruilan Dai
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (W.Y.); (Y.W.); (F.Z.); (R.D.); (Y.Z.); (D.L.); (J.C.)
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Yunan Zhou
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (W.Y.); (Y.W.); (F.Z.); (R.D.); (Y.Z.); (D.L.); (J.C.)
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Dingge Liu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (W.Y.); (Y.W.); (F.Z.); (R.D.); (Y.Z.); (D.L.); (J.C.)
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Jin Cheng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (W.Y.); (Y.W.); (F.Z.); (R.D.); (Y.Z.); (D.L.); (J.C.)
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (W.Y.); (Y.W.); (F.Z.); (R.D.); (Y.Z.); (D.L.); (J.C.)
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| | - Yingfang Ao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing 100191, China; (W.Y.); (Y.W.); (F.Z.); (R.D.); (Y.Z.); (D.L.); (J.C.)
- Beijing Key Laboratory of Sports Injuries, Beijing 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing 100191, China
| |
Collapse
|
5
|
Li Y, Li H, Wang L, Xie W, Yuan D, Wen Z, Zhang T, Lai J, Xiong Z, Shan Y, Jiang W. The p65-LOC727924-miR-26a/KPNA3-p65 regulatory loop mediates vasoactive intestinal peptide effects on osteoarthritis chondrocytes. Int Immunopharmacol 2023; 122:110518. [PMID: 37392568 DOI: 10.1016/j.intimp.2023.110518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/10/2023] [Accepted: 06/13/2023] [Indexed: 07/03/2023]
Abstract
Loss and dysfunction of articular chondrocytes, which disrupt the homeostasis of extracellular matrix formation and breakdown, promote the onset of osteoarthritis (OA). Targeting inflammatory pathways is an important therapeutic strategy for OA. Vasoactive intestinal peptide (VIP) is an immunosuppressive neuropeptide with potent anti-inflammatory effects; however, its role and mechanism in OA remain unclear. In this study, microarray expression profiling from the Gene Expression Omnibus database and integrative bioinformatics analyses were performed to identify differentially expressed lncRNAs in OA samples. qRT-PCR validation of the top ten different expressed lncRNAs indicated that the expression level of intergenic non-protein coding RNA 2203 (LINC02203, also named LOC727924) was the highest in OA cartilage compared to normal cartilage. Hence, the LOC727924 function was further investigated. LOC727924 was upregulated in OA chondrocytes, with a dominant sub-localization in the cytoplasm. In OA chondrocytes, LOC727924 knockdown boosted cell viability, suppressed cell apoptosis, reactive oxygen species (ROS) accumulation, increased aggrecan and collagen II, decreased matrix metallopeptidase (MMP)-3/13 and ADAM metallopeptidase with thrombospondin type 1 motif (ADAMTS)-4/5 levels, and reduced the levels of tumor necrosis factor alpha (TNF-α), interleukin 1 beta (IL-1β), and interleukin 6 (IL-6). LOC727924 could interact with the microRNA 26a (miR-26a)/ karyopherin subunit alpha 3 (KPNA3) axis by competitively targeting miR-26a for KPNA3 binding, therefore down-regulating miR-26a and upregulating KPNA3; in OA chondrocytes, miR-26a inhibition partially abolished LOC727924 knockdown effects on chondrocytes. miR-26a inhibited the nuclear translocation of p65 through targeting KPNA3 and p65 transcriptionally activated LOC727924, forming a p65-LOC727924-miR-26a/KPNA3-p65 regulatory loop to modulate OA chondrocyte phenotypes. In vitro, VIP improved OA chondrocyte proliferation and functions, down-regulated LOC727924, KPNA3, and p65 expression, and upregulated miR-26a expression; in vivo, VIP ameliorated destabilization of the medial meniscus (DMM)-induced damages on the mouse knee joint, down-regulated KPNA3, inhibited the nuclear translocation of p65. In conclusion, the p65-LOC727924-miR-26a/KPNA3-p65 regulatory loop modulates OA chondrocyte apoptosis, ROS accumulation, extracellular matrix (ECM) deposition, and inflammatory response in vitro and OA development in vivo, being one of the mechanisms mediating VIP ameliorating OA.
Collapse
Affiliation(s)
- Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Lijie Wang
- Department of Bone and Joint, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Wenqing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Dongliang Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China; Xiangya School of Medicine, Central South University, Changsha 410083, Hunan, China
| | - Zeqin Wen
- Xiangya School of Medicine, Central South University, Changsha 410083, Hunan, China
| | - Tiancheng Zhang
- Department of Bone and Joint, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Jieyu Lai
- Xiangya School of Medicine, Central South University, Changsha 410083, Hunan, China
| | - Zixuan Xiong
- Xiangya School of Medicine, Central South University, Changsha 410083, Hunan, China
| | - Yunhan Shan
- Xiangya School of Medicine, Central South University, Changsha 410083, Hunan, China
| | - Wei Jiang
- Department of Bone and Joint, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China.
| |
Collapse
|
6
|
Aleem AW, Rai MF, Cai L, Brophy RH. Gene Expression in Glenoid Articular Cartilage Varies Across Acute Instability, Chronic Instability, and Osteoarthritis. J Bone Joint Surg Am 2023:00004623-990000000-00776. [PMID: 37011069 DOI: 10.2106/jbjs.22.01124] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
BACKGROUND Shoulder instability is a common pathology associated with an elevated risk of osteoarthritis (OA). Little is known about gene expression in the cartilage of the glenohumeral joint after dislocation events, particularly as it relates to the risk of posttraumatic OA. This study tested the hypothesis that gene expression in glenoid cartilage varies among acute instability (<3 dislocations), chronic instability (≥3 dislocations), and OA. METHODS Articular cartilage was collected from the anteroinferior glenoid of consenting patients undergoing shoulder stabilization surgery (n = 17) or total shoulder arthroplasty (n = 16). Digital quantitative polymerase chain reaction was used to assess the relative expression of 57 genes (36 genes from OA risk allele studies, 21 genes from differential expression studies), comparing (1) OA versus instability (acute and chronic combined), (2) acute versus chronic instability, (3) OA versus acute instability, and (4) OA versus chronic instability. RESULTS The expression of 11 genes from OA risk allele studies and 9 genes from differential expression studies was significantly different between cartilage from patients with instability and those with OA. Pro-inflammatory genes from differential expression studies and genes from OA risk allele studies were more highly expressed in cartilage in the OA group compared with the instability group, which expressed higher levels of extracellular matrix and pro-anabolic genes. The expression of 14 genes from OA risk allele studies and 4 genes from differential expression studies, including pro-inflammatory genes, anti-anabolic genes, and multiple genes from OA risk allele studies, was higher in the acute instability group compared with the chronic instability group. Cartilage in the OA group displayed higher expression of CCL3, CHST11, GPR22, PRKAR2B, and PTGS2 than cartilage in the group with acute or chronic instability. Whereas cartilage in both the acute and chronic instability groups had higher expression of collagen genes, cartilage in the OA group had expression of a subset of genes from OA risk allele studies or from differential expression studies that was lower than in the acute group and higher than in the chronic group. CONCLUSIONS Glenoid cartilage has an inflammatory and catabolic phenotype in shoulders with OA but an anabolic phenotype in shoulders with instability. Cartilage from shoulders with acute instability displayed greater (cellular) metabolic activity compared with shoulders with chronic instability. CLINICAL RELEVANCE This exploratory study identified genes of interest, such as CCL3, CHST11, GPR22, PRKAR2B, and PTGS2, that have elevated expression in osteoarthritic glenoid cartilage. These findings provide new biological insight into the relationship between shoulder instability and OA, which could lead to strategies to predict and potentially modify patients' risk of degenerative arthritis due to shoulder instability.
Collapse
Affiliation(s)
- Alexander W Aleem
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri
- Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Lei Cai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Robert H Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
7
|
Gouldin AG, Patel NK, Golladay GJ, Puetzer JL. Advanced glycation end-product accumulation differs by location and sex in aged osteoarthritic human menisci. Osteoarthritis Cartilage 2023; 31:363-373. [PMID: 36494052 PMCID: PMC10088070 DOI: 10.1016/j.joca.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/15/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVE There is a clear link between increasing age and meniscus degeneration, leading to increased injury, osteoarthritis (OA) progression, and often total knee replacement. Advanced glycation end-products (AGEs) are non-enzymatic crosslinks and adducts that accumulate in collagen with age, altering tissue mechanics and cell function, ultimately leading to increased injury and inflammation. AGEs, both fluorescent and non-fluorescent, play a central role in age-related degradation of tissues throughout the body; however, little is known about their role in meniscus degeneration. The objective of this study was to characterize changes in aged OA menisci, specifically evaluating zonal AGE accumulation, to gain a better understanding of changes that may lead to age-related meniscal degeneration. METHOD Deidentified human menisci (N = 48, 52-84 years old) were obtained from subjects undergoing total knee replacement. Changes in extracellular matrix (ECM) were assessed by gross morphology, confocal analysis, and biochemical assays. Deoxyribonucleic acid (DNA), glycosaminoglycan (GAG), collagen, and AGE accumulation were compared with patient age, zonal region, and patient sex. RESULTS There were minimal changes in DNA, GAG, and collagen concentration with age or zone. However, collagen fraying and AGEs increased with age, with more AGEs accumulating in the meniscal horns compared to the central body and in male menisci compared to females. CONCLUSIONS Overall, this work provides greater insights into regional changes that occur in human menisci with age and OA. These results suggest AGEs may play a role in the degeneration of the meniscus, with AGEs being a possible target to reduce age-related tears, degeneration, and OA progression.
Collapse
Affiliation(s)
- A G Gouldin
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States.
| | - N K Patel
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States.
| | - G J Golladay
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States.
| | - J L Puetzer
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, United States; Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
8
|
Monibi FA, Pannellini T, Croen B, Otero M, Warren R, Rodeo SA. Targeted transcriptomic analyses of RNA isolated from formalin-fixed and paraffin-embedded human menisci. J Orthop Res 2022; 40:1104-1112. [PMID: 34370349 PMCID: PMC8825887 DOI: 10.1002/jor.25153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/14/2021] [Accepted: 08/03/2021] [Indexed: 02/04/2023]
Abstract
Formalin-fixed and paraffin-embedded (FFPE) biospecimens are a valuable and widely-available resource for diagnostic and research applications. With biobanks of tissue samples available in many institutions, FFPE tissues could prove to be a valuable resource for translational orthopaedic research. The purpose of this study was to characterize the molecular profiles and degree of histologic degeneration on archival fragments of FFPE human menisci obtained during arthroscopic partial meniscectomy. We used FFPE menisci for multiplexed gene expression analysis using the NanoString nCounter® platform, and for histological assessment using a quantitative scoring system. In total, 17 archival specimens were utilized for integrated histologic and molecular analyses. The median patient age was 22 years (range: 14-62). We found that the genes with the highest normalized counts were those typically expressed in meniscal fibrocartilage. Gene expression differences were identified in patient cohorts based on age (≤40 years), including genes associated with the extracellular matrix and tissue repair. The majority of samples showed mild to moderate histologic degeneration. Based on these data, we conclude that FFPE human menisci can be effectively utilized for molecular evaluation following a storage time as long as 11 years. Statement of Clinical Significance: The integration of histological and transcriptomic analyses described in this study will be useful for future studies investigating the basis for biological classification of meniscus specimens in patients. Further exploration into the genes and pathways uncovered by this study may suggest targets for biomarker discovery and identify patients at greater risk for osteoarthritis once the meniscus is torn.
