1
|
Jurynec MJ, Nosyreva E, Thompson D, Munoz C, Novak KA, Matheson DJ, Kazmers NH, Syeda R. PIEZO1 variants that reduce open channel probability are associated with familial osteoarthritis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.03.24312969. [PMID: 39281748 PMCID: PMC11398433 DOI: 10.1101/2024.09.03.24312969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
The synovial joints senses and responds to a multitude of physical forces to maintain joint homeostasis. Disruption of joint homeostasis results in development of osteoarthritis (OA), a disease characterized by loss of joint space, degeneration of articular cartilage, remodeling of bone and other joint tissues, low-grade inflammation, and pain. How changes in mechanosensing in the joint contribute to OA susceptibility remains elusive. PIEZO1 is a major mechanosensitive cation channel in the joint directly regulated by mechanical stimulus. To test whether altered PIEZO1 channel activity causes increased OA susceptibility, we determined whether variants affecting PIEZO1 are associated with dominant inheritance of age-associated familial OA. We identified four rare coding variants affecting PIEZO1 that are associated with familial hand OA. Single channel analyses demonstrated that all four PIEZO1 mutant channels act in a dominant-negative manner to reduce the open probability of the channel in response to pressure. Furthermore, we show that a GWAS mutation in PIEZO1 associated with reduced joint replacement results in increased channel activity when compared with WT and the mutants. Our data support the hypothesis that reduced PIEZO1 activity confers susceptibility to age-associated OA whereas increased PIEZO1 activity may be associated with reduced OA susceptibility.
Collapse
Affiliation(s)
- Michael J Jurynec
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 84108
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112
| | - Elena Nosyreva
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390
| | - David Thompson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390
| | - Crystal Munoz
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390
| | - Kendra A Novak
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 84108
| | - Derek J Matheson
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 84108
| | - Nikolas H Kazmers
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 84108
| | - Ruhma Syeda
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390
| |
Collapse
|
2
|
Arruda AL, Katsoula G, Chen S, Reimann E, Kreitmaier P, Zeggini E. The Genetics and Functional Genomics of Osteoarthritis. Annu Rev Genomics Hum Genet 2024; 25:239-257. [PMID: 39190913 DOI: 10.1146/annurev-genom-010423-095636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Osteoarthritis is the most prevalent whole-joint degenerative disorder, and is characterized by the degradation of articular cartilage and the underlying bone structures. Almost 600 million people are affected by osteoarthritis worldwide. No curative treatments are available, and management strategies focus mostly on pain relief. Here, we provide a comprehensive overview of the available human genetic and functional genomics studies for osteoarthritis to date and delineate how these studies have helped shed light on disease etiopathology. We highlight genetic discoveries from genome-wide association studies and provide a detailed overview of molecular-level investigations in osteoarthritis tissues, including methylation-, transcriptomics-, and proteomics-level analyses. We review how functional genomics data from different molecular levels have helped to prioritize effector genes that can be used as drug targets or drug-repurposing opportunities. Finally, we discuss future directions with the potential to drive a step change in osteoarthritis research.
Collapse
Affiliation(s)
- Ana Luiza Arruda
- Graduate School of Experimental Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- Munich School for Data Science, Helmholtz Munich, Neuherberg, Germany
| | - Georgia Katsoula
- Graduate School of Experimental Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| | - Shibo Chen
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
| | - Ene Reimann
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Peter Kreitmaier
- Graduate School of Experimental Medicine, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Munich, Neuherberg, Germany;
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany
| |
Collapse
|
3
|
Richette P, Latourte A. Hand osteoarthritis: A fresh look. Joint Bone Spine 2024; 91:105652. [PMID: 37797830 DOI: 10.1016/j.jbspin.2023.105652] [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] [Accepted: 11/16/2022] [Indexed: 10/07/2023]
Abstract
Hand osteoarthritis (OA) has been the subject of numerous publications in recent years, particularly in the fields of imaging and therapeutics. The imaging studies revealed a good correlation between the presence of synovitis and/or subchondral edema and arthritic joint pain. Several randomized controlled trials (RCTs) have assessed the efficacy of biologics and conventional DMARDs in patients with symptomatic hand OA. No less than six RCTs have evaluated the symptomatic and, in some cases, structural efficacy of anti-IL-1, anti-TNF or anti-IL-6 drugs. Overall, the results of these trials were disappointing - none of them demonstrated superiority over placebo. There were also two negative trials with hydroxychloroquine. In the end, the only trial that was positive evaluated 10mg oral prednisone versus placebo for 6 weeks in patients with flares of hand OA and synovitis visible on ultrasound. While that trial confirms the role of inflammation in hand OA, it should obviously not encourage the long-term use of corticosteroids as a symptomatic treatment.
