1
|
Christiansen BA, Lin YY, Lee CA. Response to the commentary on "Surgical restabilization reduces the progression of post-traumatic osteoarthritis initiated by ACL rupture in mice". Osteoarthritis Cartilage 2025; 33:656-657. [PMID: 39952346 DOI: 10.1016/j.joca.2025.02.772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Affiliation(s)
| | - Yu-Yang Lin
- University of California Davis Health, Department of Orthopaedic Surgery, USA.
| | - Cassandra A Lee
- University of California Davis Health, Department of Orthopaedic Surgery, USA.
| |
Collapse
|
2
|
Frost MR, Ball BK, Pendyala M, Douglas SR, Brubaker DK, Chan DD. Computational Translation of Mouse Models of Osteoarthritis Predicts Human Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639777. [PMID: 40060529 PMCID: PMC11888325 DOI: 10.1101/2025.02.23.639777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Objective Translation of biological insights from preclinical studies to human disease is a pressing challenge in biomedical research, including in osteoarthritis. Translatable Components Regression (TransComp-R) is a computational framework that has previously been used to synthesize preclinical and human OA data to identify biological pathways predictive of human disease conditions. We aimed to evaluate the translatability of two common murine models of post-traumatic osteoarthritis - surgical destabilization of the medial meniscus (DMM) and noninvasive anterior cruciate ligament rupture (ACLR) - to transcriptomics cartilage data from human OA outcomes. Design Transcriptomics cartilage data of DMM and ACLR mouse and human data was acquired from Gene Expression Omnibus. TransComp-R was used to project human OA data into a mouse model (DMM or ACLR) principal component analysis space. The principal components (PCs) were regressed against human OA conditions using increasing complexity of linear regression models incorporating human demographic covariates of OA, sex, and age. Biological pathways of the mouse PCs that significantly stratified human OA and control groups were then interpreted using Gene Set Enrichment Analysis. Results From the TransComp-R model, we identified different enriched biological pathways across DMM and ACLR models. While PCs among the DMM models revealed pathways associated with cell signaling and metabolism, ACLR PCs represented immune function and cellular pathways associated with OA condition. The immune pathways presented in the ACLR further highlighted the potential relevance of the OA pathways observed in human conditions. Conclusions The ACLR mouse model more successfully predicted human OA conditions, particularly with the human control groups without a history of joint injury or disease. Cross-species translational approaches support the selection of preclinical models intended for therapeutic discovery and pathway analysis in humans.
Collapse
Affiliation(s)
- Maya R Frost
- Weldon School of Biomedical Engineering, Purdue University
| | - Brendan K Ball
- Weldon School of Biomedical Engineering, Purdue University
| | - Meghana Pendyala
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute
| | | | - Douglas K Brubaker
- Center for Global Health and Diseases, Department of Pathology, School of Medicine, Case Western Reserve University
- Blood Heart Lung Immunology Research Center, University Hospitals Cleveland Medical Center
| | - Deva D Chan
- Weldon School of Biomedical Engineering, Purdue University
- School of Mechanical Engineering, Purdue University
| |
Collapse
|
3
|
Chun JM, Park JH, Moon BC, Baek SJ. Transcriptomic insights into the anti-inflammatory mechanisms of Protaetia brevitarsis seulensis larvae in IL-1β-driven chondrosarcoma cells. Biomed Pharmacother 2025; 183:117866. [PMID: 39862704 DOI: 10.1016/j.biopha.2025.117866] [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: 11/12/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025] Open
Abstract
Osteoarthritis (OA) is a complex, degenerative, multi-factorial joint disease. Because of the difficulty in treating OA, developing new targeting strategies that can be used to understand its molecular mechanisms is critical. Protaetia brevitarsis seulensis larvae offer much therapeutic value; however, the presence of various active compounds and the multi-factorial risk factors for OA render the precise mechanisms of action unclear. A systematic transcriptome analysis was used to investigate the key mechanisms of action of P. brevitarsis seulensis larvae aqueous extract (PBSL) and its compounds on OA. Major mechanisms and transcription factors of PBSL were analyzed by profiling gene expression changes in interleukin (IL)-1β-induced human chondrosarcoma cell (SW1353) treated with PBSL. An in vitro assay was performed to validate the efficacy of the novel mechanism and targets of PBSL. PBSL exerted anti-inflammatory effects on SW1353 cells by regulating many molecular pathways. The IL-6/JAK/STAT3 pathway was significantly downregulated by PBSL, and STAT3 was identified as a major transcription factor regulating PBSL-induced target gene expression. Of the six PBSL compounds, the major compound was regulated by the IL-6/JAK/STAT3 pathway. This study provided potential novel mechanisms and transcription factors for PBSL and its active compounds against OA and indicated that inhibiting the IL-6/JAK/STAT3 pathway is a therapeutic target for treating OA.
Collapse
Affiliation(s)
- Jin Mi Chun
- Digital Health Research Division, Korean Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korean Institute of Oriental Medicine, Naju 58245, Republic of Korea
| | - Byeong Cheol Moon
- Herbal Medicine Resources Research Center, Korean Institute of Oriental Medicine, Naju 58245, Republic of Korea
| | - Su-Jin Baek
- Korean Medicine Data Division, Korean Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| |
Collapse
|
4
|
McCool JL, Sebastian A, Hum NR, Wilson SP, Davalos OA, Murugesh DK, Amiri B, Morfin C, Christiansen BA, Loots GG. CD206+ Trem2+ macrophage accumulation in the murine knee joint after injury is associated with protection against post-traumatic osteoarthritis in MRL/MpJ mice. PLoS One 2025; 20:e0312587. [PMID: 39752388 PMCID: PMC11698337 DOI: 10.1371/journal.pone.0312587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/09/2024] [Indexed: 01/06/2025] Open
Abstract
Post-traumatic osteoarthritis (PTOA) is a painful joint disease characterized by the degradation of bone, cartilage, and other connective tissues in the joint. PTOA is initiated by trauma to joint-stabilizing tissues, such as the anterior cruciate ligament, medial meniscus, or by intra-articular fractures. In humans, ~50% of joint injuries progress to PTOA, while the rest spontaneously resolve. To better understand molecular programs contributing to PTOA development or resolution, we examined injury-induced fluctuations in immune cell populations and transcriptional shifts by single-cell RNA sequencing of synovial joints in PTOA-susceptible C57BL/6J (B6) and PTOA-resistant MRL/MpJ (MRL) mice. We identified significant differences in monocyte and macrophage subpopulations between MRL and B6 joints. A potent myeloid-driven anti-inflammatory response was observed in MRL injured joints that significantly contrasted the pro-inflammatory signaling seen in B6 joints. Multiple CD206+ macrophage populations classically described as M2 were found enriched in MRL injured joints. These CD206+ macrophages also robustly expressed Trem2, a receptor involved in inflammation and myeloid cell activation. These data suggest that the PTOA resistant MRL mouse strain displays an enhanced capacity of clearing debris and apoptotic cells induced by inflammation after injury due to an increase in activated M2 macrophages within the synovial tissue and joint space.