Collapse
Affiliation(s)
| | | | - Brett Croen
- Hospital for Special Surgery, NY, NY,Drexel University College of Medicine, Philadelphia, PA
| | | | | | | |
Collapse
|
9
|
Zaki S, Blaker CL, Little CB. OA foundations - experimental models of osteoarthritis. Osteoarthritis Cartilage 2022; 30:357-380. [PMID: 34536528 DOI: 10.1016/j.joca.2021.03.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis (OA) is increasingly recognised as a disease of diverse phenotypes with variable clinical presentation, progression, and response to therapeutic intervention. This same diversity is readily apparent in the many animal models of OA. However, model selection, study design, and interpretation of resultant findings, are not routinely done in the context of the target human (or veterinary) patient OA sub-population or phenotype. This review discusses the selection and use of animal models of OA in discovery and therapeutic-development research. Beyond evaluation of the different animal models on offer, this review suggests focussing the approach to OA-animal model selection on study objective(s), alignment of available models with OA-patient sub-types, and the resources available to achieve valid and translatable results. How this approach impacts model selection is discussed and an experimental design checklist for selecting the optimal model(s) is proposed. This approach should act as a guide to new researchers and a reminder to those already in the field, as to issues that need to be considered before embarking on in vivo pre-clinical research. The ultimate purpose of using an OA animal model is to provide the best possible evidence if, how, when and where a molecule, pathway, cell or process is important in clinical disease. By definition this requires both model and study outcomes to align with and be predictive of outcomes in patients. Keeping this at the forefront of research using pre-clinical OA models, will go a long way to improving the quality of evidence and its translational value.
Collapse
Affiliation(s)
- S Zaki
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Australia; Raymond Purves Bone and Joint Research Laboratory, Australia.
| | - C L Blaker
- Raymond Purves Bone and Joint Research Laboratory, Australia; Murray Maxwell Biomechanics Laboratory, The Kolling Institute, University of Sydney Faculty of Medicine and Health, At Royal North Shore Hospital, Australia.
| | - C B Little
- Raymond Purves Bone and Joint Research Laboratory, Australia.
| |
Collapse
|
10
|
Chen M, Zhou S, Shi H, Gu H, Wen Y, Chen L. Identification and validation of pivotal genes related to age-related meniscus degeneration based on gene expression profiling analysis and in vivo and in vitro models detection. BMC Med Genomics 2021; 14:237. [PMID: 34587952 PMCID: PMC8482591 DOI: 10.1186/s12920-021-01088-6] [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: 06/08/2021] [Accepted: 09/20/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The componential and structural change in the meniscus with aging would increase the tissue vulnerability of the meniscus, which would induce meniscus tearing. Here, we investigated the molecular mechanism of age-related meniscus degeneration with gene expression profiling analysis, and validate pivotal genes in vivo and in vitro models. METHODS The GSE45233 dataset, including 6 elderly meniscus samples and 6 younger meniscus samples, was downloaded from the Gene Expression Omnibus (GEO) database. To screen the differential expression of mRNAs and identify the miRNAs targeting hub genes, we completed a series of bioinformatics analyses, including functional and pathway enrichment, protein-protein interaction network, hub genes screening, and construction of a lncRNA-miRNA-mRNA network. Furthermore, crucial genes were examined in human senescent menisci, mouse senescent meniscus tissues and mouse meniscus cells stimulated by IL-1β. RESULTS In total, the most significant 4 hub genes (RRM2, AURKB, CDK1, and TIMP1) and 5 miRNAs (hsa-miR-6810-5p, hsa-miR-4676-5p, hsa-miR-6877-5p, hsa-miR-8085, and hsa-miR-6133) that regulated such 4 hub genes, were finally identified. Moreover, these hub genes were decreased in meniscus cells in vitro and meniscus tissues in vivo, which indicated that hub genes were related to meniscus senescence and could serve as potential biomarkers for age-related meniscus tearing. CONCLUSIONS In short, the integrated analysis of gene expression profile, co-expression network, and models detection identified pivotal genes, which elucidated the possible molecular basis underlying the senescence meniscus and also provided prognosis clues for early-onset age-related meniscus tearing.
Collapse
Affiliation(s)
- Ming Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Siqi Zhou
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.,Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Huasong Shi
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Hanwen Gu
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Yinxian Wen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China. .,Joint Disease Research Center, Wuhan University, Wuhan, 430071, China.
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedics Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China. .,Joint Disease Research Center, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
11
|
Lee KI, Gamini R, Olmer M, Ikuta Y, Hasei J, Baek J, Alvarez-Garcia O, Grogan SP, D'Lima DD, Asahara H, Su AI, Lotz MK. Mohawk is a transcription factor that promotes meniscus cell phenotype and tissue repair and reduces osteoarthritis severity. Sci Transl Med 2021; 12:12/567/eaan7967. [PMID: 33115953 DOI: 10.1126/scitranslmed.aan7967] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 02/06/2020] [Accepted: 09/21/2020] [Indexed: 12/13/2022]
Abstract
Meniscus tears are common knee injuries and a major osteoarthritis (OA) risk factor. Knowledge gaps that limit the development of therapies for meniscus injury and degeneration concern transcription factors that control the meniscus cell phenotype. Analysis of RNA sequencing data from 37 human tissues in the Genotype-Tissue Expression database and RNA sequencing data from meniscus and articular cartilage showed that transcription factor Mohawk (MKX) is highly enriched in meniscus. In human meniscus cells, MKX regulates the expression of meniscus marker genes, OA-related genes, and other transcription factors, including Scleraxis (SCX), SRY Box 5 (SOX5), and Runt domain-related transcription factor 2 (RUNX2). In mesenchymal stem cells (MSCs), the combination of adenoviral MKX (Ad-MKX) and transforming growth factor-β3 (TGF-β3) induced a meniscus cell phenotype. When Ad-MKX-transduced MSCs were seeded on TGF-β3-conjugated decellularized meniscus scaffold (DMS) and inserted into experimental tears in meniscus explants, they increased glycosaminoglycan content, extracellular matrix interconnectivity, cell infiltration into the DMS, and improved biomechanical properties. Ad-MKX injection into mouse knee joints with experimental OA induced by surgical destabilization of the meniscus suppressed meniscus and cartilage damage, reducing OA severity. Ad-MKX injection into human OA meniscus tissue explants corrected pathogenic gene expression. These results identify MKX as a previously unidentified key transcription factor that regulates the meniscus cell phenotype. The combination of Ad-MKX with TGF-β3 is effective for differentiation of MSCs to a meniscus cell phenotype and useful for meniscus repair. MKX is a promising therapeutic target for meniscus tissue engineering, repair, and prevention of OA.
Collapse
Affiliation(s)
- Kwang Il Lee
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Ramya Gamini
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Merissa Olmer
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Yasunari Ikuta
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Joe Hasei
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Jihye Baek
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA.,Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA 92037, USA
| | | | - Shawn P Grogan
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA.,Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA 92037, USA
| | - Darryl D D'Lima
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA.,Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA 92037, USA
| | - Hiroshi Asahara
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA
| | - Andrew I Su
- Department of Integrative, Structural and Computational Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Martin K Lotz
- Department of Molecular Medicine, Scripps Research, La Jolla, CA 92037, USA.
| |
Collapse
|
12
|
Abstract
There is a well-established historical observation that structural joint damage by plain X-ray correlates poorly with symptomatic disease in osteoarthritis (OA). This is often attributed to the inability to visualise soft-tissue pathology within the joint and the recognition of heterogeneous patient factors that drive central pain sensitisation. A major issue is the relative paucity of mechanistic studies in which molecular pathogenesis of pain is interrogated in relation to tissue pathology. Nonetheless, in recent years, three broad approaches have been deployed to attempt to address this: correlative clinical studies of peripheral and central pain outcomes using magnetic resonance imaging, where soft-tissue processes can be visualised; molecular studies on tissue from patients with OA; and careful molecular interrogation of preclinical models of OA across the disease time course. Studies have taken advantage of established clinical molecular targets such as nerve growth factor. Not only is the regulation of nerve growth factor within the joint being used to explore the relationship between tissue pathology and the origins of pain in OA, but it also provides a core model on which other molecules present within the joint can modulate the pain response. In this narrative review, how molecular and pathological tissue change relates to joint pain in OA will be discussed. Finally, a model for how tissue damage may lead to pain over the disease course will be proposed.
Collapse
|
13
|
Koduru SV, Elcheva IA, Leberfinger AN, Ravnic DJ. In silico analysis of RNA and small RNA sequencing data from human BM-MSCs and differentiated osteocytes, chondrocytes and tenocytes. ENGINEERED REGENERATION 2021. [DOI: 10.1016/j.engreg.2020.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
14
|
Schwer J, Rahman MM, Stumpf K, Rasche V, Ignatius A, Dürselen L, Seitz AM. Degeneration Affects Three-Dimensional Strains in Human Menisci: In situ MRI Acquisition Combined With Image Registration. Front Bioeng Biotechnol 2020; 8:582055. [PMID: 33042980 PMCID: PMC7526678 DOI: 10.3389/fbioe.2020.582055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 08/28/2020] [Indexed: 11/20/2022] Open
Abstract
Degenerative changes of menisci contribute to the evolution of osteoarthritis in the knee joint, because they alter the load transmission to the adjacent articular cartilage. Identifying alterations in the strain response of meniscal tissue under compression that are associated with progressive degeneration may uncover links between biomechanical function and meniscal degeneration. Therefore, the goal of this study was to investigate how degeneration effects the three-dimensional (3D; axial, circumferential, radial) strain in different anatomical regions of human menisci (anterior and posterior root attachment; anterior and posterior horn; pars intermedia) under simulated compression. Magnetic resonance imaging (MRI) was performed to acquire image sequences of 12 mild and 12 severe degenerated knee joints under unloaded and loaded [25%, 50% and 100% body weight (BW)] conditions using a customized loading device. Medial and lateral menisci as well as their root attachments were manually segmented. Intensity-based rigid and non-rigid image registration were performed to obtain 3D deformation fields under the respective load levels. Finally, the 3D voxels were transformed into hexahedral finite-element models and direction-dependent local strain distributions were determined. The axial compressive strain in menisci and meniscal root attachments significantly increased on average from 3.1% in mild degenerated joints to 7.3% in severe degenerated knees at 100% BW (p ≤ 0.021). In severe degenerated knee joints, the menisci displayed a mean circumferential strain of 0.45% (mild: 0.35%) and a mean radial strain of 0.41% (mild: 0.37%) at a load level of 100% BW. No significant changes were observed in the circumferential or radial directions between mild and severe degenerated knee joints for all load levels (p > 0.05). In conclusion, high-resolution MRI was successfully combined with image registration to investigate spatial strain distributions of the meniscus and its attachments in response to compression. The results of the current study highlight that the compressive integrity of the meniscus decreases with progressing tissue degeneration, whereas the tensile properties are maintained.