Collapse
Affiliation(s)
- Pascal Richette
- Service de rhumatologie, hôpital Lariboisière, Paris, France.
| | | |
Collapse
|
4
|
Dai J, Jiang H, Yang Z, Chen C, Tang X. A functional variant of ALDH1A2 is associated with hand osteoarthritis in the Chinese population. Int J Immunogenet 2024; 51:157-163. [PMID: 38441233 DOI: 10.1111/iji.12662] [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: 11/07/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 05/08/2024]
Abstract
Genome-wide association study identified common variants within the ALDH1A2 gene as the susceptible loci of hand osteoarthritis (HOA) in UK and Iceland populations. Located in chromosome 15, ALDH1A2 encodes aldehyde dehydrogenase family 1 member A2, which is an enzyme that catalyses the synthesis of retinoic acid from retinaldehyde. Our purposes were to replicate the association of functional variant in ALDH1A2 with the development of HOA in the Chinese population. Variant rs12915901 of ALDH1A2 was genotyped in 872 HOA patients and 1223 healthy controls. Subchondral bone samples were collected from 40 patients who had undergone a trapeziectomy, and the tissue expression of ALDH1A2 was analysed. The chi-square analysis was used to compare the frequency of genotype and risk allele between the HOA cases and controls. The Student t test was used to compare the mRNA expression of ALDH1A2 between patients with genotype AA/AG and those with genotype GG. The frequency of genotype AA was significantly higher in HOA patients than in the controls (7.6% vs. 5.1%, p = .01). The frequency of allele A was significantly higher in the patients than in the controls (28.9% vs. 24.6%, p = .005). The mRNA expression of ALDH1A2 was 1.31-folds higher in patients with genotype GG than in the patients with genotype AA/AG (0.000617 ± 0.000231 vs. 0.000471 ± 0.000198, p = .04). Variant rs12915901 of ALDH1A2 contributed to the susceptibility of HOA in the Chinese population. Allele A of rs12915901 can add to the risk of HOA possibly via down-regulation of ALDH1A2 expression.
Collapse
Affiliation(s)
- Jian Dai
- Department of Orthopedics Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China
| | - Haitao Jiang
- Department of Orthopedics Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China
| | - Zhaoqi Yang
- Postgraduate in Orthopedics Surgery, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Chen
- Department of Orthopedics Surgery, Third People's Hospital of Jiujiang City, Jiujiang, Jiangxi, China
| | - Xiaoming Tang
- Department of Orthopedics Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China
| |
Collapse
|
5
|
Duan X, Hu H, Wang L, Chen L. Aldehyde dehydrogenase 1 family: A potential molecule target for diseases. Cell Biol Int 2024. [PMID: 38800962 DOI: 10.1002/cbin.12188] [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: 08/08/2023] [Revised: 04/22/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024]
Abstract
Aldehyde dehydrogenase 1 (ALDH1), a crucial aldehyde metabolizing enzyme, has six family members. The ALDH1 family is expressed in various tissues, with a significant presence in the liver. It plays a momentous role in several pathophysiological processes, including aldehyde detoxification, oxidative stress, and lipid peroxidation. Acetaldehyde detoxification is the fundamental function of the ALDH1 family in participating in vital pathological mechanisms. The ALDH1 family can catalyze retinal to retinoic acid (RA) that is a hormone-signaling molecule and plays a vital role in the development and adult tissues. Furthermore, there is a need for further and broader research on the role of the ALDH1 family as a signaling molecule. The ALDH1 family is widely recognized as a cancer stem cell (CSC) marker and plays a significant role in the proliferation, invasion, metastasis, prognosis, and drug resistance of cancer. The ALDH1 family also participates in other human diseases, such as neurodegenerative diseases, osteoarthritis, diabetes, and atherosclerosis. It can inhibit disease progression by inhibiting/promoting the expression/activity of the ALDH1 family. In this review, we comprehensively analyze the tissue distribution, and functions of the ALDH1 family. Additionally, we review the involvement of the ALDH1 family in diseases, focusing on the underlying pathological mechanisms and briefly talk about the current status and development of ALDH1 family inhibitors. The ALDH1 family presents new possibilities for treating diseases, with both its upstream and downstream pathways serving as promising targets for therapeutic intervention. This offers fresh perspectives for drug development in the field of disease research.