Collapse
Affiliation(s)
- Jillian L. McCool
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
- School of Natural Sciences, University of California Merced, Merced, CA, United States of America
| | - Aimy Sebastian
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Stephen P. Wilson
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Oscar A. Davalos
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Deepa K. Murugesh
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Beheshta Amiri
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
| | - Cesar Morfin
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States of America
| | - Blaine A. Christiansen
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States of America
| | - Gabriela G. Loots
- Lawrence Livermore National Laboratory, Physical and Life Science Directorate, Livermore, CA, United States of America
- School of Natural Sciences, University of California Merced, Merced, CA, United States of America
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States of America
| |
Collapse
|
5
|
Timkovich AE, Holling GA, Afzali MF, Kisiday J, Santangelo KS. TLR4 antagonism provides short-term but not long-term clinical benefit in a full-depth cartilage defect mouse model. Connect Tissue Res 2024; 65:26-40. [PMID: 37898909 PMCID: PMC11271750 DOI: 10.1080/03008207.2023.2269257] [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: 12/14/2022] [Revised: 08/23/2023] [Accepted: 10/05/2023] [Indexed: 10/31/2023]
Abstract
PURPOSE/AIM Cartilage injury and subsequent osteoarthritis (OA) are debilitating conditions affecting millions worldwide. As there are no cures for these ailments, novel therapies are needed to suppress disease pathogenesis. Given that joint injuries are known to produce damage-associated molecular patterns (DAMPs), our central premise is that the Toll-like receptor 4 (TLR4) pathway is a principal driver in the early response to cartilage damage and subsequent pathology. We postulate that TLR4 activation is initiated/perpetuated by DAMPs released following joint damage. Thus, antagonism of the TLR4 pathway immediately after injury may suppress the development of joint surface defects. MATERIALS AND METHODS Two groups were utilized: (1) 8-week-old, male C57BL6 mice treated systemically with a known TLR4 antagonist and (2) mice injected with vehicle control. A full-depth cartilage lesion on the midline of the patellofemoral groove was created in the right knee of each mouse. The left knee was used as a sham surgery control. Gait changes were evaluated over 4 weeks using a quantitative gait analysis system. At harvest, knee joints were processed for pathologic assessment, Nanostring® transcript expression, and immunohistochemistry (IHC). RESULTS Short-term treatment with a TLR4 antagonist at 14-days significantly improved relevant gait parameters; improved cartilage metrics and modified Mankin scores were also seen. Additionally, mRNA expression and IHC showed reduced expression of inflammatory mediators in animals treated with the TLR4 antagonist. CONCLUSIONS Collectively, this work demonstrates that systemic treatment with a TLR4 antagonist is protective to further cartilage damage 14-days post-injury in a murine model of induced disease.
Collapse
Affiliation(s)
- Ariel E. Timkovich
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - G. Aaron Holling
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Maryam F. Afzali
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - John Kisiday
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Kelly S. Santangelo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
6
|
Mendez ME, Murugesh DK, Christiansen BA, Loots GG. Antibiotic Treatment Prior to Injury Abrogates the Detrimental Effects of LPS in STR/ort Mice Susceptible to Osteoarthritis Development. JBMR Plus 2023; 7:e10759. [PMID: 37614305 PMCID: PMC10443070 DOI: 10.1002/jbm4.10759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 08/25/2023] Open
Abstract
Post traumatic osteoarthritis (PTOA) is a form of secondary osteoarthritis (OA) that develops in ~50% of cases of severe articular joint injuries and leads to chronic and progressive degradation of articular cartilage and other joint tissues. PTOA progression can be exacerbated by repeated injury and systemic inflammation. Few studies have examined approaches for blunting or slowing down PTOA progression with emphasis on systemic inflammation; most arthritis studies focused on the immune system have been in the context of rheumatoid arthritis. To examine how the gut microbiome affects systemic inflammation during PTOA development, we used a chronic antibiotic treatment regimen starting at weaning for 6 weeks before anterior cruciate ligament (ACL) rupture in STR/ort mice combined with lipopolysaccharide (LPS)-induced systemic inflammation. STR/ort mice develop spontaneous OA as well as a more severe PTOA phenotype than C57Bl/6J mice. By 6 weeks post injury, histological examination showed a more robust cartilage staining in the antibiotic-treated (AB) STR/ort mice than in the untreated STR/ort controls. Furthermore, we also examined the effects of AB treatment on systemic inflammation and found that the effects of LPS administration before injury are also blunted by AB treatment in STR/ort mice. The AB- or AB+LPS-treated STR/ort injured joints more closely resembled the C57Bl/6J VEH OA phenotypes than the vehicle- or LPS-treated STR/ort, suggesting that antibiotic treatment has the potential to slow disease progression and should be further explored therapeutically as prophylactic post injury. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Melanie E Mendez
- Lawrence Livermore National Laboratories, Physical and Life Sciences DirectorateLivermoreCAUSA
| | - Deepa K Murugesh
- Lawrence Livermore National Laboratories, Physical and Life Sciences DirectorateLivermoreCAUSA
| | - Blaine A Christiansen
- Department of Orthopaedic SurgeryUniversity of California Davis HealthSacramentoCAUSA
| | - Gabriela G Loots
- Lawrence Livermore National Laboratories, Physical and Life Sciences DirectorateLivermoreCAUSA
- Department of Orthopaedic SurgeryUniversity of California Davis HealthSacramentoCAUSA
| |
Collapse
|
7
|
Husin NA, Rahman S, Karunakaran R, Bhore SJ. Transcriptome analysis during fruit developmental stages in durian (Durio zibethinus Murr.) var. D24. Genet Mol Biol 2023; 45:e20210379. [PMID: 36622241 PMCID: PMC9830936 DOI: 10.1590/1678-4685-gmb-2021-0379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 10/18/2022] [Indexed: 01/10/2023] Open
Abstract
Durian (Durio zibethinus Murr.) fruits are famous for their unique aroma. This study analysed the Durian fruit transcriptome to discover the expression patterns of genes and to understand their regulation. Three developmental stages of Durian fruit, namely, early [90 days post-anthesis (DPA)], mature (120 DPA), and ripen (127 DPA), were studied. The Illumina HiSeq platform was used for sequencing. The sequence data were analysed using four different mapping aligners and statistical methods: CLC Genomic Workbench, HISAT2+DESeq2, Tophat+Cufflinks, and HISAT2+edgeR. The analyses showed that over 110 million clean reads were mapped to the Durian genome, yielding 19,976, 11,394, 17,833, and 24,351 differentially expressed genes during 90-127 days post-anthesis. Many identified differentially expressed genes were linked to the fruit ripening processes. The data analysis suggests that most genes with increased expression at the ripening stage were primarily involved in the metabolism of cofactors and vitamins, nucleotide metabolism, and carbohydrate metabolism. Significantly expressed genes from the young to mature stage were mainly associated with carbohydrate metabolism, amino acid metabolism, and cofactor and vitamin metabolism. The transcriptome data will serve as a foundation for understanding Durian fruit development-specific genes and could be helpful in fruit's trait improvement.