Collapse
Affiliation(s)
- Jonas Schwer
- Institute of Orthopaedic Research and Biomechanics, Centre for Trauma Research Ulm, Ulm University Medical Centre, Ulm, Germany
| | - Muhammed Masudur Rahman
- Institute of Orthopaedic Research and Biomechanics, Centre for Trauma Research Ulm, Ulm University Medical Centre, Ulm, Germany.,Department of Mechanical Engineering, University of Connecticut, Storrs, CT, United States
| | - Kilian Stumpf
- Experimental Cardiovascular Imaging, Department of Internal Medicine II, University Hospital Ulm, Ulm, Germany
| | - Volker Rasche
- Experimental Cardiovascular Imaging, Department of Internal Medicine II, University Hospital Ulm, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopaedic Research and Biomechanics, Centre for Trauma Research Ulm, Ulm University Medical Centre, Ulm, Germany
| | - Lutz Dürselen
- Institute of Orthopaedic Research and Biomechanics, Centre for Trauma Research Ulm, Ulm University Medical Centre, Ulm, Germany
| | - Andreas Martin Seitz
- Institute of Orthopaedic Research and Biomechanics, Centre for Trauma Research Ulm, Ulm University Medical Centre, Ulm, Germany
| |
Collapse
|
15
|
Rai MF, Brophy RH, Rosen V. Molecular biology of meniscus pathology: Lessons learned from translational studies and mouse models. J Orthop Res 2020; 38:1895-1904. [PMID: 32068295 PMCID: PMC7802285 DOI: 10.1002/jor.24630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/11/2020] [Indexed: 02/04/2023]
Abstract
Injury to any individual structure in the knee interrupts the overall function of the joint and initiates a cascade of biological and biomechanical changes whose endpoint is often osteoarthritis (OA). The knee meniscus is an integral component of knee biomechanics and may also contribute to the biological homeostasis of the joint. Meniscus injury altering knee function is associated with a high risk of OA progression, and may also be involved in the initiation of OA. As the relationship between meniscus injury and OA is very complex; despite the availability of transcript level data on human meniscus injury and meniscus mediated OA, mechanistic studies are lacking, and available human data are difficult to validate in the absence of patient-matched noninjured control tissues. As similarities exist between human and mouse knee joint structure and function, investigators have begun to use cutting-edge genetic and genomic tools to examine the usefulness of the mouse as a model to study the intricate relationship between meniscus injury and OA. In this review, we use evidence from human meniscus research to identify critical barriers hampering our understanding of meniscus injury induced OA and discuss strategies to overcome these barriers, including those that can be examined in a mouse model of injury-mediated OA.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America,Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Robert H. Brophy
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, United States of America
| |
Collapse
|
16
|
Lamplot JD, Rai MF, Tompkins WP, Friedman MV, Schmidt EJ, Sandell LJ, Brophy RH. Gene Expression in Meniscal Tears at the Time of Arthroscopic Partial Meniscectomy Predicts the Progression of Osteoarthritis Within 6 Years of Surgery. Orthop J Sports Med 2020; 8:2325967120936275. [PMID: 32923494 PMCID: PMC7446268 DOI: 10.1177/2325967120936275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022] Open
Abstract
Background: While knees with meniscal tears are associated with a heightened risk of developing osteoarthritis (OA), it is difficult to predict which patients are at the greatest risk for OA. Gene signatures in menisci that are resected during arthroscopic partial meniscectomy (APM) may provide insight into the risk of OA progression. Hypothesis: Meniscal gene signatures at the time of APM will predict radiographic OA progression. Study Design: Case series; Level of evidence, 4. Methods: Meniscal fragments were collected from 38 patients without OA during clinically indicated APM of the medial meniscus. The expression of 28 candidate genes with known roles in cartilage homeostasis, OA, extracellular matrix degradation, and obesity was assessed by quantitative real-time polymerase chain reaction. Weightbearing radiographs obtained before surgery and at final follow-up were graded by a musculoskeletal radiologist using the Kellgren-Lawrence classification of OA. The association of meniscal gene expression at baseline with the progression of radiographic OA was determined. Results: Gene expression and baseline and follow-up radiographic data were available from 31 patients (81.6%) at a mean follow-up of 6.2 ± 1.3 years. Patients without OA progression had significantly higher expression of 7 genes: MMP9 (5.1-fold; P = .002), IL8 (2.9-fold; P = .016), CCL3 (3.7-fold; P = .032), CCL3L1 (4.5-fold; P = .008), CXCL6 (6.2-fold; P = .010), LEP (5.2-fold; P = .004), and RETN (46-fold; P = .008). Conclusion: Gene expression in the meniscus at the time of APM may be associated with the risk for progression of OA after surgery. Elevated expression of the aforementioned genes may reflect a chondroprotective response. Stratifying the risk for OA progression after APM could facilitate targeted interventions to delay or prevent the development of OA. Further studies in a larger cohort with an extended follow-up, and inclusion of additional genes, are warranted to better characterize this association.
Collapse
Affiliation(s)
- Joseph D Lamplot
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, Missouri, USA.,Department of Cell Biology & Physiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - William P Tompkins
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| | - Michael V Friedman
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Eric J Schmidt
- Department of Health Sciences, University of Lynchburg, Lynchburg, Virginia, USA
| | - Linda J Sandell
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, Missouri, USA.,Department of Cell Biology & Physiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Robert H Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
17
|
Brophy RH, Schmidt EJ, Cai L, Rai MF. Duration of symptoms prior to partial meniscectomy is not associated with the expression of osteoarthritis genes in the injured meniscus. J Orthop Res 2020; 38:1268-1278. [PMID: 31876303 PMCID: PMC7225063 DOI: 10.1002/jor.24574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/21/2019] [Indexed: 02/04/2023]
Abstract
While there is emerging data on how duration of symptoms prior to surgery relates to outcomes of patients undergoing arthroscopic partial meniscectomy, little is known about how duration of symptoms relates to the biology of the knee in these patients. The purpose of this study was to test the hypothesis that duration of symptoms prior to arthroscopic partial meniscectomy is associated with expression of osteoarthritis (OA)-related genes in the meniscus. We collected resected meniscus from patients (N = 76) undergoing clinically indicated arthroscopic partial meniscectomy from knees without advanced degenerative changes. RNA from 64 patients was analyzed for 28 candidate OA transcripts by real-time polymerase chain reaction (PCR). RNA was also probed for identification of novel genes by RNA microarray in 12 patients followed by validation of selected candidates by real-time PCR. The association of gene expression with duration of symptoms prior to surgery was tested. Additional screening was performed with known OA genetic risk alleles assembled from published literature and with gene transcripts differentially expressed between non-OA and OA cartilage and menisci. Our data revealed that duration of symptoms did not predict expression of OA genes in the meniscus, other than limited association with CXCL3, BMP2, and HLA-DQA1. Microarray identified new genes and pathways with unknown role(s) in meniscus injury and OA and validation of a subset of genes by real-time PCR showed expression pattern highly concordant with the microarray data. While duration of symptoms prior to arthroscopic partial meniscectomy does not significantly alter the expression of OA related genes, the association with novel genes and pathways deserves further investigation.
Collapse
Affiliation(s)
- Robert H. Brophy
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Eric J. Schmidt
- School of Physician Assistant Medicine, College of Health Sciences, University of Lynchburg College, Lynchburg, VA, USA
| | - Lei Cai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Muhammad Farooq Rai
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO, 63110, USA,Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| |
Collapse
|
18
|
Sun H, Wen X, Li H, Wu P, Gu M, Zhao X, Zhang Z, Hu S, Mao G, Ma R, Liao W, Zhang Z. Single-cell RNA-seq analysis identifies meniscus progenitors and reveals the progression of meniscus degeneration. Ann Rheum Dis 2020; 79:408-417. [PMID: 31871141 PMCID: PMC7034356 DOI: 10.1136/annrheumdis-2019-215926] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/21/2019] [Accepted: 12/07/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVES The heterogeneity of meniscus cells and the mechanism of meniscus degeneration is not well understood. Here, single-cell RNA sequencing (scRNA-seq) was used to identify various meniscus cell subsets and investigate the mechanism of meniscus degeneration. METHODS scRNA-seq was used to identify cell subsets and their gene signatures in healthy human and degenerated meniscus cells to determine their differentiation relationships and characterise the diversity within specific cell types. Colony-forming, multi-differentiation assays and a mice meniscus injury model were used to identify meniscus progenitor cells. We investigated the role of degenerated meniscus progenitor (DegP) cell clusters during meniscus degeneration using computational analysis and experimental verification. RESULTS We identified seven clusters in healthy human meniscus, including five empirically defined populations and two novel populations. Pseudotime analysis showed endothelial cells and fibrochondrocyte progenitors (FCP) existed at the pseudospace trajectory start. Melanoma cell adhesion molecule ((MCAM)/CD146) was highly expressed in two clusters. CD146+ meniscus cells differentiated into osteoblasts and adipocytes and formed colonies. We identified changes in the proportions of degenerated meniscus cell clusters and found a cluster specific to degenerative meniscus with progenitor cell characteristics. The reconstruction of four progenitor cell clusters indicated that FCP differentiation into DegP was an aberrant process. Interleukin 1β stimulation in healthy human meniscus cells increased CD318+ cells, while TGFβ1 attenuated the increase in CD318+ cells in degenerated meniscus cells. CONCLUSIONS The identification of meniscus progenitor cells provided new insights into cell-based meniscus tissue engineering, demonstrating a novel mechanism of meniscus degeneration, which contributes to the development of a novel therapeutic strategy.
Collapse
Affiliation(s)
- Hao Sun
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Guangzhou, China
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Xingzhao Wen
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Hongyi Li
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Peihui Wu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Minghui Gu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Xiaoyi Zhao
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Ziji Zhang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Shu Hu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Guping Mao
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Ruofan Ma
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - Weiming Liao
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| | - Zhiqi Zhang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, China
| |
Collapse
|
19
|
|
20
|
Abstract
Meniscal injuries are common in patients with varus or valgus malalignment, but consensus is lacking as to when surgery should address the meniscal injury only and when it should be combined with an osteotomy. Several factors need to be evaluated to provide the most appropriate treatment in each case. Here we highlight the most relevant literature on the subject and suggest a rationale for surgical treatment.
Collapse
Affiliation(s)
- Pablo Eduardo Gelber
- ICATME-Hospital Universitari Dexeus, Universitat Autònoma de Barcelona, Barcelona, Spain; Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Bjorn Barenius
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Intitutet, Solnavägen 1, Solna, Stockholm 17177, Sweden
| | - Simone Perelli
- ICATME-Hospital Universitari Dexeus, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
21
|
Zhang Y, Wang F, Bao L, Li J, Shi Z, Wang J. Cyclic hydrostatic compress force regulates apoptosis of meniscus fibrochondrocytes via integrin α5β1. Physiol Res 2019; 68:639-649. [DOI: 10.33549/physiolres.934088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Meniscus is a semilunar fibrocartilaginous tissue, serving important roles in load buffering, stability, lubrication, proprioception, and nutrition of the knee joint. The degeneration and damage of meniscus has been proved to be a risk factor of knee osteoarthritis. Mechanical stimulus is a critical factor of the development, maintenance and repair of the meniscus fibrochondrocytes. However, the mechanism of the mechano-transduction process remains elusive. Here we reported that cyclic hydrostatic compress force (CHCF) treatment promotes proliferation and inhibits apoptosis of the isolated primary meniscus fibrochondrocytes (PMFs), via upregulating the expression level of integrin α5β1. Consequently, increased phosphorylated-ERK1/2 and phosphorylated-PI3K, and decreased caspase-3 were detected. These effects of CHCF treatment can be abolished by integrin α5β1 inhibitor or specific siRNA transfection. These data indicate that CHCF regulates apoptosis of PMFs via integrin α5β1-FAK-PI3K/ERK pathway, which may be an important candidate approach during meniscus degeneration.