Collapse
Affiliation(s)
- Xiangning Duan
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
| | - Haoliang Hu
- Changde Research Centre for Artificial Intelligence and Biomedicine, Zoology Key Laboratory of Hunan Higher Education, College of Life and Environmental Sciences, Hunan University of Arts and Science, Changde, Hunan, China
| | - Lingzhi Wang
- Department of Pharmacy, The First Affiliated Hospital of Jishou University, Jishou, Hunan, China
| | - Linxi Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, Hunan, China
| |
Collapse
|
6
|
McDonnell E, Orr SE, Barter MJ, Rux D, Brumwell A, Wrobel N, Murphy L, Overmann LM, Sorial AK, Young DA, Soul J, Rice SJ. Epigenetic mechanisms of osteoarthritis risk in human skeletal development. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.05.24306832. [PMID: 38766055 PMCID: PMC11100852 DOI: 10.1101/2024.05.05.24306832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The epigenome, including the methylation of cytosine bases at CG dinucleotides, is intrinsically linked to transcriptional regulation. The tight regulation of gene expression during skeletal development is essential, with ~1/500 individuals born with skeletal abnormalities. Furthermore, increasing evidence is emerging to link age-associated complex genetic musculoskeletal diseases, including osteoarthritis (OA), to developmental factors including joint shape. Multiple studies have shown a functional role for DNA methylation in the genetic mechanisms of OA risk using articular cartilage samples taken from aged patients. Despite this, our knowledge of temporal changes to the methylome during human cartilage development has been limited. We quantified DNA methylation at ~700,000 individual CpGs across the epigenome of developing human articular cartilage in 72 samples ranging from 7-21 post-conception weeks, a time period that includes cavitation of the developing knee joint. We identified significant changes in 8% of all CpGs, and >9400 developmental differentially methylated regions (dDMRs). The largest hypermethylated dDMRs mapped to transcriptional regulators of early skeletal patterning including MEIS1 and IRX1. Conversely, the largest hypomethylated dDMRs mapped to genes encoding extracellular matrix proteins including SPON2 and TNXB and were enriched in chondrocyte enhancers. Significant correlations were identified between the expression of these genes and methylation within the hypomethylated dDMRs. We further identified 811 CpGs at which significant dimorphism was present between the male and female samples, with the majority (68%) being hypermethylated in female samples. Following imputation, we captured the genotype of these samples at >5 million variants and performed epigenome-wide methylation quantitative trait locus (mQTL) analysis. Colocalization analysis identified 26 loci at which genetic variants exhibited shared impacts upon methylation and OA genetic risk. This included loci which have been previously reported to harbour OA-mQTLs (including GDF5 and ALDH1A2), yet the majority (73%) were novel (including those mapping to CHST3, FGF1 and TEAD1). To our knowledge, this is the first extensive study of DNA methylation across human articular cartilage development. We identify considerable methylomic plasticity within the development of knee cartilage and report active epigenomic mediators of OA risk operating in prenatal joint tissues.
Collapse
Affiliation(s)
- Euan McDonnell
- Computational Biology Facility, University of Liverpool, MerseyBio, Crown Street, United Kingdom
| | - Sarah E Orr
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Matthew J Barter
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Danielle Rux
- Orthopaedic Surgery, UConn Health, Farmington, Connecticut, USA
| | - Abby Brumwell
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Nicola Wrobel
- Edinburgh Clinical Research Facility, University of Edinburgh, Edinburgh, United Kingdom
| | - Lee Murphy
- Edinburgh Clinical Research Facility, University of Edinburgh, Edinburgh, United Kingdom
| | - Lynne M Overmann
- Human Developmental Biology Resource, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Antony K Sorial
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - David A Young
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Jamie Soul
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Sarah J Rice
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
7
|
Henkel C, Erikstrup C, Ostrowski SR, Pedersen OB, Troelsen A. Genetics may affect the risk of undergoing surgery for rhizarthrosis. J Orthop Res 2024; 42:1001-1008. [PMID: 38263870 DOI: 10.1002/jor.25753] [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: 08/10/2023] [Revised: 10/25/2023] [Accepted: 11/28/2023] [Indexed: 01/25/2024]
Abstract
Osteoarthritis is a prevalent and severe disease. Involvement of the trapeziometacarpal joint is common and can lead to both pain and disability. Genetics are known to affect the risk of osteoarthritis, but it remains unclear how genetics affect disease trajectories. In this study, we investigated whether the genetic associations of trapeziometacarpal osteoarthritis (rhizarthrosis) vary with the need for surgical treatment. The study was conducted as a case-control genome-wide association study using individuals from the Copenhagen Hospital Biobank pain and degenerative musculoskeletal disease study and the Danish Blood Donor Study (N = 208,342). We identified patients diagnosed with rhizarthrosis and grouped them by treatment status, resulting in two case groups: surgical (N = 1083) and nonsurgical (N = 1888). The case groups were tested against osteoarthritis-free controls in two genome-wide association studies. We then compared variants suggestive of association (p < 10-6) in either of these analyses directly between the treatment groups (surgical vs. nonsurgical rhizarthrosis). We identified 10 variants suggestive of association with either surgical (seven variants) or nonsurgical (three variants) rhizarthrosis. None of the variants reached nominal significance in the opposite treatment group (p ≥ 0.14), and all 10 variants were significantly different between the treatment groups at a false discovery rate of 5%. These results suggest possible differences in the genetic associations of rhizarthrosis depending on surgical treatment. Clinical significance: Uncovering genetic differences between clinically distinct patient groups can reveal biological determinants of disease trajectories.