Collapse
Affiliation(s)
- Nurul Arneida Husin
- AIMST University, Faculty of Applied Sciences, Department of Biotechnology, Kedah, Malaysia.,Monash University Malaysia, Jeffrey Cheah School of Medicine and Health Sciences, Malaysia
| | - Sadequr Rahman
- Monash University Malaysia, School of Science and Tropical Medicine and Biology Platform, Malaysia.
| | - Rohini Karunakaran
- AIMST University, Faculty of Medicine, Unit of Biochemistry, Kedah, Malaysia.,Institute of Bioinformatics, Saveetha School of Engineering, Department of Computational Biology, Chennai, India
| | | |
Collapse
|
8
|
Gavile CM, Kazmers NH, Novak KA, Meeks HD, Yu Z, Thomas JL, Hansen C, Barker T, Jurynec MJ. Familial Clustering and Genetic Analysis of Severe Thumb Carpometacarpal Joint Osteoarthritis in a Large Statewide Cohort. J Hand Surg Am 2022; 47:923-933. [PMID: 36184273 PMCID: PMC9547951 DOI: 10.1016/j.jhsa.2022.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/29/2022] [Accepted: 08/04/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE Our goals were to identify individuals who required surgery for thumb carpometacarpal (CMC) joint osteoarthritis (OA), determine if CMC joint OA clusters in families, define the magnitude of familial risk of CMC joint OA, identify risk factors associated with CMC joint OA, and identify rare genetic variants that segregate with familial CMC joint OA. METHODS We searched the Utah Population Database to identify a cohort of CMC joint OA patients who required surgery. Affected individuals were mapped to pedigrees to identify high-risk families with excess clustering of CMC joint OA. Cox regression models were used to calculate familial risk of CMC joint OA in related individuals. Risk factors were evaluated using logistic regression models. Whole exome sequencing was used to identify rare coding variants associated with familial CMC joint OA. RESULTS We identified 550 pedigrees with excess clustering of severe CMC joint OA. The relative risk of CMC joint OA requiring surgical treatment was elevated significantly in first- and third-degree relatives of affected individuals, and significant associations with advanced age, female sex, obesity, and tobacco use were observed. We discovered candidate genes that dominantly segregate with severe CMC joint OA in 4 independent families, including a rare variant in Chondroitin Sulfate Synthase 3 (CHSY3). CONCLUSIONS Familial clustering of severe CMC joint OA was observed in a statewide population. Our data indicate that genetic and environmental factors contribute to the disease process, further highlighting the multifactorial nature of the disease. Genomic analyses suggest distinct biological processes are involved in CMC joint OA pathogenesis. CLINICAL RELEVANCE Awareness of associated comorbidities may guide the diagnosis of CMC joint OA in at-risk populations and help identify individuals who may not do well with nonoperative treatment. Further pursuit of the genes associated with severe CMC joint OA may lead to assays for detection of early stages of disease and have therapeutic potential.
Collapse
Affiliation(s)
| | | | - Kendra A Novak
- Department of Orthopaedics, University of Utah, Salt Lake City, UT
| | - Huong D Meeks
- Huntsman Cancer Institute, Utah Population Database, University of Utah, Salt Lake City, UT
| | - Zhe Yu
- Huntsman Cancer Institute, Utah Population Database, University of Utah, Salt Lake City, UT
| | - Joy L Thomas
- Intermountain Healthcare, Precision Genomics, St. George, UT
| | - Channing Hansen
- Intermountain Healthcare, Biorepository, South Salt Lake City, UT
| | - Tyler Barker
- Department of Orthopaedics, University of Utah, Salt Lake City, UT; Intermountain Healthcare, Precision Genomics, Murray, UT; Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT
| | - Michael J Jurynec
- Department of Orthopaedics, University of Utah, Salt Lake City, UT; Department of Human Genetics, University of Utah, Salt Lake City, UT.
| |
Collapse
|
9
|
Sebastian A, Hum NR, McCool JL, Wilson SP, Murugesh DK, Martin KA, Rios-Arce ND, Amiri B, Christiansen BA, Loots GG. Single-cell RNA-Seq reveals changes in immune landscape in post-traumatic osteoarthritis. Front Immunol 2022; 13:938075. [PMID: 35967299 PMCID: PMC9373730 DOI: 10.3389/fimmu.2022.938075] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/06/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is the most common joint disease, affecting over 300 million people world-wide. Accumulating evidence attests to the important roles of the immune system in OA pathogenesis. Understanding the role of various immune cells in joint degeneration or joint repair after injury is vital for improving therapeutic strategies for treating OA. Post-traumatic osteoarthritis (PTOA) develops in ~50% of individuals who have experienced an articular trauma like an anterior cruciate ligament (ACL) rupture. Here, using the high resolution of single-cell RNA sequencing, we delineated the temporal dynamics of immune cell accumulation in the mouse knee joint after ACL rupture. Our study identified multiple immune cell types in the joint including neutrophils, monocytes, macrophages, B cells, T cells, NK cells and dendritic cells. Monocytes and macrophage populations showed the most dramatic changes after injury. Further characterization of monocytes and macrophages reveled 9 major subtypes with unique transcriptomics signatures, including a tissue resident Lyve1hiFolr2hi macrophage population and Trem2hiFcrls+ recruited macrophages, both showing enrichment for phagocytic genes and growth factors such as Igf1, Pdgfa and Pdgfc. We also identified several genes induced or repressed after ACL injury in a cell type-specific manner. This study provides new insight into PTOA-associated changes in the immune microenvironment and highlights macrophage subtypes that may play a role in joint repair after injury.