Collapse
Affiliation(s)
| | | | | | | | | | - J. Wang
- Department of orthopedic surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
22
|
Rai MF, Tycksen ED, Cai L, Yu J, Wright RW, Brophy RH. Distinct degenerative phenotype of articular cartilage from knees with meniscus tear compared to knees with osteoarthritis. Osteoarthritis Cartilage 2019; 27:945-955. [PMID: 30797944 PMCID: PMC6536326 DOI: 10.1016/j.joca.2019.02.792] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/29/2019] [Accepted: 02/07/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To compare the transcriptome of articular cartilage from knees with meniscus tears to knees with end-stage osteoarthritis (OA). DESIGN Articular cartilage was collected from the non-weight bearing medial intercondylar notch of knees undergoing arthroscopic partial meniscectomy (APM; N = 10, 49.7 ± 10.8 years, 50% females) for isolated medial meniscus tears and knees undergoing total knee arthroplasty (TKA; N = 10, 66.0 ± 7.6 years, 70% females) due to end-stage OA. Ribonucleic acid (RNA) preparation was subjected to SurePrint G3 human 8 × 60K RNA microarrays to probe differentially expressed transcripts followed by computational exploration of underlying biological processes. Real-time polymerase chain reaction amplification was performed on selected transcripts to validate microarray data. RESULTS We observed that 81 transcripts were significantly differentially expressed (45 elevated, 36 repressed) between APM and TKA samples (≥ 2 fold) at a false discovery rate of ≤ 0.05. Among these, CFD, CSN1S1, TSPAN11, CSF1R and CD14 were elevated in the TKA group, while CHI3L2, HILPDA, COL3A1, COL27A1 and FGF2 were highly expressed in APM group. A few long intergenic non-coding RNAs (lincRNAs), small nuclear RNAs (snoRNAs) and antisense RNAs were also differentially expressed between the two groups. Transcripts up-regulated in TKA cartilage were enriched for protein localization and activation, chemical stimulus, immune response, and toll-like receptor signaling pathway. Transcripts up-regulated in APM cartilage were enriched for mesenchymal cell apoptosis, epithelial morphogenesis, canonical glycolysis, extracellular matrix organization, cartilage development, and glucose catabolic process. CONCLUSIONS This study suggests that APM and TKA cartilage express distinct sets of OA transcripts. The gene profile in cartilage from TKA knees represents an end-stage OA whereas in APM knees it is clearly earlier in the degenerative process.
Collapse
MESH Headings
- Adult
- Aged
- Arthroplasty, Replacement, Knee
- Cartilage, Articular/metabolism
- Case-Control Studies
- Caseins/genetics
- Chitinases/genetics
- Collagen Type III/genetics
- Complement Factor D/genetics
- Female
- Fibrillar Collagens/genetics
- Fibroblast Growth Factor 2/genetics
- Gene Expression Profiling
- Humans
- Lipopolysaccharide Receptors/genetics
- Male
- Meniscectomy
- Middle Aged
- Neoplasm Proteins/genetics
- Osteoarthritis, Knee/genetics
- Osteoarthritis, Knee/surgery
- Phenotype
- RNA/metabolism
- RNA, Antisense/metabolism
- RNA, Long Noncoding/metabolism
- RNA, Messenger/metabolism
- RNA, Small Nuclear/metabolism
- Real-Time Polymerase Chain Reaction
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Tetraspanins/genetics
- Tibial Meniscus Injuries/genetics
- Tibial Meniscus Injuries/surgery
Collapse
Affiliation(s)
- M F Rai
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - E D Tycksen
- Genome Technology Access Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - L Cai
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - J Yu
- Genome Technology Access Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - R W Wright
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - R H Brophy
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
23
|
Osteoarthritis following meniscus and ligament injury: insights from translational studies and animal models. Curr Opin Rheumatol 2019; 31:70-79. [DOI: 10.1097/bor.0000000000000566] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
24
|
Huang P, Gu J, Wu J, Geng L, Hong Y, Wang S, Wang M. Microarray analysis of the molecular mechanisms associated with age and body mass index in human meniscal injury. Mol Med Rep 2018; 19:93-102. [PMID: 30483788 PMCID: PMC6297773 DOI: 10.3892/mmr.2018.9685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 10/04/2018] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to identify genes and functional pathways associated with meniscal injuries affected by age or body mass index (BMI) using microarray analysis. The GSE45233 gene expression dataset with 12 injured meniscus samples associated with age and BMI and GSE66635 dataset with 12 injured and 12 normal meniscus samples were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified based on age or BMI in GSE45233. DEGs between injured and normal meniscus samples in GSE66635 were also identified. Common DEGs between GSE45233 and GSE66635 were identified as feature genes associated with age or BMI, followed by protein-protein interaction (PPI) network and functional pathway enrichment analyses for the feature genes. Finally, the GSE51588 genome-wide expression profile was then downloaded from the GEO database to validate the results. A total of 1,328 DEGs were identified. Of these, 28 age-associated and 20 BMI-associated meniscal injury genes were obtained. B-cell lymphoma-2 (Bcl-2) and matrix metalloproteinase-14 were identified as hub genes in the PPI networks. Functional pathway enrichment analysis revealed that vascular endothelial growth factor A (VEGFA), transferrin (TF) and Bcl-2 were involved in the hypoxia-inducible factor 1 signaling pathway. TF was involved in the mineral absorption function pathway associated with BMI. Additionally, TF and VEGFA were identified to be overlapping candidate genes of GSE45233 and GSE66635, and DEGs in GSE51588. Therefore, VEGFA, TF, and Bcl-2 may be important genes for human meniscal injuries. Additional evaluations of these results are required.
Collapse
Affiliation(s)
- Peiyan Huang
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Jun Gu
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Junguo Wu
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Lei Geng
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Yang Hong
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Siqun Wang
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| | - Minghai Wang
- Department of Orthopedic Surgery, Shanghai Fifth People's Hospital Affiliated to Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
25
|
Grogan SP, Duffy SF, Pauli C, Lotz MK, D’Lima DD. Gene expression profiles of the meniscus avascular phenotype: A guide for meniscus tissue engineering. J Orthop Res 2018; 36:1947-1958. [PMID: 29411909 PMCID: PMC6326361 DOI: 10.1002/jor.23864] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 02/05/2018] [Indexed: 02/04/2023]
Abstract
Avascular (Avas) meniscus regeneration remains a challenge, which is partly a consequence of our limited knowledge of the cells that maintain this tissue region. In this study, we utilized microarrays to characterize gene expression profiles of intact human Avas meniscus tissue and of cells following culture expansion. Using these data, we examined various 3D culture conditions to redifferentiate Avas cells toward the tissue phenotype. RNA was isolated from either the tissue directly or following cell isolation and 2 weeks in monolayer culture. RNA was hybridized on human genome arrays. Differentially expressed (DE) genes were identified by ranking analysis. DAVID pathway analysis was performed and visualized using STRING analysis. Quantitative PCR (qPCR) on additional donor menisci (tissues and cells) were used to validate array data. Avas cells cultured in 3D were subjected to qPCR to compare with the array-generated data. A total of 387 genes were DE based on differentiation state (>3-fold change; p < 0.01). In Avas-cultured cells, the upregulated pathways included focal adhesion, ECM-receptor interaction, regulation of actin cytoskeleton, and PDGF Signaling. In 3D-cultured Avas cells, TGFβ1 or combinations of TGFβ1 and BMP6 were most effective to promote an Avas tissue phenotype. THBS2 and THBS4 expression levels were identified as a means to denote meniscus cell phenotype status. We identified the key gene expression profiles, new markers and pathways involved in characterizing the Avas meniscus phenotype in the native state and during in vitro dedifferentiation and redifferentiation. These data served to screen 3D conditions to generate meniscus-like neotissues. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1947-1958, 2018.
Collapse
Affiliation(s)
- Shawn P Grogan
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA
| | - Stuart F. Duffy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Chantal Pauli
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Martin K Lotz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Darryl D D’Lima
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA,Corresponding author: Darryl D D’Lima, MD, PhD, Shiley Center for Orthopaedic Research and Education at Scripps Clinic, 11025 North Torrey Pines Road, Suite 200, La Jolla, CA 92037, Tel 858 332 0166 Fax 858 332 0669,
| |
Collapse
|
26
|
McIlwraith CW, Kawcak CE, Frisbie DD, Little CB, Clegg PD, Peffers MJ, Karsdal MA, Ekman S, Laverty S, Slayden RA, Sandell LJ, Lohmander LS, Kraus VB. Biomarkers for equine joint injury and osteoarthritis. J Orthop Res 2018; 36:823-831. [PMID: 28921609 DOI: 10.1002/jor.23738] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/07/2017] [Indexed: 02/04/2023]
Abstract
We report the results of a symposium aimed at identifying validated biomarkers that can be used to complement clinical observations for diagnosis and prognosis of joint injury leading to equine osteoarthritis (OA). Biomarkers might also predict pre-fracture change that could lead to catastrophic bone failure in equine athletes. The workshop was attended by leading scientists in the fields of equine and human musculoskeletal biomarkers to enable cross-disciplinary exchange and improve knowledge in both. Detailed proceedings with strategic planning was written, added to, edited and referenced to develop this manuscript. The most recent information from work in equine and human osteoarthritic biomarkers was accumulated, including the use of personalized healthcare to stratify OA phenotypes, transcriptome analysis of anterior cruciate ligament (ACL) and meniscal injuries in the human knee. The spectrum of "wet" biomarker assays that are antibody based that have achieved usefulness in both humans and horses, imaging biomarkers and the role they can play in equine and human OA was discussed. Prediction of musculoskeletal injury in the horse remains a challenge, and the potential usefulness of spectroscopy, metabolomics, proteomics, and development of biobanks to classify biomarkers in different stages of equine and human OA were reviewed. The participants concluded that new information and studies in equine musculoskeletal biomarkers have potential translational value for humans and vice versa. OA is equally important in humans and horses, and the welfare issues associated with catastrophic musculoskeletal injury in horses add further emphasis to the need for good validated biomarkers in the horse. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:823-831, 2018.
Collapse
Affiliation(s)
- C Wayne McIlwraith
- Orthopaedic Research Center, Barbara Cox Anthony University Chair in Orthopaedics, Colorado State University, 300 West Drake Road, Fort Collins, Colorado 80523
| | - Christopher E Kawcak
- Orthopaedic Research Center, Barbara Cox Anthony University Chair in Orthopaedics, Colorado State University, 300 West Drake Road, Fort Collins, Colorado 80523
| | - David D Frisbie
- Orthopaedic Research Center, Barbara Cox Anthony University Chair in Orthopaedics, Colorado State University, 300 West Drake Road, Fort Collins, Colorado 80523
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Labs, The University of Sydney, Sydney, Australia
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Mandy J Peffers
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | | | - Stina Ekman
- Department of Biomedicine and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Sheila Laverty
- Department of Clinical Sciences, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - Richard A Slayden
- Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, Colorado
| | - Linda J Sandell
- Department of Orthopaedic Surgery, Washington University, St. Louis, Missouri
| | - L S Lohmander
- Department of Clinical Sciences Lund, Orthopaedics, Lund University, Lund, Sweden
| | - Virginia B Kraus
- Duke Molecular Physiology Institute and Division of Rheumatology, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
27
|
Brophy RH, Zhang B, Cai L, Wright RW, Sandell LJ, Rai MF. Transcriptome comparison of meniscus from patients with and without osteoarthritis. Osteoarthritis Cartilage 2018; 26:422-432. [PMID: 29258882 PMCID: PMC6007850 DOI: 10.1016/j.joca.2017.12.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 11/13/2017] [Accepted: 12/08/2017] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To assess the impact of osteoarthritis (OA) on the meniscus by comparing transcripts and biological processes in the meniscus between patients with and without OA. DESIGN RNA microarrays were used to identify transcripts differentially expressed (DE) in meniscus obtained from 12 OA and 12 non-OA patients. The non-OA specimens were obtained at the time of arthroscopic partial meniscectomy. Real-time PCR was performed on selected transcripts. Biological processes and gene-networking was examined computationally. Transcriptome signatures were mapped with 37 OA-related transcripts to evaluate how meniscus gene expression relates to that of OA cartilage. RESULTS We identified 168 transcripts significantly DE between OA (75 elevated, 93 repressed) and non-OA samples (≥1.5-fold). Among these, CSN1S1, COL10A1, WIF1, and SPARCL1 were the most prominent transcripts elevated in OA meniscus, POSTN and VEGFA were most highly repressed in OA meniscus. Transcripts elevated in OA meniscus represented response to external stimuli, cell migration and cell localization while those repressed in OA meniscus represented histone deacetylase activity (related to epigenetics) and skeletal development. Numerous long non-coding RNAs (lncRNAs) were DE between the two groups. When segregated by OA-related transcripts, two distinct clustering patterns appeared: OA meniscus appeared to be more inflammatory while non-OA meniscus exhibited a "repair" phenotype. CONCLUSIONS Numerous transcripts with potential relevance to the pathogenesis of OA are DE in OA and non-OA meniscus. These data suggest an involvement of epigenetically regulated histone deacetylation in meniscus tears as well as expression of lncRNAs. Patient clustering based on transcripts related to OA in articular cartilage confirmed distinct phenotypes between injured (non-OA) and OA meniscus.