Collapse
Affiliation(s)
- Cecilie Henkel
- Clinical Orthopaedic Research Hvidovre (CORH), Department of Orthopaedic Surgery, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Sisse R Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Clinical Academic Group: Research OsteoArthritis Denmark (CAG ROAD), Greater Copenhagen Health Science Partners, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ole B Pedersen
- Clinical Academic Group: Research OsteoArthritis Denmark (CAG ROAD), Greater Copenhagen Health Science Partners, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Zealand University Hospital Køge, Køge, Denmark
| | - Anders Troelsen
- Clinical Orthopaedic Research Hvidovre (CORH), Department of Orthopaedic Surgery, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Clinical Academic Group: Research OsteoArthritis Denmark (CAG ROAD), Greater Copenhagen Health Science Partners, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Tyurin A, Akhiiarova K, Minniakhmetov I, Mokrysheva N, Khusainova R. The Genetic Markers of Knee Osteoarthritis in Women from Russia. Biomedicines 2024; 12:782. [PMID: 38672138 PMCID: PMC11048526 DOI: 10.3390/biomedicines12040782] [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: 02/06/2024] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Osteoarthritis is a chronic progressive joint disease that clinically debuts at the stage of pronounced morphologic changes, which makes treatment difficult. In this regard, an important task is the study of genetic markers of the disease, which have not been definitively established, due to the clinical and ethnic heterogeneity of the studied populations. To find the genetic markers for the development of knee osteoarthritis (OA) in women from the Volga-Ural region of Russia, we conducted research in two stages using different genotyping methods, such as the restriction fragment length polymorphism (RFLP) measurement, TaqMan technology and competitive allele-specific PCR-KASPTM. In the first stage, we studied polymorphic variants of candidate genes (ACAN, ADAMTS5, CHST11, SOX9, COL1A1) for OA development. The association of the *27 allele of the VNTR locus of the ACAN gene was identified (OR = 1.6). In the second stage, we replicated the GWAS results (ASTN2, ALDH1A2, DVWA, CHST11, GNL3, NCOA3, FILIP/SENP1, MCF2L, GLT8D, DOT1L) for knee OA studies. The association of the *T allele of the rs7639618 locus of the DVWA gene was detected (OR = 1.54). Thus, the VNTR locus of ACAN and the rs7639618 locus of DVWA are risk factors for knee OA in women from the Volga-Ural region of Russia.
Collapse
Affiliation(s)
- Anton Tyurin
- Internal Medicine Department, Bashkir State Medical University, 450008 Ufa, Russia;
| | - Karina Akhiiarova
- Internal Medicine Department, Bashkir State Medical University, 450008 Ufa, Russia;
| | - Ildar Minniakhmetov
- Endocrinology Research Centre, Dmitriya Ulianova Street, 11, 117036 Moscow, Russia; (I.M.); (N.M.); (R.K.)
| | - Natalia Mokrysheva
- Endocrinology Research Centre, Dmitriya Ulianova Street, 11, 117036 Moscow, Russia; (I.M.); (N.M.); (R.K.)
| | - Rita Khusainova
- Endocrinology Research Centre, Dmitriya Ulianova Street, 11, 117036 Moscow, Russia; (I.M.); (N.M.); (R.K.)
- Medical Genetics Department, Bashkir State Medical University, 450008 Ufa, Russia
| |
Collapse
|
9
|
Bean MB, Favero M, Ramonda R, Scanzello CR. Erosive Hand Osteoarthritis: Recent Advances and Future Treatments. Curr Rheumatol Rep 2024; 26:103-111. [PMID: 38214806 PMCID: PMC10965372 DOI: 10.1007/s11926-023-01130-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2023] [Indexed: 01/13/2024]
Abstract
PURPOSE OF THE REVIEW Erosive hand osteoarthritis (EHOA) is an aggressive form of hand osteoarthritis that leads to significant disability, and recent data suggests that it is increasing in prevalence. This review provides an update of our current understanding of epidemiology, genetic associations, biomarkers, pathogenesis, and treatment of EHOA, with particular focus on studies published within the last 5 years. RECENT FINDINGS New studies of EHOA have identified new genetic loci associated with disease, including variants in genes involved in inflammation and bone remodeling. Preclinical studies implicate pathways of innate immunity, including some that may be causal in the condition. Recent novel studies showed that inflammatory features identified by ultrasound and MRI are associated with development of erosive lesions over time on conventional radiography. In the future, these imaging modalities may be useful in identifying patients at risk of adverse outcomes. Promising new findings in genetics, biomarkers, and treatment targets will hopefully allow for future therapeutic options for this debilitating condition.
Collapse
Affiliation(s)
- Marta B Bean
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| | - Marta Favero
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
- Medicine Unit 1, Ca' Foncello Hospital, Treviso, Italy
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine-DIMED, Padova University Hospital, Padova, Italy
| | - Carla R Scanzello
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Medicine & Translational Musculoskeletal Research Center, Corp. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| |
Collapse
|
10
|
Tashjian RZ, Zitnay J, Kazmers NH, Veerabhadraiah SR, Zelada AC, Honeggar M, Chalmers PN, Henninger HB, Jurynec MJ. Estrogen and testosterone supplementation improves tendon healing and functional recovery after rotator cuff repair. J Orthop Res 2024; 42:259-266. [PMID: 37756152 DOI: 10.1002/jor.25695] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/21/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023]
Abstract
Failure of healing after rotator cuff repair (RCR) is common. The purpose of the current study was to evaluate the effect of systemic estrogen or testosterone supplementation on tendon healing after RCR. Seventy-two adult male mice were utilized for all experiments. The supraspinatus tendon was transected and repaired with 6-0 Prolene suture on the left shoulder of 51 animals. Mice were segregated into three groups postoperative: (1) vehicle group (VG; n = 18), (2) estrogen group (EST; n = 17), and (3) testosterone group (TST; n = 16). An unrepaired control group (unrepaired, n = 21) did not have surgery. Utilizing these animals, histological analysis, activity testing, biomechanical testing and RNA sequencing (RNA-seq) was performed. At 8 weeks post-RCR, TST, and EST supplementation improved the overall histologic structure of the repaired enthesis site. No differences in ultimate failure loads or stiffness were detected between VG, EST, and TST groups after biomechanical testing. RCR caused a reduction in wheel activity compared to unrepaired controls and supplementation with TST restored wheel activity. RNA-seq analysis indicated that estrogen and testosterone regulated different pathways associated with enthesis healing, including a suppression of inflammatory signaling. Supplementation with sex hormones improved the structure of the repaired tendon enthesis and significantly regulated expression of diverse pathways regulating multiple biological processes. Testosterone administration following RCR restored wheel activity without having a detrimental impact on biomechanical strength. Future human studies of sex hormone supplementation after RCR are warranted as supplementation in an animal model may improve tendon enthesis healing.