Collapse
Affiliation(s)
- Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- *Correspondence: Aimy Sebastian, ; Gabriela G. Loots,
| | - Nicholas R. Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Jillian L. McCool
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- School of Natural Sciences, University of California Merced, Merced, CA, United States
| | - Stephen P. Wilson
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Deepa K. Murugesh
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Kelly A. Martin
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Naiomy Deliz Rios-Arce
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Beheshta Amiri
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Blaine A. Christiansen
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, United States
| | - Gabriela G. Loots
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, United States
- School of Natural Sciences, University of California Merced, Merced, CA, United States
- *Correspondence: Aimy Sebastian, ; Gabriela G. Loots,
| |
Collapse
|
10
|
Rios‐Arce ND, Murugesh DK, Hum NR, Sebastian A, Jbeily EH, Christiansen BA, Loots GG. Pre‐existing Type 1 Diabetes Mellitus Blunts the Development of
Post‐Traumatic
Osteoarthritis. JBMR Plus 2022; 6:e10625. [PMID: 35509635 PMCID: PMC9059474 DOI: 10.1002/jbm4.10625] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/26/2022] [Accepted: 03/09/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Naiomy D. Rios‐Arce
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Deepa K. Murugesh
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Nicholas R. Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
| | - Elias H. Jbeily
- Department of Orthopedic Surgery UC Davis Medical Center Sacramento CA USA
| | | | - Gabriela G. Loots
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories Livermore CA USA
- Molecular and Cell Biology School of Natural Sciences, UC Merced Merced CA USA
| |
Collapse
|
11
|
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
|
12
|
Turlo AJ, McDermott BT, Barr ED, Riggs CM, Boyde A, Pinchbeck GL, Clegg PD. Gene expression analysis of subchondral bone, cartilage, and synovium in naturally occurring equine palmar/plantar osteochondral disease. J Orthop Res 2022; 40:595-603. [PMID: 33993513 DOI: 10.1002/jor.25075] [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: 10/19/2020] [Revised: 04/23/2021] [Accepted: 05/03/2021] [Indexed: 02/04/2023]
Abstract
Osteoarthritis (OA) is a disease of the entire joint but the relationship between pathological events in various joint tissues is poorly understood. We examined concurrent changes in bone, cartilage, and synovium in a naturally occurring equine model of joint degeneration. Joints (n = 64) were grossly assessed for palmar/plantar osteochondral disease (POD) in racehorses that required euthanasia for unrelated reasons and assigned a grade of 0 (n = 34), 1 (n = 17), 2 or 3 (n = 13) using a recognized grading scheme. Synovium, cartilage, and subchondral bone were collected for histological and gene expression analysis. Relations between POD grade, cartilage histological score, and gene expression levels were examined using one-way analysis of variance or Kruskal-Wallis test and Spearman's correlation coefficient with corrections for multiple comparisons. Cartilage histological score increased in joints with POD grade 1 (p = 0.002) and 2 or 3 (p < 0.001) compared to 0. At grade 1, expression of COL1A1, COL2A1, and MMP1 increased and BGN decreased in subchondral bone while expression of BGN and ACAN decreased in cartilage. These changes further progressed at grades 2 and 3. POD grades 2 and 3 were associated with decreased expression of osteoclast inhibitor OPG and increased markers of cartilage degeneration (MMP13, COL1A1). Expression of the vascular endothelial growth factor decreased with POD grade and negatively correlated with cartilage histological score. Synovium showed no histological or transcriptomic changes related to pathology grade. Cartilage degeneration in POD is likely to be secondary to remodeling of the subchondral bone. Limited activation of proinflammatory and catabolic genes and moderate synovial pathology suggests distinct molecular phenotype of POD compared with OA.
Collapse
Affiliation(s)
- Agnieszka J Turlo
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Benjamin T McDermott
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | | | - Chris M Riggs
- Department of Veterinary Clinical Services, Hong Kong Jockey Club, Sha Tin Racecourse, New Territories, Hong Kong SAR, China
| | - Alan Boyde
- Dental Physical Sciences, Oral BioEngineering, Queen Mary University of London, Mile End Campus, London, UK
| | - Gina L Pinchbeck
- Department of Epidemiology and Population Health, Institute of Infection and Global Health, School of Veterinary Science, University of Liverpool, Liverpool, UK
| | - Peter D Clegg
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
13
|
Kallenbach JG, Freeberg MAT, Abplanalp D, Alenchery RG, Ajalik RE, Muscat S, Myers JA, Ashton JM, Loiselle A, Buckley MR, van Wijnen AJ, Awad HA. Altered TGFB1 regulated pathways promote accelerated tendon healing in the superhealer MRL/MpJ mouse. Sci Rep 2022; 12:3026. [PMID: 35194136 PMCID: PMC8863792 DOI: 10.1038/s41598-022-07124-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 02/11/2022] [Indexed: 12/23/2022] Open
Abstract
To better understand the molecular mechanisms of tendon healing, we investigated the Murphy Roth's Large (MRL) mouse, which is considered a model of mammalian tissue regeneration. We show that compared to C57Bl/6J (C57) mice, injured MRL tendons have reduced fibrotic adhesions and cellular proliferation, with accelerated improvements in biomechanical properties. RNA-seq analysis revealed that differentially expressed genes in the C57 healing tendon at 7 days post injury were functionally linked to fibrosis, immune system signaling and extracellular matrix (ECM) organization, while the differentially expressed genes in the MRL injured tendon were dominated by cell cycle pathways. These gene expression changes were associated with increased α-SMA+ myofibroblast and F4/80+ macrophage activation and abundant BCL-2 expression in the C57 injured tendons. Transcriptional analysis of upstream regulators using Ingenuity Pathway Analysis showed positive enrichment of TGFB1 in both C57 and MRL healing tendons, but with different downstream transcriptional effects. MRL tendons exhibited of cell cycle regulatory genes, with negative enrichment of the cell senescence-related regulators, compared to the positively-enriched inflammatory and fibrotic (ECM organization) pathways in the C57 tendons. Serum cytokine analysis revealed decreased levels of circulating senescence-associated circulatory proteins in response to injury in the MRL mice compared to the C57 mice. These data collectively demonstrate altered TGFB1 regulated inflammatory, fibrosis, and cell cycle pathways in flexor tendon repair in MRL mice, and could give cues to improved tendon healing.
Collapse
Affiliation(s)
- Jacob G Kallenbach
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Margaret A T Freeberg
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - David Abplanalp
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Rahul G Alenchery
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Raquel E Ajalik
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Samantha Muscat
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacquelyn A Myers
- UR Genomics Research Center (GRC), University of Rochester Medical Center, Rochester, NY, USA
| | - John M Ashton
- UR Genomics Research Center (GRC), University of Rochester Medical Center, Rochester, NY, USA
| | - Alayna Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA
| | - Mark R Buckley
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Hani A Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Orthopaedics, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY, 14642, USA.
| |
Collapse
|
14
|
Mechanical Cues: Bidirectional Reciprocity in the Extracellular Matrix Drives Mechano-Signalling in Articular Cartilage. Int J Mol Sci 2021; 22:ijms222413595. [PMID: 34948394 PMCID: PMC8707858 DOI: 10.3390/ijms222413595] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 12/29/2022] Open
Abstract
The composition and organisation of the extracellular matrix (ECM), particularly the pericellular matrix (PCM), in articular cartilage is critical to its biomechanical functionality; the presence of proteoglycans such as aggrecan, entrapped within a type II collagen fibrillar network, confers mechanical resilience underweight-bearing. Furthermore, components of the PCM including type VI collagen, perlecan, small leucine-rich proteoglycans—decorin and biglycan—and fibronectin facilitate the transduction of both biomechanical and biochemical signals to the residing chondrocytes, thereby regulating the process of mechanotransduction in cartilage. In this review, we summarise the literature reporting on the bidirectional reciprocity of the ECM in chondrocyte mechano-signalling and articular cartilage homeostasis. Specifically, we discuss studies that have characterised the response of articular cartilage to mechanical perturbations in the local tissue environment and how the magnitude or type of loading applied elicits cellular behaviours to effect change. In vivo, including transgenic approaches, and in vitro studies have illustrated how physiological loading maintains a homeostatic balance of anabolic and catabolic activities, involving the direct engagement of many PCM molecules in orchestrating this slow but consistent turnover of the cartilage matrix. Furthermore, we document studies characterising how abnormal, non-physiological loading including excessive loading or joint trauma negatively impacts matrix molecule biosynthesis and/or organisation, affecting PCM mechanical properties and reducing the tissue’s ability to withstand load. We present compelling evidence showing that reciprocal engagement of the cells with this altered ECM environment can thus impact tissue homeostasis and, if sustained, can result in cartilage degradation and onset of osteoarthritis pathology. Enhanced dysregulation of PCM/ECM turnover is partially driven by mechanically mediated proteolytic degradation of cartilage ECM components. This generates bioactive breakdown fragments such as fibronectin, biglycan and lumican fragments, which can subsequently activate or inhibit additional signalling pathways including those involved in inflammation. Finally, we discuss how bidirectionality within the ECM is critically important in enabling the chondrocytes to synthesise and release PCM/ECM molecules, growth factors, pro-inflammatory cytokines and proteolytic enzymes, under a specified load, to influence PCM/ECM composition and mechanical properties in cartilage health and disease.