Collapse
Affiliation(s)
- R H Brophy
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - B Zhang
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine at Barnes-Jewish Hospital, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - L Cai
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - R W Wright
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - L J Sandell
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Department of Cell Biology and Physiology, Washington University School of Medicine at Barnes-Jewish Hospital, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Engineering & Applied Science, 1 Brookings Drive, St. Louis, MO 63130, USA.
| | - M F Rai
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Department of Cell Biology and Physiology, Washington University School of Medicine at Barnes-Jewish Hospital, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
28
|
Rai MF, Tycksen ED, Sandell LJ, Brophy RH. Advantages of RNA-seq compared to RNA microarrays for transcriptome profiling of anterior cruciate ligament tears. J Orthop Res 2018; 36:484-497. [PMID: 28749036 PMCID: PMC5787041 DOI: 10.1002/jor.23661] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 07/19/2017] [Indexed: 02/04/2023]
Abstract
Microarrays and RNA-seq are at the forefront of high throughput transcriptome analyses. Since these methodologies are based on different principles, there are concerns about the concordance of data between the two techniques. The concordance of RNA-seq and microarrays for genome-wide analysis of differential gene expression has not been rigorously assessed in clinically derived ligament tissues. To demonstrate the concordance between RNA-seq and microarrays and to assess potential benefits of RNA-seq over microarrays, we assessed differences in transcript expression in anterior cruciate ligament (ACL) tissues based on time-from-injury. ACL remnants were collected from patients with an ACL tear at the time of ACL reconstruction. RNA prepared from torn ACL remnants was subjected to Agilent microarrays (N = 24) and RNA-seq (N = 8). The correlation of biological replicates in RNA-seq and microarrays data was similar (0.98 vs. 0.97), demonstrating that each platform has high internal reproducibility. Correlations between the RNA-seq data and the individual microarrays were low, but correlations between the RNA-seq values and the geometric mean of the microarrays values were moderate. The cross-platform concordance for differentially expressed transcripts or enriched pathways was linearly correlated (r = 0.64). RNA-Seq was superior in detecting low abundance transcripts and differentiating biologically critical isoforms. Additional independent validation of transcript expression was undertaken using microfluidic PCR for selected genes. PCR data showed 100% concordance (in expression pattern) with RNA-seq and microarrays data. These findings demonstrate that RNA-seq has advantages over microarrays for transcriptome profiling of ligament tissues when available and affordable. Furthermore, these findings are likely transferable to other musculoskeletal tissues where tissue collection is challenging and cells are in low abundance. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:484-497, 2018.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 660 S. Euclid Ave., St. Louis, MO 63110, United States,Department of Cell Biology and Physiology, Washington University School of Medicine at Barnes-Jewish Hospital, 660 S. Euclid Ave., St. Louis, MO 63110, United States,Corresponding author: Muhammad Farooq Rai, Ph.D., Department of Orthopaedic Surgery, Washington University School of Medicine at Barnes-Jewish Hospital, MS 8233, 660 South Euclid Avenue, St. Louis, MO 63110 United States, Ph: 314-286-0955; Fax: 314-362-0334;
| | - Eric D. Tycksen
- Genome Technology Access Center, Washington University School of Medicine at Barnes-Jewish Hospital, 660 S. Euclid Ave., St. Louis, MO 63110, United States
| | - Linda J. Sandell
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 660 S. Euclid Ave., St. Louis, MO 63110, United States,Department of Cell Biology and Physiology, Washington University School of Medicine at Barnes-Jewish Hospital, 660 S. Euclid Ave., St. Louis, MO 63110, United States,Department of Biomedical Engineering, Washington University School of Medicine at Barnes-Jewish Hospital, 660 S. Euclid Ave., St. Louis, MO 63110, United States
| | - Robert H. Brophy
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, 660 S. Euclid Ave., St. Louis, MO 63110, United States
| |
Collapse
|
29
|
Barboza E, Hudson J, Chang WP, Kovats S, Towner RA, Silasi-Mansat R, Lupu F, Kent C, Griffin TM. Profibrotic Infrapatellar Fat Pad Remodeling Without M1 Macrophage Polarization Precedes Knee Osteoarthritis in Mice With Diet-Induced Obesity. Arthritis Rheumatol 2017; 69:1221-1232. [PMID: 28141918 DOI: 10.1002/art.40056] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/24/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To test the hypothesis that high-fat (HF) diet-induced obesity increases proinflammatory cytokine expression, macrophage infiltration, and M1 polarization in the infrapatellar fat pad (IFP) prior to knee cartilage degeneration. METHODS We characterized the effect of HF feeding on knee OA pathology, body adiposity, and glucose intolerance in male C57BL/6J mice and identified a diet duration that induces metabolic dysfunction prior to cartilage degeneration. Magnetic resonance imaging and histomorphology were used to quantify changes in the epididymal, subcutaneous, and infrapatellar fat pads and in adipocyte sizes. Finally, we used targeted gene expression and protein arrays, immunohistochemistry, and flow cytometry to quantify differences in fat pad markers of inflammation and immune cell populations. RESULTS Twenty weeks of feeding with an HF diet induced marked obesity, glucose intolerance, and early osteoarthritis (OA), including osteophytes and cartilage tidemark duplication. This duration of HF feeding increased the IFP volume. However, it did not increase IFP inflammation, macrophage infiltration, or M1 macrophage polarization as observed in epididymal fat. Furthermore, leptin protein levels were reduced. This protection from obesity-induced inflammation corresponded to increased IFP fibrosis and the absence of adipocyte hypertrophy. CONCLUSION The IFP does not recapitulate classic abdominal adipose tissue inflammation during the early stages of knee OA in an HF diet-induced model of obesity. Consequently, these findings do not support the hypothesis that IFP inflammation is an initiating factor of obesity-induced knee OA. Furthermore, the profibrotic and antihypertrophic responses of IFP adipocytes to HF feeding suggest that intraarticular adipocytes are subject to distinct spatiotemporal structural and metabolic regulation among fat pads.
Collapse
Affiliation(s)
| | | | | | - Susan Kovats
- Oklahoma Medical Research Foundation, Oklahoma City
| | | | | | - Florea Lupu
- Oklahoma Medical Research Foundation, Oklahoma City
| | - Collin Kent
- Oklahoma Medical Research Foundation, Oklahoma City
| | - Timothy M Griffin
- Oklahoma Medical Research Foundation, Reynolds Oklahoma Center on Aging, and University of Oklahoma Health Sciences Center, Oklahoma City
| |
Collapse
|
30
|
|
31
|
Melrose J, Fuller ES, Little CB. The biology of meniscal pathology in osteoarthritis and its contribution to joint disease: beyond simple mechanics. Connect Tissue Res 2017; 58:282-294. [PMID: 28121190 DOI: 10.1080/03008207.2017.1284824] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The meniscal cartilages in the knee function to improve congruity of the medial and lateral femoro-tibial joints and play critical roles in load distribution and joint stability. Meniscal tears of various configurations are one of the most common conditions of the knee and are associated with an increased risk of developing osteoarthritis (OA). While this risk has been largely attributed to loss of the biomechanical functions of the menisci, there is accumulating evidence suggesting that other aspects of meniscal biology may play a role in determining the long-term consequences of meniscal damage for joint health. In this narrative review, we examine the existing literature and present some new data implicating synthesis and secretion of enzymes and other pro-catabolic mediators by injured and degenerate menisci, contributing to the pathological change in other knee joint tissues in OA.
Collapse
Affiliation(s)
- James Melrose
- a Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Sydney Medical School Northern , University of Sydney, Royal North Shore Hospital , St. Leonards , Australia.,b Graduate School of Biomedical Engineering , University of New South Wales , Sydney , Australia
| | - Emily S Fuller
- a Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Sydney Medical School Northern , University of Sydney, Royal North Shore Hospital , St. Leonards , Australia
| | - Christopher B Little
- a Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute, Northern Sydney Local Health District, Sydney Medical School Northern , University of Sydney, Royal North Shore Hospital , St. Leonards , Australia
| |
Collapse
|
32
|
Brophy RH, Sandell LJ, Rai MF. Traumatic and Degenerative Meniscus Tears Have Different Gene Expression Signatures. Am J Sports Med 2017; 45:114-120. [PMID: 27604189 PMCID: PMC5969913 DOI: 10.1177/0363546516664889] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Meniscus tears are classified as traumatic or degenerative based on the tear pattern. There is little evidence demonstrating biological differences between the 2 tear types. HYPOTHESIS Gene expression signatures in the injured meniscus are different between traumatic (vertical) and degenerative (complex, horizontal, or flap) tears. STUDY DESIGN Controlled laboratory study. METHODS Samples of the torn meniscus from the white-white zone were removed at the time of clinically indicated partial meniscectomy from 48 patients (37 with degenerative tears and 11 with traumatic tears). mRNA expression in the injured menisci was measured by quantitative real-time polymerase chain reaction for selected molecular markers of osteoarthritis, inflammation, and cartilage homeostasis (eg, cytokines/chemokines, aggrecanases/metalloproteinases, transcription factors, cartilage matrix genes, and adipokines). The tear pattern (traumatic or degenerative) and location (medial or lateral) were recorded for each patient. Gene expression differences between degenerative and traumatic tears were computed after adjusting for patients' age, sex, and body mass index and for location of the resected meniscus (medial/lateral). RESULTS Gene expression in meniscus tears varied by pattern. Chemokines ( IL8 [ P < .001] and CXCL6 [ P < .001]) and matrix metalloproteinases ( MMP1 [ P = .011] and MMP3 [ P = .016]) were expressed at a significantly higher level in traumatic tears compared with degenerative tears. In contrast, COL1A1 was expressed at a lower level in traumatic tears compared with degenerative tears ( P = .058). None of the genes tested demonstrated significant differences between medial and lateral meniscus tears. CONCLUSION Traumatic meniscus tears overall exhibited a higher inflammatory/catabolic response as evidenced by higher levels of chemokine and matrix metalloproteinase expression than degenerative tears. These findings suggest that there is a (molecular) biological distinction between traumatic and degenerative tears. CLINICAL RELEVANCE The catabolic/inflammatory differences between traumatic and degenerative tears may be relevant to treatment decisions regarding the meniscus as well as advance our understanding of how meniscus tears relate to the development of knee osteoarthritis.