Collapse
Affiliation(s)
- Robert Z Tashjian
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jared Zitnay
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Nikolas H Kazmers
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | | | - Antonio C Zelada
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Matthew Honeggar
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Peter N Chalmers
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Heath B Henninger
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Michael J Jurynec
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
11
|
Roelofs AJ, De Bari C. Osteoarthritis year in review 2023: Biology. Osteoarthritis Cartilage 2024; 32:148-158. [PMID: 37944663 DOI: 10.1016/j.joca.2023.11.002] [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: 07/11/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
Great progress continues to be made in our understanding of the multiple facets of osteoarthritis (OA) biology. Here, we review the major advances in this field and progress towards therapy development over the past year, highlighting a selection of relevant published literature from a PubMed search covering the year from the end of April 2022 to the end of April 2023. The selected articles have been arranged in themes. These include 1) molecular regulation of articular cartilage and implications for OA, 2) mechanisms of subchondral bone remodelling, 3) role of synovium and inflammation, 4) role of age-related changes including cartilage matrix stiffening, cellular senescence, mitochondrial dysfunction, metabolic dysfunction, and impaired autophagy, and 5) peripheral mechanisms of OA pain. Progress in the understanding of the cellular and molecular mechanisms responsible for the multiple aspects of OA biology is unravelling novel therapeutic targets for disease modification.
Collapse
Affiliation(s)
- Anke J Roelofs
- Arthritis and Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Cosimo De Bari
- Arthritis and Regenerative Medicine Laboratory, Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
12
|
Iqbal L, Zameer U, Iqbal Malick M. Exploring Talarozole as a Novel Therapeutic Approach for Osteoarthritis: Insights From Experimental Studies. CLINICAL MEDICINE INSIGHTS. ARTHRITIS AND MUSCULOSKELETAL DISORDERS 2024; 17:11795441231222494. [PMID: 38292917 PMCID: PMC10823836 DOI: 10.1177/11795441231222494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/07/2023] [Indexed: 02/01/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by the slow degeneration of joint components that primarily affects the elderly. There is currently no cure for OA; thus, treatment focuses on symptom reduction. This article investigates the potential of talarozole, a retinoic acid metabolism-blocking agent (RAMBA), as a new treatment for hand OA. Talarozole showed promising results by inhibiting retinoic acid degradation and increasing its levels in the body. Six hours after destabilization of the medial meniscus, talarozole significantly reduced inflammation in mice's cartilage. The findings underscore the importance of the protein encoded by the ALDH1A2 gene in retinoic acid metabolism, shedding light on its potential implications for the management of OA. Maintaining adequate retinoic acid levels may help to reduce mechano-inflammatory gene regulation. Furthermore, RAMBAs like talarozole may emerge as disease-modifying OA therapies, promising improved symptom control and slower disease progression. In conclusion, this research provides critical genetic insights into severe hand OA and promotes talarozole as a prospective therapy option. These findings pave the door for additional research that could revolutionize OA treatment by targeting retinoic acid metabolism to reduce symptoms and slow disease progression.