Collapse
|
15
|
Rai MF. Back to basics: Transcriptomics studies for deep phenotyping of osteoarthritis. OSTEOARTHRITIS AND CARTILAGE OPEN 2021; 3:100166. [PMID: 36474769 PMCID: PMC9718213 DOI: 10.1016/j.ocarto.2021.100166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/06/2021] [Indexed: 11/21/2022] Open
|
16
|
Singh P, Wang M, Mukherjee P, Lessard SG, Pannellini T, Carballo CB, Rodeo SA, Goldring MB, Otero M. Transcriptomic and epigenomic analyses uncovered Lrrc15 as a contributing factor to cartilage damage in osteoarthritis. Sci Rep 2021; 11:21107. [PMID: 34702854 PMCID: PMC8548547 DOI: 10.1038/s41598-021-00269-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/05/2021] [Indexed: 01/03/2023] Open
Abstract
In osteoarthritis (OA), articular chondrocytes display phenotypic and functional changes associated with epigenomic alterations. These changes contribute to the disease progression, which is characterized by dysregulated reparative processes and abnormal extracellular matrix remodeling leading to cartilage degradation. Recent studies using a murine model of posttraumatic OA highlighted the contribution of changes in DNA hydroxymethylation (5hmC) to OA progression. Here, we integrated transcriptomic and epigenomic analyses in cartilage after induction of OA to show that the structural progression of OA is accompanied by early transcriptomic and pronounced DNA methylation (5mC) changes in chondrocytes. These changes accumulate over time and are associated with recapitulation of developmental processes, including cartilage development, chondrocyte hypertrophy, and ossification. Our integrative analyses also uncovered that Lrrc15 is differentially methylated and expressed in OA cartilage, and that it may contribute to the functional and phenotypic alterations of chondrocytes, likely coordinating stress responses and dysregulated extracellular matrix remodeling.
Collapse
Affiliation(s)
- Purva Singh
- Hospital for Special Surgery, HSS Research Institute, New York, NY, 10021, USA
| | - Mengying Wang
- Hospital for Special Surgery, HSS Research Institute, New York, NY, 10021, USA.,School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | | | - Samantha G Lessard
- Hospital for Special Surgery, HSS Research Institute, New York, NY, 10021, USA
| | - Tania Pannellini
- Hospital for Special Surgery, HSS Research Institute, New York, NY, 10021, USA
| | - Camila B Carballo
- Hospital for Special Surgery, HSS Research Institute, New York, NY, 10021, USA
| | - Scott A Rodeo
- Hospital for Special Surgery, HSS Research Institute, New York, NY, 10021, USA.,Weill Cornell Medicine, New York, NY, 10021, USA
| | - Mary B Goldring
- Hospital for Special Surgery, HSS Research Institute, New York, NY, 10021, USA.,Weill Cornell Medicine, New York, NY, 10021, USA
| | - Miguel Otero
- Hospital for Special Surgery, HSS Research Institute, New York, NY, 10021, USA. .,Weill Cornell Medicine, New York, NY, 10021, USA. .,Hospital for Special Surgery, Orthopedic Soft Tissue Research Program, HSS Research Institute, Room 603, 535 East 70th Street, New York, NY, 10021, USA.
| |
Collapse
|
17
|
Single-Cell RNA-Seq Reveals Transcriptomic Heterogeneity and Post-Traumatic Osteoarthritis-Associated Early Molecular Changes in Mouse Articular Chondrocytes. Cells 2021; 10:cells10061462. [PMID: 34200880 PMCID: PMC8230441 DOI: 10.3390/cells10061462] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/25/2022] Open
Abstract
Articular cartilage is a connective tissue lining the surfaces of synovial joints. When the cartilage severely wears down, it leads to osteoarthritis (OA), a debilitating disease that affects millions of people globally. The articular cartilage is composed of a dense extracellular matrix (ECM) with a sparse distribution of chondrocytes with varying morphology and potentially different functions. Elucidating the molecular and functional profiles of various chondrocyte subtypes and understanding the interplay between these chondrocyte subtypes and other cell types in the joint will greatly expand our understanding of joint biology and OA pathology. Although recent advances in high-throughput OMICS technologies have enabled molecular-level characterization of tissues and organs at an unprecedented resolution, thorough molecular profiling of articular chondrocytes has not yet been undertaken, which may be in part due to the technical difficulties in isolating chondrocytes from dense cartilage ECM. In this study, we profiled articular cartilage from healthy and injured mouse knee joints at a single-cell resolution and identified nine chondrocyte subtypes with distinct molecular profiles and injury-induced early molecular changes in these chondrocytes. We also compared mouse chondrocyte subpopulations to human chondrocytes and evaluated the extent of molecular similarity between mice and humans. This work expands our view of chondrocyte heterogeneity and rapid molecular changes in chondrocyte populations in response to joint trauma and highlights potential mechanisms that trigger cartilage degeneration.
Collapse
|
18
|
Guan M, Pan D, Zhang M, Leng X, Yao B. Deer antler extract potentially facilitates xiphoid cartilage growth and regeneration and prevents inflammatory susceptibility by regulating multiple functional genes. J Orthop Surg Res 2021; 16:208. [PMID: 33752715 PMCID: PMC7983396 DOI: 10.1186/s13018-021-02350-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Deer antler is a zoological exception due to its fantastic characteristics, including amazing growth rate and repeatable regeneration. Deer antler has been used as a key ingredient in traditional Chinese medicine relating to kidney and bone health for centuries. The aim of this study was to dissect the molecular regulation of deer antler extract (DAE) on xiphoid cartilage (XC). METHODS The DAE used in this experiment was same as the one that was prepared as previously described. The specific pathogen-free (SPF) grade Sprague-Dawley (SD) rats were randomly divided into blank group (n =10) and DAE group (n =10) after 1-week adaptive feeding. The DAE used in this experiment was same as the one that was prepared as previously described. The rats in DAE group were fed with DAE for 3 weeks at a dose of 0.2 g/kg per day according to the body surface area normalization method, and the rats in blank group were fed with drinking water. Total RNA was extracted from XC located in the most distal edge of the sternum. Illumina RNA sequencing (RNA-seq) in combination with quantitative real-time polymerase chain reaction (qRT-PCR) validation assay was carried out to dissect the molecular regulation of DAE on XC. RESULTS We demonstrated that DAE significantly increased the expression levels of DEGs involved in cartilage growth and regeneration, but decreased the expression levels of DEGs involved in inflammation, and mildly increased the expression levels of DEGs involved in chondrogenesis and chondrocyte proliferation. CONCLUSIONS Our findings suggest that DAE might serve as a complementary therapeutic regent for cartilage growth and regeneration to treat cartilage degenerative disease, such as osteoarthritis.