Collapse
Affiliation(s)
- Robert H. Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, United States,Corresponding author: Department of Orthopaedic Surgery, Washington University School of Medicine, 14532, South Outer Forty Drive, Chesterfield, MO 63017, USA, , Tel: 314-514-3564; Fax: 314-514-3689
| | - Linda J. Sandell
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, United States,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, United States
| | - M. Farooq Rai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
33
|
Brophy RH, Sandell LJ, Cheverud JM, Rai MF. Gene expression in human meniscal tears has limited association with early degenerative changes in knee articular cartilage. Connect Tissue Res 2017; 58:295-304. [PMID: 27435997 PMCID: PMC5931210 DOI: 10.1080/03008207.2016.1211114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
UNLABELLED Purpose/Aim: Meniscus tears are a common injury to the knee associated with the development of osteoarthritis. Gene expression in the injured meniscus may be associated with early degeneration in the articular cartilage. The purpose of this study was to test the hypothesis that gene expression in meniscus tears is associated with early degenerative changes in the articular cartilage at the time of partial meniscectomy. MATERIALS AND METHODS Torn meniscus was removed at the time of partial meniscectomy in 68 patients without radiographic osteoarthritis. Meniscal mRNA expression was measured by quantitative PCR for multiple molecular markers of osteoarthritis and cartilage homeostasis. The presence of early degenerative changes in the knee was recorded by X-ray (N = 63), magnetic resonance imaging (MRI, N = 48), and arthroscopy (N = 63). Gene expression was tested for correlation with the presence/absence of degenerative changes after adjusting for age, sex, and body mass index. RESULTS Overall gene expression varied significantly with degenerative changes based on X-ray (P = 0.047) and MRI (P = 0.018). The linear combination of gene variation was also significant. However, only adiponectin (ADIPOQ) (P = 0.015) was expressed at a significantly lower level in patients with chondrosis on MRI, while the expression of ADIPOQ (P = 0.035) and resistin (RETN) (P = 0.017) was higher in patients with early degenerative changes on X-ray. None of the genes varied significantly with presence/absence of chondrosis as measured by arthroscopy. CONCLUSIONS There is an overall association of gene expression in meniscal tears to early degenerative changes in the knee, but only a limited number of specific genes demonstrate this relationship. The roles of adiponectin and resistin in knee injury and osteoarthritis deserve further study.
Collapse
Affiliation(s)
- Robert H. Brophy
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA,
| | - Linda J. Sandell
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA,Department of Biomedical Engineering, Washington University in St. Louis, MO, USA
| | | | - Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
34
|
Abstract
The knee is a fascinating yet complex joint. Researchers and clinicians agree that the joint is an organ comprised of highly specialized intrinsic and extrinsic tissues contributing to both health and disease. Key to the function and movement of the knee are the menisci, exquisite fibrocartilage structures that are critical structures for maintaining biological and biomechanical integrity of the joint. The biological/physiological functions of the menisci must be understood at the tissue, cellular and even molecular levels in order to determine clinically relevant methods for assessing it and influencing it. By investigating normal and pathological functions at the basic science level, we can begin to translate data to patients. The objective of this article is to provide an overview of this translational pathway so that progression toward improved diagnostic, preventative, and therapeutic strategies can be effectively pursued. We have thoroughly examined the pathobiological, biomarker, and imaging aspects of meniscus research. This translational approach can be effective toward optimal diagnosis, prevention, and treatment for the millions of patients who suffer from meniscal disorders each year.
Collapse
Affiliation(s)
- James L Cook
- a University of Missouri Department of Orthopaedic Surgery and Thompson Laboratory for Regenerative Orthopaedics , Columbia , MO , USA
| | - Keiichi Kuroki
- a University of Missouri Department of Orthopaedic Surgery and Thompson Laboratory for Regenerative Orthopaedics , Columbia , MO , USA
| | - Aaron M Stoker
- a University of Missouri Department of Orthopaedic Surgery and Thompson Laboratory for Regenerative Orthopaedics , Columbia , MO , USA
| | - Farrah A Monibi
- a University of Missouri Department of Orthopaedic Surgery and Thompson Laboratory for Regenerative Orthopaedics , Columbia , MO , USA
| | - Brandon L Roller
- b Department of Radiology , Wake Forest Baptist Medical Center , Winston-Salem , NC , USA
| |
Collapse
|
35
|
Brophy RH, Tycksen ED, Sandell LJ, Rai MF. Changes in Transcriptome-Wide Gene Expression of Anterior Cruciate Ligament Tears Based on Time From Injury. Am J Sports Med 2016; 44:2064-75. [PMID: 27159315 DOI: 10.1177/0363546516643810] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Anterior cruciate ligament (ACL) tears are a common injury. The healing potential of the injured ACL is poorly understood and is considered limited. Therefore, most ACL tears that are treated surgically undergo reconstruction rather than repair. However, there has been renewed interest recently in repairing ACL tears despite unanswered questions regarding the healing capacity of the ACL. HYPOTHESIS Gene expression in the injured ACL varies with time from injury. STUDY DESIGN Descriptive laboratory study. METHODS Transcriptome-wide expression profiles of 24 human ACL remnants recovered at the time of surgical reconstruction were analyzed using the Agilent human 8x60K microarray platform. Gene ontology was performed on differentially expressed transcripts based on time from injury (acute, <3 months; intermediate, 3-12 months; chronic, >12 months). A subset of transcripts with large fold changes in expression between any 2 categories was validated via microfluidic digital polymerase chain reaction. RESULTS Numerous transcripts representing important biological processes were differentially expressed by time from injury. The most significant changes were noted between the acute and chronic groups. Expression of several extracellular matrix genes- namely, POSTN, COL5A1, COL1A1, and COL12A1-was lower in the chronic tears compared with acute and intermediate tears. In acute tears, processes representing angiogenesis and stem cell differentiation were affected. In intermediate tears, processes representing stem cell proliferation concomitant with cellular component organization/cellular localization were altered. In ACL tears more than 12 months out from injury, processes denoting myosin filament organization, cellular component organization/cell localization, and extracellular matrix organization were affected. CONCLUSION These findings are consistent with initial repair activity in the injured ACL, which declines with time from injury. Individual genes identified in this study, such as periostin, deserve further investigation into their role in tissue repair. CLINICAL RELEVANCE The decreased healing capacity of ACL tears over time is relevant to the development of effective techniques for repairing ACL tears and may have some significance for ACL reconstruction techniques as well. The potential for healing appears to be greatest in acute ACL tears, suggesting this window should be the focus of research for ACL repair.
Collapse
Affiliation(s)
- Robert H Brophy
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, St Louis, Missouri, USA
| | - Eric D Tycksen
- Genome Technology Access Center, Washington University in St Louis, St Louis, Missouri, USA
| | - Linda J Sandell
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, St Louis, Missouri, USA Department of Biomedical Engineering, Washington University in St Louis at Engineering and Applied Sciences, St Louis, Missouri, USA Department of Cell Biology and Physiology, Washington University School of Medicine at Barnes-Jewish Hospital, St Louis, Missouri, USA
| | - Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes-Jewish Hospital, St Louis, Missouri, USA
| |
Collapse
|
36
|
Vanderman KS, Loeser RF, Chubinskaya S, Anderson A, Ferguson CM. Reduced response of human meniscal cells to Osteogenic Protein 1 during osteoarthritis and pro-inflammatory stimulation. Osteoarthritis Cartilage 2016; 24:1036-46. [PMID: 26778533 PMCID: PMC4875791 DOI: 10.1016/j.joca.2015.12.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 11/22/2015] [Accepted: 12/27/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Many cell types lose responsiveness to anabolic factors during inflammation and disease. Osteogenic Protein 1 (OP1/BMP7) was evaluated for the ability to enhance extracellular matrix synthesis in healthy and OA meniscus cells. Mechanisms of cell response to OP1 were explored. DESIGN Meniscus and cartilage tissues from healthy tissue donors and osteoarthritis (OA) patients undergoing total knee arthroplasties were acquired. Primary cell cultures were stimulated with OP1 and/or inflammatory factors (IL1α, IL1β, or fibronectin fragments (FnF)) and cellular responses were analyzed by RT-qPCR and immunoblots. Frozen section immunohistochemistry (IHC) was conducted to assess OP1 and receptor proteins in normal and OA meniscus. RESULTS OP1 treatment of normal meniscus cells resulted in significant, dose-dependent increases in ACAN (aggrecan) and COL2A1, and decreased MMP13 gene transcription, while only ACAN was upregulated (P < 0.01) at the highest dose of OP1 in OA meniscus cells. OP1 induced significantly more ACAN gene transcription in normal meniscus than normal articular cartilage (P = 0.05), and no differences between normal and OA cartilage were detected. Receptor expression and kinetics of canonical signaling activation were similar between normal and OA specimens. Normal meniscus cells treated with inflammatory factors were refractory to OP1 stimulation. Smad1 phosphorylation at an inhibitory site was induced (P = 0.01 for both normal and OA meniscus) by inflammatory cytokine treatment. CONCLUSIONS The meniscus demonstrates resistance to OP1 stimulation in OA and in the presence of inflammatory mediators. MAPK-mediated Smad1 linker phosphorylation is a possible mediator of the loss of anabolic extracellular matrix production in the inflammatory cytokine affected meniscus.
Collapse
Affiliation(s)
- K S Vanderman
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | - R F Loeser
- Department of Medicine and the Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599-7280, USA.
| | - S Chubinskaya
- Department of Biochemistry, Rush University Medical Center, 1653 W, Congress Parkway, Chicago, IL 60612, USA.
| | - A Anderson
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | - C M Ferguson
- Department of Orthopaedic Surgery, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| |
Collapse
|
37
|
Van Der Straeten C, Byttebier P, Eeckhoudt A, Victor J. Meniscal Allograft Transplantation Does Not Prevent or Delay Progression of Knee Osteoarthritis. PLoS One 2016; 11:e0156183. [PMID: 27228174 PMCID: PMC4881946 DOI: 10.1371/journal.pone.0156183] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/10/2016] [Indexed: 11/23/2022] Open
Abstract
Background Meniscal tears are common knee injuries. Meniscal allograft transplantation (MAT) has been advocated to alleviate symptoms and delay osteoarthritis (OA) after meniscectomy. We investigated (1) the long-term outcome of MAT as a treatment of symptomatic meniscectomy, (2) most important factors affecting survivorship and (3) OA progression. Methods From 1989 till 2013, 329 MAT were performed in 313 patients. Clinical and radiographic results and MAT survival were evaluated retrospectively. Failure was defined as conversion to knee arthroplasty (KA) or total removal of the MAT. Results Mean age at surgery was 33 years (15–57); 60% were males. No-to-mild cartilage damage was found in 156 cases, moderate-to-severe damage in 130. Simultaneous procedures in 118 patients included cartilage procedures, osteotomy or ACL-reconstruction. At a mean follow-up of 6.8 years (0.2–24.3years), 5 patients were deceased and 48 lost (14.6%), 186 MAT were in situ (56.5%) whilst 90 (27.4%) had been removed, including 63 converted to a KA (19.2%). Cumulative allograft survivorship was 15.1% (95% CI:13.9–16.3) at 24.0 years. In patients <35 years at surgery, survival was significantly better (24.1%) compared to ≥35 years (8.0%) (p = 0.017). In knees with no-to-mild cartilage damage more allografts survived (43.0%) compared to moderate-to-severe damage (6.6%) (p = 0.003). Simultaneous osteotomy significantly deteriorated survival (0% at 24.0 years) (p = 0.010). 61% of patients underwent at least one additional surgery (1–11) for clinical symptoms after MAT. Consecutive radiographs showed significant OA progression at a mean of 3.8 years (p<0.0001). Incremental Kellgren-Lawrence grade was +1,1 grade per 1000 days (2,7yrs). Conclusions MAT did not delay or prevent tibiofemoral OA progression. 19.2% were converted to a knee prosthesis at a mean of 10.3 years. Patients younger than 35 with no-to-mild cartilage damage may benefit from MAT for relief of symptoms (survivorship 51.9% at 20.2 years), but patients and healthcare payers and providers should be aware of the high number of surgical re-interventions.