Collapse
Affiliation(s)
- Laraib Iqbal
- Department of Medicine, Karachi Medical & Dental College, Karachi, Pakistan
| | - Ushna Zameer
- Department of Medicine, Karachi Medical & Dental College, Karachi, Pakistan
| | | |
Collapse
|
13
|
Wu D, Khan FA, Zhang K, Pandupuspitasari NS, Negara W, Guan K, Sun F, Huang C. Retinoic acid signaling in development and differentiation commitment and its regulatory topology. Chem Biol Interact 2024; 387:110773. [PMID: 37977248 DOI: 10.1016/j.cbi.2023.110773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Retinoic acid (RA), the derivative of vitamin A/retinol, is a signaling molecule with important implications in health and disease. It is a well-known developmental morphogen that functions mainly through the transcriptional activity of nuclear RA receptors (RARs) and, uncommonly, through other nuclear receptors, including peroxisome proliferator-activated receptors. Intracellular RA is under spatiotemporally fine-tuned regulation by synthesis and degradation processes catalyzed by retinaldehyde dehydrogenases and P450 family enzymes, respectively. In addition to dictating the transcription architecture, RA also impinges on cell functioning through non-genomic mechanisms independent of RAR transcriptional activity. Although RA-based differentiation therapy has achieved impressive success in the treatment of hematologic malignancies, RA also has pro-tumor activity. Here, we highlight the relevance of RA signaling in cell-fate determination, neurogenesis, visual function, inflammatory responses and gametogenesis commitment. Genetic and post-translational modifications of RAR are also discussed. A better understanding of RA signaling will foster the development of precision medicine to improve the defects caused by deregulated RA signaling.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | | | - Windu Negara
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
14
|
Zhu L, Vincent TL. Genome-Wide Association Studies to Drug: Identifying Retinoic Acid Metabolism Blocking Agents to Suppress Mechanoflammation in Osteoarthritis. DNA Cell Biol 2023; 42:527-531. [PMID: 37418291 DOI: 10.1089/dna.2023.0197] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023] Open
Abstract
Osteoarthritis (OA) is a highly prevalent debilitating joint disease for which there are currently no licensed disease-modifying treatments. The pathogenesis of OA is complex, involving genetic, mechanical, biochemical, and environmental factors. Cartilage injury, arguably the most important driving factor in OA development, is able to activate both protective and inflammatory pathways within the tissue. Recently, >100 genetic risk variants for OA have been identified through Genome Wide Association Studies, which provide a powerful tool to validate existing putative disease pathways and discover new ones. Using such an approach, hypomorphic variants within the aldehyde dehydrogenase 1 family member A2 (ALDH1A2) gene were shown to be associated with increased risk of severe hand OA. ALDH1A2 encodes the enzyme that synthesizes all-trans retinoic acid (atRA), an intracellular signaling molecule. This review summarizes the influence of the genetic variants on expression and function of ALDH1A2 in OA cartilage, its role in the mechanical injury response of cartilage, and its potent anti-inflammatory effect after cartilage injury. In doing so it identifies atRA metabolism-blocking agents as potential treatments for suppressing mechanoflammation in OA.
Collapse
Affiliation(s)
- Linyi Zhu
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| | - Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
15
|
Kaya S, Bailey KN, Schurman CA, Evans DS, Alliston T. Bone-cartilage crosstalk informed by aging mouse bone transcriptomics and human osteoarthritis genome-wide association studies. Bone Rep 2023; 18:101647. [PMID: 36636109 PMCID: PMC9830153 DOI: 10.1016/j.bonr.2022.101647] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/28/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022] Open
Abstract
Subchondral bone participates in crosstalk with articular cartilage to maintain joint homeostasis, and disruption of either tissue results in overall joint degeneration. Among the subchondral bone changes observed in osteoarthritis (OA), subchondral bone plate (SBP) thickening has a time-dependent relationship with cartilage degeneration and has recently been shown to be regulated by osteocytes. Here, we evaluate the effect of age on SBP thickness and cartilage degeneration in aging mice. We find that SBP thickness significantly increases by 18-months of age, corresponding temporally with increased cartilage degeneration. To identify factors in subchondral bone that may participate in bone cartilage crosstalk or OA, we leveraged mouse transcriptomic data from one joint tissue compartment - osteocyte-enriched bone - to search for enrichment with human OA in UK Biobank and Arthritis Research UK Osteoarthritis Genetics (arcOGEN) GWAS using the mouse2human (M2H, www.mouse2human.org) strategy. Genes differentially expressed in aging mouse bone are significantly enriched for human OA, showing joint site-specific (knee vs. hip) relationships, exhibit temporal associations with age, and unique gene clusters are implicated in each type of OA. Application of M2H identifies genes with known and unknown functions in osteocytes and OA development that are clinically associated with human OA. Altogether, this work prioritizes genes with a potential role in bone/cartilage crosstalk for further mechanistic study based on their association with human OA in GWAS.
Collapse
Affiliation(s)
- Serra Kaya
- Department of Orthopaedic Surgery, University of California San Francisco, CA, United States of America
| | - Karsyn N. Bailey
- Department of Orthopaedic Surgery, University of California San Francisco, CA, United States of America
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, United States of America
| | - Charles A. Schurman
- Department of Orthopaedic Surgery, University of California San Francisco, CA, United States of America
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, United States of America
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, United States of America
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, CA, United States of America
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, United States of America
| |
Collapse
|
16
|
RNA-Seq Reveals the mRNAs, miRNAs, and lncRNAs Expression Profile of Knee Joint Synovial Tissue in Osteoarthritis Patients. J Clin Med 2023; 12:jcm12041449. [PMID: 36835984 PMCID: PMC9968173 DOI: 10.3390/jcm12041449] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/19/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
Osteoarthritis (OA) is a chronic disease common in the elderly population and imposes significant health and economic burden. Total joint replacement is the only currently available treatment but does not prevent cartilage degeneration. The molecular mechanism of OA, especially the role of inflammation in disease progression, is incompletely understood. We collected knee joint synovial tissue samples of eight OA patients and two patients with popliteal cysts (controls), measured the expression levels of lncRNAs, miRNAs, and mRNAs in these tissues by RNA-seq, and identified differentially expressed genes (DEGs) and key pathways. In the OA group, 343 mRNAs, 270 lncRNAs, and 247 miRNAs were significantly upregulated, and 232 mRNAs, 109 lncRNAs, and 157 miRNAs were significantly downregulated. mRNAs potentially targeted by lncRNAs were predicted. Nineteen overlapped miRNAs were screened based on our sample data and GSE 143514 data. Pathway enrichment and functional annotation analyses showed that the inflammation-related transcripts CHST11, ALDH1A2, TREM1, IL-1β, IL-8, CCL5, LIF, miR-146a-5p, miR-335-5p, lncRNA GAS5, LINC02288, and LOC101928134 were differentially expressed. In this study, inflammation-related DEGs and non-coding RNAs were identified in synovial samples, suggesting that competing endogenous RNAs have a role in OA. TREM1, LIF, miR146-5a, and GAS5 were identified to be OA-related genes and potential regulatory pathways. This research helps elucidate the pathogenesis of OA and identify novel therapeutic targets for this disorder.