Collapse
Affiliation(s)
- Mengqi Guan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117 China
| | - Daian Pan
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117 China
| | - Mei Zhang
- Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, Jilin, 130117 China
| | - Xiangyang Leng
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117 China
| | - Baojin Yao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130117 China
| |
Collapse
|
19
|
Soul J, Barter MJ, Little CB, Young DA. OATargets: a knowledge base of genes associated with osteoarthritis joint damage in animals. Ann Rheum Dis 2021; 80:376-383. [PMID: 33077471 PMCID: PMC7892386 DOI: 10.1136/annrheumdis-2020-218344] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/21/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To collate the genes experimentally modulated in animal models of osteoarthritis (OA) and compare these data with OA transcriptomics data to identify potential therapeutic targets. METHODS PubMed searches were conducted to identify publications describing gene modulations in animal models. Analysed gene expression data were retrieved from the SkeletalVis database of analysed skeletal microarray and RNA-Seq expression data. A network diffusion approach was used to predict new genes associated with OA joint damage. RESULTS A total of 459 genes were identified as having been modulated in animal models of OA, with ageing and post-traumatic (surgical) models the most prominent. Ninety-eight of the 143 genes (69%) genetically modulated more than once had a consistent effect on OA joint damage severity. Several discrepancies between different studies were identified, providing lessons on interpretation of these data. We used the data collected along with OA gene expression data to expand existing annotations and prioritise the most promising therapeutic targets, which we validated using the latest reported associations. We constructed an online database OATargets to allow researchers to explore the collated data and integrate it with existing OA and skeletal gene expression data. CONCLUSIONS We present a comprehensive survey and online resource for understanding gene regulation of animal model OA pathogenesis.
Collapse
Affiliation(s)
- Jamie Soul
- Skeletal Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| | - Matthew J Barter
- Skeletal Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| | - Christopher B Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute, The University of Sydney, St Leonards, New South Wales, Australia
| | - David A Young
- Skeletal Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, Tyne and Wear, UK
| |
Collapse
|
20
|
Mendez ME, Sebastian A, Murugesh DK, Hum NR, McCool JL, Hsia AW, Christiansen BA, Loots GG. LPS-Induced Inflammation Prior to Injury Exacerbates the Development of Post-Traumatic Osteoarthritis in Mice. J Bone Miner Res 2020; 35:2229-2241. [PMID: 32564401 PMCID: PMC7689775 DOI: 10.1002/jbmr.4117] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 06/08/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA) is a debilitating and painful disease characterized by the progressive loss of articular cartilage. Post-traumatic osteoarthritis (PTOA) is an injury-induced type of OA that persists in an asymptomatic phase for years before it becomes diagnosed in ~50% of injured individuals. Although PTOA is not classified as an inflammatory disease, it has been suggested that inflammation could be a major driver of PTOA development. Here we examined whether a state of systemic inflammation induced by lipopolysaccharide (LPS) administration 5-days before injury would modulate PTOA outcomes. RNA-seq analysis at 1-day post-injury followed by micro-computed tomography (μCT) and histology characterization at 6 weeks post-injury revealed that LPS administration causes more severe PTOA phenotypes. These phenotypes included significantly higher loss of cartilage and subchondral bone volume. Gene expression analysis showed that LPS alone induced a large cohort of inflammatory genes previously shown to be elevated in synovial M1 macrophages of rheumatoid arthritis (RA) patients, suggesting that systemic LPS produces synovitis. This synovitis was sufficient to promote PTOA in MRL/MpJ mice, a strain previously shown to be resistant to PTOA. The synovium of LPS-treated injured joints displayed an increase in cellularity, and immunohistological examination confirmed that this increase was in part attributable to an elevation in type 1 macrophages. LPS induced the expression of Tlr7 and Tlr8 in both injured and uninjured joints, genes known to be elevated in RA. We conclude that inflammation before injury is an important risk factor for the development of PTOA and that correlating patient serum endotoxin levels or their state of systemic inflammation with PTOA progression may help develop new, effective treatments to lower the rate of PTOA in injured individuals. © 2020 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Melanie E Mendez
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA.,School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Aimy Sebastian
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Deepa K Murugesh
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Nicholas R Hum
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA.,School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Jillian L McCool
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA.,School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Allison W Hsia
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, USA
| | - Blaine A Christiansen
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, USA
| | - Gabriela G Loots
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA.,School of Natural Sciences, University of California Merced, Merced, CA, USA
| |
Collapse
|
21
|
Identification of TGFβ signatures in six murine models mimicking different osteoarthritis clinical phenotypes. Osteoarthritis Cartilage 2020; 28:1373-1384. [PMID: 32659345 DOI: 10.1016/j.joca.2020.06.008] [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: 03/04/2020] [Revised: 06/12/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE TGFβ is a key player in cartilage homeostasis and OA pathology. However, few data are available on the role of TGFβ signalling in the different OA phenotypes. Here, we analysed the TGFβ pathway by transcriptomic analysis in six mouse models of OA. METHOD We have brought together seven expert laboratories in OA pathophysiology and, used inter-laboratories standard operating procedures and quality controls to increase experimental reproducibility and decrease bias. As none of the available OA models covers the complexity and heterogeneity of the human disease, we used six different murine models of knee OA: from post-traumatic/mechanical models (meniscectomy (MNX), MNX and hypergravity (HG-MNX), MNX and high fat diet (HF-MNX), MNX and seipin knock-out (SP-MNX)) to aging-related OA and inflammatory OA (collagenase-induced OA (CIOA)). Four controls (MNX-sham, young, SP-sham, CIOA-sham) were added. OsteoArthritis Research Society International (OARSI)-based scoring of femoral condyles and ribonucleic acid (RNA) extraction from tibial plateau samples were done by single operators as well as the transcriptomic analysis of the TGFβ family pathway by Custom TaqMan® Array Microfluidic Cards. RESULTS The transcriptomic analysis revealed specific gene signatures in each of the six models; however, no gene was deregulated in all six OA models. Of interest, we found that the combinatorial Gdf5-Cd36-Ltbp4 signature might discriminate distinct subgroups of OA: Cd36 upregulation is a hallmark of MNX-related OA while Gdf5 and Ltbp4 upregulation is related to MNX-induced OA and CIOA. CONCLUSION These findings stress the OA animal model heterogeneity and the need of caution when extrapolating results from one model to another.