Collapse
Affiliation(s)
- Catherine Van Der Straeten
- Musculoskeletal Sciences and Technology, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- * E-mail:
| | - Paul Byttebier
- Department Orthopaedics and Traumatology, Ghent University Hospital, Ghent, Belgium
| | - Annelies Eeckhoudt
- Department Orthopaedics and Traumatology, Ghent University Hospital, Ghent, Belgium
| | - Jan Victor
- Department Orthopaedics and Traumatology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
38
|
Rai MF, Sandell LJ, Zhang B, Wright RW, Brophy RH. RNA Microarray Analysis of Macroscopically Normal Articular Cartilage from Knees Undergoing Partial Medial Meniscectomy: Potential Prediction of the Risk for Developing Osteoarthritis. PLoS One 2016; 11:e0155373. [PMID: 27171008 PMCID: PMC4865200 DOI: 10.1371/journal.pone.0155373] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/27/2016] [Indexed: 11/24/2022] Open
Abstract
Objectives (i) To provide baseline knowledge of gene expression in macroscopically normal articular cartilage, (ii) to test the hypothesis that age, body-mass-index (BMI), and sex are associated with cartilage RNA transcriptome, and (iii) to predict individuals at potential risk for developing “pre-osteoarthritis” (OA) based on screening of genetic risk-alleles associated with OA and gene transcripts differentially expressed between normal and OA cartilage. Design Healthy-appearing cartilage was obtained from the medial femoral notch of 12 knees with a meniscus tear undergoing arthroscopic partial meniscectomy. Cartilage had no radiographic, magnetic-resonance-imaging or arthroscopic evidence for degeneration. RNA was subjected to Affymetrix microarrays followed by validation of selected transcripts by microfluidic digital polymerase-chain-reaction. The underlying biological processes were explored computationally. Transcriptome-wide gene expression was probed for association with known OA genetic risk-alleles assembled from published literature and for comparison with gene transcripts differentially expressed between healthy and OA cartilage from other studies. Results We generated a list of 27,641 gene transcripts in healthy cartilage. Several gene transcripts representing numerous biological processes were correlated with age and BMI and differentially expressed by sex. Based on disease-specific Ingenuity Pathways Analysis, gene transcripts associated with aging were enriched for bone/cartilage disease while the gene expression profile associated with BMI was enriched for growth-plate calcification and OA. When segregated by genetic risk-alleles, two clusters of study patients emerged, one cluster containing transcripts predicted by risk studies. When segregated by OA-associated gene transcripts, three clusters of study patients emerged, one of which is remarkably similar to gene expression pattern in OA. Conclusions Our study provides a list of gene transcripts in healthy-appearing cartilage. Preliminary analysis into groupings based on OA risk-alleles and OA-associated gene transcripts reveals a subset of patients expressing OA transcripts. Prospective studies in larger cohorts are needed to assess whether these patterns are predictive for OA.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes Jewish Hospital, St. Louis, Missouri, United States of America
- * E-mail:
| | - Linda J. Sandell
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes Jewish Hospital, St. Louis, Missouri, United States of America
- Department of Cell Biology and Physiology, Washington University School of Medicine at Barnes Jewish Hospital, St. Louis, Missouri, United States of America
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Bo Zhang
- Department of Developmental Biology, Center of Regenerative Medicine, Washington University School of Medicine at Barnes Jewish Hospital, St. Louis, Missouri, United States of America
| | - Rick W. Wright
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes Jewish Hospital, St. Louis, Missouri, United States of America
| | - Robert H. Brophy
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine at Barnes Jewish Hospital, St. Louis, Missouri, United States of America
| |
Collapse
|
39
|
Leal MF, Arliani GG, Astur DC, Franciozi CE, Debieux P, Andreoli CV, Smith MC, Pochini ADC, Ejnisman B, Cohen M. Comprehensive selection of reference genes for expression studies in meniscus injury using quantitative real-time PCR. Gene 2016; 584:60-68. [PMID: 26968891 DOI: 10.1016/j.gene.2016.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 02/11/2016] [Accepted: 03/04/2016] [Indexed: 11/29/2022]
Abstract
The meniscus plays critical roles in the knee function. Meniscal tears can lead to knee osteoarthritis. Gene expression analysis may be a useful tool for understanding meniscus tears, and reverse-transcription quantitative polymerase chain reaction (RT-qPCR) has become an effective method for such studies. However, this technique requires the use of suitable reference genes for data normalization. We evaluated the suitability of six reference genes (18S, ACTB, B2M, GAPDH, HPRT1 and TBP) using meniscus samples of (1) 19 patients with isolated meniscal tears, (2) 20 patients with meniscal tears and combined anterior cruciate ligament injury (ACL), and (3) 11 controls without meniscal tears. The stability of the candidate reference genes was determined using the NormFinder, geNorm, BestKeeper DataAssist and RefFinder software packages and comparative ΔCt method. Overall, HPRT1 was the best single reference gene. However, GenEx software demonstrated that two or more reference genes should be used for gene expression normalization, which was confirmed when we evaluated TGFβR1 expression using several reference gene combinations. HPRT1+TBP was the most frequently identified pair from the analysis of samples of (1) meniscal tear samples of patients with a concomitant ACL tears, (2) all meniscal tears, and (3) all samples. HPRT1+GAPDH was the most frequently identified pair from the analysis of samples of isolated meniscal tear samples and controls. In the analysis involving only controls, GAPDH+18S was the most frequently identified pair. In the analysis of only isolated meniscal tear samples and in the analysis of meniscal tear samples of patients with concomitant ACL tears and controls, both HPRT1+TBP and HPRT1+GAPDH were identified as suitable pairs. If the gene expression study aims to compare non-injured meniscus, isolated meniscal tears and meniscal tears of patients with ACL tears as three independent groups, the trio of HPRT1+TBP+GAPDH is the most suitable combination of reference genes.
Collapse
Affiliation(s)
- Mariana Ferreira Leal
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, 04038-032, São Paulo, SP, Brazil; Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, 04023-001, São Paulo, SP, Brazil.
| | - Gustavo Gonçalves Arliani
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, 04038-032, São Paulo, SP, Brazil
| | - Diego Costa Astur
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, 04038-032, São Paulo, SP, Brazil
| | - Carlos Eduardo Franciozi
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, 04038-032, São Paulo, SP, Brazil
| | - Pedro Debieux
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, 04038-032, São Paulo, SP, Brazil
| | - Carlos Vicente Andreoli
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, 04038-032, São Paulo, SP, Brazil
| | - Marília Cardoso Smith
- Disciplina de Genética, Departamento de Morfologia e Genética, Universidade Federal de São Paulo, 04023-001, São Paulo, SP, Brazil
| | - Alberto de Castro Pochini
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, 04038-032, São Paulo, SP, Brazil
| | - Benno Ejnisman
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, 04038-032, São Paulo, SP, Brazil
| | - Moises Cohen
- Departamento de Ortopedia e Traumatologia, Universidade Federal de São Paulo, 04038-032, São Paulo, SP, Brazil
| |
Collapse
|
40
|
Lee HR, Shon OJ, Park SI, Kim HJ, Kim S, Ahn MW, Do SH. Platelet-Rich Plasma Increases the Levels of Catabolic Molecules and Cellular Dedifferentiation in the Meniscus of a Rabbit Model. Int J Mol Sci 2016; 17:ijms17010120. [PMID: 26784189 PMCID: PMC4730361 DOI: 10.3390/ijms17010120] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/24/2015] [Accepted: 01/11/2016] [Indexed: 01/10/2023] Open
Abstract
Despite the susceptibility to frequent intrinsic and extrinsic injuries, especially in the inner zone, the meniscus does not heal spontaneously owing to its poor vascularity. In this study, the effect of platelet-rich plasma (PRP), containing various growth factors, on meniscal mechanisms was examined under normal and post-traumatic inflammatory conditions. Isolated primary meniscal cells of New Zealand white (NZW) rabbits were incubated for 3, 10, 14 and 21 days with PRP(−), 10% PRP (PRP(+)), IL(+) or IL(+)PRP(+). The meniscal cells were collected and examined using reverse-transcription polymerase chain reaction (RT-PCR). Culture media were examined by immunoblot analyses for matrix metalloproteinases (MMP) catabolic molecules. PRP containing growth factors improved the cellular viability of meniscal cells in a concentration-dependent manner at Days 1, 4 and 7. However, based on RT-PCR, meniscal cells demonstrated dedifferentiation, along with an increase in type I collagen in the PRP(+) and in IL(+)PRP(+). In PRP(+), the aggrecan expression levels were lower than in the PRP(−) until Day 21. The protein levels of MMP-1 and MMP-3 were higher in each PRP group, i.e., PRP(+) and IL(+)PRP(+), at each culture time. A reproducible 2-mm circular defect on the meniscus of NZW rabbit was used to implant fibrin glue (control) or PRP in vivo. After eight weeks, the lesions in the control and PRP groups were occupied with fibrous tissue, but not with meniscal cells. This study shows that PRP treatment of the meniscus results in an increase of catabolic molecules, especially those related to IL-1α-induced inflammation, and that PRP treatment for an in vivo meniscus injury accelerates fibrosis, instead of meniscal cartilage.
Collapse
Affiliation(s)
- Hye-Rim Lee
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Korea.
| | - Oog-Jin Shon
- Department of Orthopedic Surgery, College of Medicine, Yeungnam University, Daegu 705-717, Korea.
| | - Se-Il Park
- Cardiovascular Product Evaluation Center, College of Medicine, Yonsei University, Seoul 120-752, Korea.
| | - Han-Jun Kim
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Korea.
| | - Sukyoung Kim
- School of Materials Science and Engineering, Yeungnam University, Gyeongsan 712-749, Korea.
| | - Myun-Whan Ahn
- Department of Orthopedic Surgery, College of Medicine, Yeungnam University, Daegu 705-717, Korea.
| | - Sun Hee Do
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Korea.
| |
Collapse
|
41
|
Osteoarthritic changes in vervet monkey knees correlate with meniscus degradation and increased matrix metalloproteinase and cytokine secretion. Osteoarthritis Cartilage 2015; 23:1780-9. [PMID: 26033163 PMCID: PMC4642681 DOI: 10.1016/j.joca.2015.05.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 05/19/2015] [Accepted: 05/21/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Meniscus injury increases osteoarthritis risk but its pathobiology in osteoarthritis is unclear. We hypothesized that older adult vervet monkeys would exhibit knee osteoarthritic changes and the degenerative menisci from these animals would secrete matrix metalloproteinases (MMPs) and pro-inflammatory cytokines that contribute to the development of osteoarthritis. DESIGN In a cross sectional analysis of healthy young adult (9-12 years) and old (19-26 years) adult female vervet monkeys, knees were evaluated in vivo with computed tomography (CT) imaging, and joint tissues were morphologically graded at necropsy. Meniscus explants were subsequently cultured to evaluate meniscal MMP and cytokine secretion. RESULTS CT images revealed significant bony osteoarthritic changes in 80% of older monkeys which included increases in osteophyte number and meniscal calcification. Meniscus and cartilage degradation scores were greater in the older monkeys and were positively correlated (r > 0.7). Menisci from older animals exhibiting osteoarthritic changes secreted significantly more MMP-1, MMP-3, and MMP-8 than healthy menisci from younger monkeys. Older menisci without significant osteoarthritic changes secreted more IL-7 than healthy young menisci while older osteoarthritic menisci secreted more IL-7 and granulocyte-macrophage colony-stimulating factor than healthy older menisci. CONCLUSIONS Aged vervets develop naturally occurring knee osteoarthritis that includes involvement of the meniscus. Degenerative menisci secreted markedly increased amounts of matrix-degrading enzymes and inflammatory cytokines. These factors would be expected to act on the meniscus tissue and local joint tissues and may ultimately promote osteoarthritis development. These finding also suggest vervet monkeys are a useful animal model for studying the progression of osteoarthritis.