Collapse
|
17
|
Khosasih V, Liu KM, Huang CM, Liou LB, Hsieh MS, Lee CH, Tsai CY, Kuo SY, Hwa SY, Yu CL, Chang CH, Lin CJ, Hsieh SC, Cheng CY, Chen WM, Chen LK, Chuang HP, Chen YT, Tsai PC, Lu LS, H’ng WS, Zhang Y, Chen HC, Chen CH, Lee MTM, Wu JY. A Functional Polymorphism Downstream of Vitamin A Regulator Gene CYP26B1 Is Associated with Hand Osteoarthritis. Int J Mol Sci 2023; 24:ijms24033021. [PMID: 36769350 PMCID: PMC9918232 DOI: 10.3390/ijms24033021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
While genetic analyses have revealed ~100 risk loci associated with osteoarthritis (OA), only eight have been linked to hand OA. Besides, these studies were performed in predominantly European and Caucasian ancestries. Here, we conducted a genome-wide association study in the Han Chinese population to identify genetic variations associated with the disease. We recruited a total of 1136 individuals (n = 420 hand OA-affected; n = 716 unaffected control subjects) of Han Chinese ancestry. We carried out genotyping using Axiom Asia Precisi on Medicine Research Array, and we employed the RegulomeDB database and RoadMap DNase I Hypersensitivity Sites annotations to further narrow down our potential candidate variants. Genetic variants identified were tested in the Geisinger's hand OA cohort selected from the Geisinger MyCode community health initiative (MyCode®). We also performed a luciferase reporter assay to confirm the potential impact of top candidate single-nucleotide polymorphisms (SNPs) on hand OA. We identified six associated SNPs (p-value = 6.76 × 10-7-7.31 × 10-6) clustered at 2p13.2 downstream of the CYP26B1 gene. The strongest association signal identified was rs883313 (p-value = 6.76 × 10-7, odds ratio (OR) = 1.76), followed by rs12713768 (p-value = 1.36 × 10-6, OR = 1.74), near or within the enhancer region closest to the CYP26B1 gene. Our findings showed that the major risk-conferring CC haplotype of SNPs rs12713768 and rs10208040 [strong linkage disequilibrium (LD); D' = 1, r2 = 0.651] drives 18.9% of enhancer expression activity. Our findings highlight that the SNP rs12713768 is associated with susceptibility to and severity of hand OA in the Han Chinese population and that the suggested retinoic acid signaling pathway may play an important role in its pathogenesis.
Collapse
Affiliation(s)
- Vivia Khosasih
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Kai-Ming Liu
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Chung-Ming Huang
- Division of Immunology and Rheumatology, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Correspondence: (C.-M.H.); (J.-Y.W.)
| | - Lieh-Bang Liou
- Division of Rheumatology, Allergy and Immunology, New Taipei Municipal Tucheng Hospital, New Taipei City 236, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Ming-Shium Hsieh
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Orthopedics, Taipei Medical University Hospital, Taipei 110, Taiwan
- Department of Orthopedics, En Chu Kong Hospital, New Taipei 237, Taiwan
| | - Chian-Her Lee
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Orthopedics, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Chang-Youh Tsai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - San-Yuan Kuo
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Su-Yang Hwa
- Department of Orthopaedics, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chih-Hao Chang
- Department of Orthopedics, College of Medicine, National Taiwan University and National Taiwan University Hospital, Taipei 100, Taiwan
- Department of Orthopedics, National Taiwan University Hospital Jin-Shan Branch, New Taipei City 208, Taiwan
| | - Cheng-Jyh Lin
- Department of Orthopedics, China Medical University Hospital, Taichung 404, Taiwan
| | - Song-Chou Hsieh
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chun-Ying Cheng
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Orthopedic, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Wei-Ming Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Orthopaedics and Traumatology, Taipei Veteran General Hospital, Taipei 112, Taiwan
| | - Liang-Kuang Chen
- Department of Diagnostic Radiology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
| | - Hui-Ping Chuang
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Ying-Ting Chen
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Pei-Chun Tsai
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Liang-Suei Lu
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Weng-Siong H’ng
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger, Danville, PA 17822, USA
| | - Hsiang-Cheng Chen
- Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chien-Hsiun Chen
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| | - Ming Ta Michael Lee
- Genomic Medicine Institute, Geisinger, Danville, PA 17822, USA
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| | - Jer-Yuarn Wu
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
- National Center for Genome Medicine, Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- School of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Correspondence: (C.-M.H.); (J.-Y.W.)