Collapse
|
22
|
Hasegawa M, Yoshida T, Sudo A. Tenascin-C in Osteoarthritis and Rheumatoid Arthritis. Front Immunol 2020; 11:577015. [PMID: 33101302 PMCID: PMC7554343 DOI: 10.3389/fimmu.2020.577015] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Tenascin-C (TNC) is a large multimodular glycoprotein of the extracellular matrix that consists of four distinct domains. Emerging evidence suggests that TNC may be involved in the pathogenesis of osteoarthritis (OA) and rheumatoid arthritis (RA). In this review, we summarize the current understanding of the role of TNC in cartilage and in synovial biology, across both OA and RA. TNC is expressed in association with the development of articular cartilage; the expression decreases during maturation of chondrocytes and disappears almost completely in adult articular cartilage. TNC expression is increased in diseased cartilage, synovium, and synovial fluid in OA and RA. In addition, elevated circulating TNC levels have been detected in the blood of RA patients. Thus, TNC could be used as a novel biochemical marker for OA and RA, although it has no specificity as a biochemical marker for these joint disorders. In a post-traumatic OA model of aged joints, TNC deficiency was shown to enhance cartilage degeneration. Treatment with TNC domains results in different, domain-specific effects, which are also dose-dependent. For instance, some TNC fragments including the fibrinogen-like globe domain might function as endogenous inducers of synovitis and cartilage matrix degradation through binding with toll-like receptor-4, while full-length TNC promotes cartilage repair and prevents the development of OA without exacerbating synovitis. The TNC peptide TNIIIA2 also prevents cartilage degeneration without causing synovial inflammation. The clinical significance of TNC effects on cartilage and synovium is unclear and understanding the clinical significance of TNC is not straightforward.
Collapse
Affiliation(s)
- Masahiro Hasegawa
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Toshimichi Yoshida
- Department of Pathology & Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
23
|
Mendez ME, Murugesh DK, Sebastian A, Hum NR, McCloy SA, Kuhn EA, Christiansen BA, Loots GG. Antibiotic Treatment Prior to Injury Improves Post-Traumatic Osteoarthritis Outcomes in Mice. Int J Mol Sci 2020; 21:E6424. [PMID: 32899361 PMCID: PMC7503363 DOI: 10.3390/ijms21176424] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a painful and debilitating disease characterized by the chronic and progressive degradation of articular cartilage. Post-traumatic OA (PTOA) is a secondary form of OA that develops in ~50% of cases of severe articular injury. Inflammation and re-occurring injury have been implicated as contributing to the progression of PTOA after the initial injury. However, there is very little known about external factors prior to injury that could affect the risk of PTOA development. To examine how the gut microbiome affects PTOA development we used a chronic antibiotic treatment regimen starting at weaning for six weeks prior to ACL rupture, in mice. A six-weeks post-injury histological examination showed more robust cartilage staining on the antibiotic (AB)-treated mice than the untreated controls (VEH), suggesting slower disease progression in AB cohorts. Injured joints also showed an increase in the presence of anti-inflammatory M2 macrophages in the AB group. Molecularly, the phenotype correlated with a significantly lower expression of inflammatory genes Tlr5, Ccl8, Cxcl13, and Foxo6 in the injured joints of AB-treated animals. Our results indicate that a reduced state of inflammation at the time of injury and a lower expression of Wnt signaling modulatory protein, Rspo1, caused by AB treatment can slow down or improve PTOA outcomes.
Collapse
Affiliation(s)
- Melanie E. Mendez
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Deepa K. Murugesh
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Aimy Sebastian
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| | - Summer A. McCloy
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Edward A. Kuhn
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | | | - Gabriela G. Loots
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| |
Collapse
|
24
|
Zhang S, Hamid MR, Wang T, Liao J, Wen L, Zhou Y, Wei P, Zou X, Chen G, Chen J, Zhou G. RSK-3 promotes cartilage regeneration via interacting with rpS6 in cartilage stem/progenitor cells. Theranostics 2020; 10:6915-6927. [PMID: 32550912 PMCID: PMC7295041 DOI: 10.7150/thno.44875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Cartilage stem/progenitor cells (CSPC) are a promising cellular source to promote endogenous cartilage regeneration in osteoarthritis (OA). Our previous work indicates that ribosomal s6 kinase 3 (RSK-3) is a target of 4-aminobiphenyl, a chemical enhancing CSPC-mediated cartilage repair in OA. However, the primary function and mechanism of RSK-3 in CSPC-mediated cartilage pathobiology remain undefined. Methods: We systematically assessed the association of RSK-3 with OA in three mouse strains with varying susceptibility to OA (MRL/MpJ>CBA>STR/Ort), and also RSK-3-/- mice. Bioinformatic analysis was used to identify the possible mechanism of RSK-3 affecting CSPC, which was further verified in OA mice and CSPC with varying RSK-3 expression induced by chemicals or gene modification. Results: We demonstrated that the level of RSK-3 in cartilage was positively correlated with cartilage repair capacities in three mouse strains (MRL/MpJ>CBA>STR/Ort). Enhanced RSK-3 expression by 4-aminobiphenyl markedly attenuated cartilage injury in OA mice and inhibition or deficiency of RSK-3 expression, on the other hand, significantly aggravated cartilage damage. Transcriptional profiling of CSPC from mice suggested the potential role of RSK-3 in modulating cell proliferation. It was further shown that the in vivo and in vitro manipulation of the RSK-3 expression indeed affected the CSPC proliferation. Mechanistically, ribosomal protein S6 (rpS6) was activated by RSK-3 to accelerate CSPC growth. Conclusion: RSK-3 is identified as a key regulator to enhance cartilage repair, at least partly by regulating the functionality of the cartilage-resident stem/progenitor cells.
Collapse
|
25
|
Sebastian A, Murugesh DK, Mendez ME, Hum NR, Rios-Arce ND, McCool JL, Christiansen BA, Loots GG. Global Gene Expression Analysis Identifies Age-Related Differences in Knee Joint Transcriptome during the Development of Post-Traumatic Osteoarthritis in Mice. Int J Mol Sci 2020; 21:ijms21010364. [PMID: 31935848 PMCID: PMC6982134 DOI: 10.3390/ijms21010364] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/29/2019] [Accepted: 12/29/2019] [Indexed: 12/15/2022] Open
Abstract
Aging and injury are two major risk factors for osteoarthritis (OA). Yet, very little is known about how aging and injury interact and contribute to OA pathogenesis. In the present study, we examined age- and injury-related molecular changes in mouse knee joints that could contribute to OA. Using RNA-seq, first we profiled the knee joint transcriptome of 10-week-old, 62-week-old, and 95-week-old mice and found that the expression of several inflammatory-response related genes increased as a result of aging, whereas the expression of several genes involved in cartilage metabolism decreased with age. To determine how aging impacts post-traumatic arthritis (PTOA) development, the right knee joints of 10-week-old and 62-week-old mice were injured using a non-invasive tibial compression injury model and injury-induced structural and molecular changes were assessed. At six-week post-injury, 62-week-old mice displayed significantly more cartilage degeneration and osteophyte formation compared with young mice. Although both age groups elicited similar transcriptional responses to injury, 62-week-old mice had higher activation of inflammatory cytokines than 10-week-old mice, whereas cartilage/bone metabolism genes had higher expression in 10-week-old mice, suggesting that the differential expression of these genes might contribute to the differences in PTOA severity observed between these age groups.