Collapse
|
42
|
Brophy RH, Haas AK, Huston LJ, Nwosu SK, Wright RW, Wright RW. Association of Meniscal Status, Lower Extremity Alignment, and Body Mass Index With Chondrosis at Revision Anterior Cruciate Ligament Reconstruction. Am J Sports Med 2015; 43:1616-22. [PMID: 25899434 PMCID: PMC4490131 DOI: 10.1177/0363546515578838] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Knees undergoing revision anterior cruciate ligament reconstruction (rACLR) have a high prevalence of articular cartilage lesions. HYPOTHESIS The prevalence of chondrosis at the time of rACLR is associated with meniscal status and lower extremity alignment. STUDY DESIGN Cross-sectional study; Level of evidence, 3. METHODS Data from the prospective Multicenter ACL Revision Study (MARS) cohort were reviewed to identify patients with preoperative lower extremity alignment films. Lower extremity alignment was defined by the weightbearing line (WBL) as a percentage of the tibial plateau width, while the chondral and meniscal status of each weightbearing compartment was recorded at the time of surgery. Multivariable proportional odds models were constructed and adjusted for relevant factors to examine which risk factors were independently associated with the degree of medial and lateral compartment chondrosis. RESULTS The cohort included 246 patients with lower extremity alignment films at the time of rACLR. Mean (±SD) patient age was 26.9 ± 9.5 years and body mass index (BMI) was 26.4 ± 4.6. The medial compartment had more chondrosis (grade 2/3, 42%; grade 4, 6.5%) than did the lateral compartment (grade 2/3, 26%; grade 4, 6.5%). Disruption of the meniscus was noted in 35% of patients on the medial side and 16% in the lateral side. The mean WBL was 0.43 ± 0.13. Medial compartment chondrosis was associated with BMI (P = .025), alignment (P = .002), and medial meniscal status (P = .001). None of the knees with the WBL lateral to 0.625 had grade 4 chondrosis in the medial compartment. Lateral compartment chondrosis was significantly associated with age (P = .013) and lateral meniscal status (P < .001). Subjects with "intact" menisci were found to decrease their odds of having chondrosis by 64% to 84%. CONCLUSION The status of articular cartilage in the tibiofemoral compartments at the time of rACLR is related to meniscal status. Lower extremity alignment and BMI are associated with medial compartment chondrosis.
Collapse
|
43
|
Emerging targets in osteoarthritis therapy. Curr Opin Pharmacol 2015; 22:51-63. [PMID: 25863583 DOI: 10.1016/j.coph.2015.03.004] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/17/2015] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is a destructive joint disease in which the initiation may be attributed to direct injury and mechanical disruption of joint tissues, but the progressive changes are dependent on active cell-mediated processes that can be observed or inferred during the generally long time-course of the disease. Based on clinical observations and experimental studies, it is now recognized a that it is possible for individual patients to exhibit common sets of symptoms and structural abnormalities due to distinct pathophysiological pathways that act independently or in combination. Recent research that has focused on the underlying mechanisms involving biochemical cross talk among the cartilage, synovium, bone, and other joint tissues within a background of poorly characterized genetic factors will be addressed in this review.
Collapse
|
44
|
Expression of phosphocitrate-targeted genes in osteoarthritis menisci. BIOMED RESEARCH INTERNATIONAL 2014; 2014:210469. [PMID: 25525593 PMCID: PMC4265372 DOI: 10.1155/2014/210469] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/11/2014] [Accepted: 09/15/2014] [Indexed: 01/19/2023]
Abstract
Phosphocitrate (PC) inhibited calcium crystal-associated osteoarthritis (OA) in Hartley guinea pigs. However, the molecular mechanisms remain elusive. This study sought to determine PC targeted genes and the expression of select PC targeted genes in OA menisci to test hypothesis that PC exerts its disease modifying activity in part by reversing abnormal expressions of genes involved in OA. We found that PC downregulated the expression of numerous genes classified in immune response, inflammatory response, and angiogenesis, including chemokine (C-C motif) ligand 5, Fc fragment of IgG, low affinity IIIb receptor (FCGR3B), and leukocyte immunoglobulin-like receptor, subfamily B member 3 (LILRB3). In contrast, PC upregulated the expression of many genes classified in skeletal development, including collagen type II alpha1, fibroblast growth factor receptor 3 (FGFR3), and SRY- (sex determining region Y-) box 9 (SOX-9). Immunohistochemical examinations revealed higher levels of FCGR3B and LILRB3 and lower level of SOX-9 in OA menisci. These findings indicate that OA is a disease associated with immune system activation and decreased expression of SOX-9 gene in OA menisci. PC exerts its disease modifying activity on OA, at least in part, by targeting immune system activation and the production of extracellular matrix and selecting chondroprotective proteins.
Collapse
|
45
|
Human migratory meniscus progenitor cells are controlled via the TGF-β pathway. Stem Cell Reports 2014; 3:789-803. [PMID: 25418724 PMCID: PMC4235742 DOI: 10.1016/j.stemcr.2014.08.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 12/29/2022] Open
Abstract
Degeneration of the knee joint during osteoarthritis often begins with meniscal lesions. Meniscectomy, previously performed extensively after meniscal injury, is now obsolete because of the inevitable osteoarthritis that occurs following this procedure. Clinically, meniscus self-renewal is well documented as long as the outer, vascularized meniscal ring remains intact. In contrast, regeneration of the inner, avascular meniscus does not occur. Here, we show that cartilage tissue harvested from the avascular inner human meniscus during the late stages of osteoarthritis harbors a unique progenitor cell population. These meniscus progenitor cells (MPCs) are clonogenic and multipotent and exhibit migratory activity. We also determined that MPCs are likely to be controlled by canonical transforming growth factor β (TGF-β) signaling that leads to an increase in SOX9 and a decrease in RUNX2, thereby enhancing the chondrogenic potential of MPC. Therefore, our work is relevant for the development of novel cell biological, regenerative therapies for meniscus repair. Progenitor cells are found in the inner avascular part of human osteoarthritic menisci These meniscus progenitor cells (MPCs) are clonogenic, migratory, and multipotent MPCs are governed via the canonical TGF-β pathway TGF-β3 via Smad2 reduces Runx2 to enhance the chondrogenic potential of MPCs
Collapse
|
46
|
Rai MF, Patra D, Sandell LJ, Brophy RH. Relationship of gene expression in the injured human meniscus to body mass index: a biologic connection between obesity and osteoarthritis. Arthritis Rheumatol 2014; 66:2152-64. [PMID: 24692131 DOI: 10.1002/art.38643] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 03/20/2014] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Higher body mass index (BMI) increases the risk of meniscus injury and knee osteoarthritis (OA). However, it is unknown if and how obesity affects meniscus biology. We analyzed transcriptome-wide gene expression profiles of injured human menisci to test the hypothesis that meniscal gene expression signatures relate to patient BMI. METHODS Meniscus samples were obtained from patients undergoing arthroscopic partial meniscectomy. Transcriptome-wide analysis of gene expression followed by validation of selected transcripts by QuantiGene Plex assay was performed. Correlations of gene expression with BMI and relative fold changes (≥1.5-fold) in 3 BMI categories (lean [BMI 18.5-24.9 kg/m(2) ], overweight [BMI 25.0-29.9 kg/m(2) ], and obese [BMI >30.0 kg/m(2) ]) were analyzed, and integrated functional classifications were probed computationally. RESULTS The obese versus overweight comparison resulted in the largest set of differences (565 transcripts) followed by obese versus lean (280 transcripts) and overweight versus lean (125 transcripts). Biologic reproducibility was confirmed by cluster analysis of expressed transcripts. Differentially regulated transcripts represented important functional classifications. Transcripts associated with oxygen transport, calcium ion binding, and cell homeostasis were elevated with BMI, while those related to extracellular matrix deposition, cell migration, and glucosamine metabolic processes were repressed. While these functional classifications may play key roles in cartilage/meniscus homeostasis, failure of extracellular matrix deposition and increase in calcium ion binding likely contribute to OA development following meniscal injury. CONCLUSION Our results indicate greater differences in gene expression between obese and overweight groups than between overweight and lean groups. This may indicate that there is a weight threshold at which injured meniscus responds severely to increased BMI. BMI-related changes in gene expression present a plausible explanation for the role of meniscal injury in OA development among obese patients.
Collapse
Affiliation(s)
- Muhammad Farooq Rai
- Washington University School of Medicine at Barnes-Jewish Hospital, St. Louis, Missouri
| | | | | | | |
Collapse
|
47
|
Molecular insight into the association between cartilage regeneration and ear wound healing in genetic mouse models: targeting new genes in regeneration. G3-GENES GENOMES GENETICS 2013; 3:1881-91. [PMID: 24002865 PMCID: PMC3815053 DOI: 10.1534/g3.113.007302] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tissue regeneration is a complex trait with few genetic models available. Mouse strains LG/J and MRL are exceptional healers. Using recombinant inbred strains from a large (LG/J, healer) and small (SM/J, nonhealer) intercross, we have previously shown a positive genetic correlation between ear wound healing, knee cartilage regeneration, and protection from osteoarthritis. We hypothesize that a common set of genes operates in tissue healing and articular cartilage regeneration. Taking advantage of archived histological sections from recombinant inbred strains, we analyzed expression of candidate genes through branched-chain DNA technology directly from tissue lysates. We determined broad-sense heritability of candidates, Pearson correlation of candidates with healing phenotypes, and Ward minimum variance cluster analysis for strains. A bioinformatic assessment of allelic polymorphisms within and near candidate genes was also performed. The expression of several candidates was significantly heritable among strains. Although several genes correlated with both ear wound healing and cartilage healing at a marginal level, the expression of four genes representing DNA repair (Xrcc2, Pcna) and Wnt signaling (Axin2, Wnt16) pathways was significantly positively correlated with both phenotypes. Cluster analysis accurately classified healers and nonhealers for seven out of eight strains based on gene expression. Specific sequence differences between LG/J and SM/J were identified as potential causal polymorphisms. Our study suggests a common genetic basis between tissue healing and osteoarthritis susceptibility. Mapping genetic variations causing differences in diverse healing responses in multiple tissues may reveal generic healing processes in pursuit of new therapeutic targets designed to induce or enhance regeneration and, potentially, protection from osteoarthritis.
Collapse
|