Collapse
Affiliation(s)
- Aimy Sebastian
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
| | - Deepa K. Murugesh
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
| | - Melanie E. Mendez
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
- Molecular and Cell Biology, School of Natural Sciences, UC Merced, Merced, CA 95343, USA
| | - Nicholas R. Hum
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
- Molecular and Cell Biology, School of Natural Sciences, UC Merced, Merced, CA 95343, USA
| | - Naiomy D. Rios-Arce
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
| | - Jillian L. McCool
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
- Molecular and Cell Biology, School of Natural Sciences, UC Merced, Merced, CA 95343, USA
| | | | - Gabriela G. Loots
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratories, Livermore, CA 94550, USA; (A.S.); (D.K.M.); (M.E.M.); (N.R.H.); (N.D.R.-A.); (J.L.M.)
- Molecular and Cell Biology, School of Natural Sciences, UC Merced, Merced, CA 95343, USA
- Correspondence: ; Tel.: +1-925-423-0923
| |
Collapse
|
26
|
Osteoarthritis and Cartilage Regeneration: Focus on Pathophysiology and Molecular Mechanisms. Int J Mol Sci 2019; 20:ijms20246156. [PMID: 31817613 PMCID: PMC6940733 DOI: 10.3390/ijms20246156] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/12/2022] Open
|
27
|
Singleton Q, Bapat S, Fulzele S. Post-traumatic osteoarthritis (PTOA) animal model to understand pathophysiology of osteoarthritis. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S81. [PMID: 31576290 DOI: 10.21037/atm.2019.04.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Santul Bapat
- Departments of Orthopaedic Surgery, Augusta University, GA, USA
| | - Sadanand Fulzele
- Departments of Orthopaedic Surgery, Augusta University, GA, USA.,Institute of Regenerative and Reparative Medicine, Augusta University, GA, USA
| |
Collapse
|
28
|
Zhang S, Hu P, Liu T, Li Z, Huang Y, Liao J, Hamid MR, Wen L, Wang T, Mo C, Alini M, Grad S, Wang T, Chen D, Zhou G. Kartogenin hydrolysis product 4-aminobiphenyl distributes to cartilage and mediates cartilage regeneration. Theranostics 2019; 9:7108-7121. [PMID: 31695756 PMCID: PMC6831301 DOI: 10.7150/thno.38182] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 08/10/2019] [Indexed: 12/14/2022] Open
Abstract
Rationale The small molecule Kartogenin (KGN) promotes cartilage regeneration in osteoarthritis (OA) by activating stem cells differentiation, but its pharmacological mode-of-action remains unclear. KGN can be cleaved into 4-aminobiphenyl (4-ABP) and phthalic acid (PA) following enzymolysis of an amide bond. Therefore, this study investigated whether 4-ABP or PA exerted the same action as KGN. Methods KGN, 4-ABP and PA were analyzed in cartilage of mice after oral, intravenous or intra-articular administration of KGN by liquid chromatography-mass spectrometry method. Their effect on proliferation and chondrogenic differentiation of mesenchymal stem cells (MSC) was evaluated in vitro. Furthermore, their effect on cartilage preservation was tested in mice OA model induced by destabilization of medial meniscus. OA severity was quantified using OARSI histological scoring. Transcriptional analysis was used to find the possible targets of the chemicals, which were further validated. Results We demonstrated that while oral or intra-articular KGN delivery effectively ameliorated OA phenotypes in mice, only 4-ABP was detectable in cartilage. 4-ABP could induce chondrogenic differentiation and proliferation of MSC in vitro and promote cartilage repair in OA mouse models mainly by increasing the number of CD44+/CD105+ stem-cell and prevention of matrix loss. These effect of 4-ABP was stronger than that of KGN. Transcriptional profiling of 4-ABP-stimulated MSC suggested that RPS6KA2 and the PI3K-Akt pathway were 4-ABP targets; 4-ABP could activate the PI3K-Akt pathway to promote MSC proliferation and repair OA injury, which was blocked in RPS6KA2-knockdown MSC or RPS6KA2-deficient mice.Conclusion 4-ABP bio-distribution in cartilage promotes proliferation and chondrogenic differentiation of MSC, and repairs osteoarthritic lesions via PI3K-Akt pathway activation.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Peilin Hu
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Tao Liu
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
- Department of Biotherapy and Oncology, Shenzhen Luohu People's Hospital, Shenzhen, 518001, China
| | - Zhen Li
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
- AO Research Institute Davos, Davos, Switzerland
| | - Yongcan Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Departments of Orthopaedics, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 510086, China
| | - Jinqi Liao
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Md Rana Hamid
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Liru Wen
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Ting Wang
- Shenzhen Apls Cell Technologies LTD., Yinxing Scientific Building, Lonhua District, Shenzhen, 510086, China
| | - Cuiping Mo
- Shenzhen Apls Cell Technologies LTD., Yinxing Scientific Building, Lonhua District, Shenzhen, 510086, China
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | | - Tianfu Wang
- Guangdong Key Laboratory for Biomedical Measurements & Ultrasound Imaging, School of Biomedical Engineering, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| | - Di Chen
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Guangqian Zhou
- Department of Medical Cell Biology and Genetics, Guangdong Key Laboratory of Genomic Stability and Disease Prevention, Shenzhen Key Laboratory of Anti-aging and Regenerative Medicine, and Shenzhen Engineering Laboratory of Regenerative Technologies for Orthopaedic Diseases, Health Sciences Center, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW The goal of this paper is to review state-of-the-art transcriptome profiling methods and their recent applications in the field of skeletal biology. RECENT FINDINGS Next-generation sequencing of mRNA (RNA-seq) methods have been established and routinely used in skeletal biology research. RNA-seq has led to the identification of novel genes and transcription factors involved in skeletal development and disease, through its application in small and large animal models, as well as human tissue and cells. With the availability of advanced techniques such as single-cell RNA-seq, novel cell types in skeletal tissues are being identified. As the sequencing technologies are rapidly evolving, the exciting discoveries supported by transcriptomics will continue to emerge and improve our understanding of the biology of the skeleton.
Collapse
Affiliation(s)
- Ugur Ayturk
- Musculoskeletal Integrity Program, Hospital for Special Surgery, 515 East 71st St. Suite 403, New York, NY, 10021, USA.
| |
Collapse
|