| |
Collapse
|
18
|
Zhu L, Kamalathevan P, Koneva LA, Zarebska JM, Chanalaris A, Ismail H, Wiberg A, Ng M, Muhammad H, Walsby-Tickle J, McCullagh JSO, Watt FE, Sansom SN, Furniss D, Gardiner MD, Vincent TL, Riley N, Spiteri M, McNab I, Little C, Cogswell L, Critchley P, Giele H, Shirley R. Variants in ALDH1A2 reveal an anti-inflammatory role for retinoic acid and a new class of disease-modifying drugs in osteoarthritis. Sci Transl Med 2022; 14:eabm4054. [PMID: 36542696 DOI: 10.1126/scitranslmed.abm4054] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
More than 40% of individuals will develop osteoarthritis (OA) during their lifetime, yet there are currently no licensed disease-modifying treatments for this disabling condition. Common polymorphic variants in ALDH1A2, which encodes the key enzyme for synthesis of all-trans retinoic acid (atRA), are associated with severe hand OA. Here, we sought to elucidate the biological significance of this association. We first confirmed that ALDH1A2 risk variants were associated with hand OA in the U.K. Biobank. Articular cartilage was acquired from 33 individuals with hand OA at the time of routine hand OA surgery. After stratification by genotype, RNA sequencing was performed. A reciprocal relationship between ALDH1A2 mRNA and inflammatory genes was observed. Articular cartilage injury up-regulated similar inflammatory genes by a process that we have previously termed mechanoflammation, which we believe is a primary driver of OA. Cartilage injury was also associated with a concomitant drop in atRA-inducible genes, which were used as a surrogate measure of cellular atRA concentration. Both responses to injury were reversed using talarozole, a retinoic acid metabolism blocking agent (RAMBA). Suppression of mechanoflammation by talarozole was mediated by a peroxisome proliferator-activated receptor gamma (PPARγ)-dependent mechanism. Talarozole was able to suppress mechano-inflammatory genes in articular cartilage in vivo 6 hours after mouse knee joint destabilization and reduced cartilage degradation and osteophyte formation after 26 days. These data show that boosting atRA suppresses mechanoflammation in the articular cartilage in vitro and in vivo and identifies RAMBAs as potential disease-modifying drugs for OA.
Collapse
Affiliation(s)
- Linyi Zhu
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Pragash Kamalathevan
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Lada A Koneva
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Jadwiga Miotla Zarebska
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Anastasios Chanalaris
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Heba Ismail
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
- Healthy Lifespan Institute (HELSI) and Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2TN, UK
| | - Akira Wiberg
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Michael Ng
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Hayat Muhammad
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - John Walsby-Tickle
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - James S O McCullagh
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Fiona E Watt
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
- Centre for Inflammatory Disease, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| | - Stephen N Sansom
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Dominic Furniss
- Botnar Research Centre, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Matthew D Gardiner
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Tonia L Vincent
- Centre for Osteoarthritis Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
McDonald MLN, Lakshman Kumar P, Srinivasasainagendra V, Nair A, Rocco AP, Wilson AC, Chiles JW, Richman JS, Pinson SA, Dennis RA, Jagadale V, Brown CJ, Pyarajan S, Tiwari HK, Bamman MM, Singh JA. Novel genetic loci associated with osteoarthritis in multi-ancestry analyses in the Million Veteran Program and UK Biobank. Nat Genet 2022; 54:1816-1826. [PMID: 36411363 DOI: 10.1038/s41588-022-01221-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 10/05/2022] [Indexed: 11/22/2022]
Abstract
Osteoarthritis is a common progressive joint disease. As no effective medical interventions are available, osteoarthritis often progresses to the end stage, in which only surgical options such as total joint replacement are available. A more thorough understanding of genetic influences of osteoarthritis is essential to develop targeted personalized approaches to treatment, ideally long before the end stage is reached. To date, there have been no large multiancestry genetic studies of osteoarthritis. Here, we leveraged the unique resources of 484,374 participants in the Million Veteran Program and UK Biobank to address this gap. Analyses included participants of European, African, Asian and Hispanic descent. We discovered osteoarthritis-associated genetic variation at 10 loci and replicated findings from previous osteoarthritis studies. We also present evidence that some osteoarthritis-associated regions are robust to population ancestry. Drug repurposing analyses revealed enrichment of targets of several medication classes and provide potential insight into the etiology of beneficial effects of antiepileptics on osteoarthritis pain.
Collapse
Affiliation(s)
- Merry-Lynn N McDonald
- Birmingham Veterans Affairs Health Care System (BVAHCS), Birmingham, AL, USA.
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Preeti Lakshman Kumar
- Birmingham Veterans Affairs Health Care System (BVAHCS), Birmingham, AL, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Vinodh Srinivasasainagendra
- Birmingham Veterans Affairs Health Care System (BVAHCS), Birmingham, AL, USA
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ashwathy Nair
- Birmingham Veterans Affairs Health Care System (BVAHCS), Birmingham, AL, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Alison P Rocco
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Ava C Wilson
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joe W Chiles
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Joshua S Richman
- Birmingham Veterans Affairs Health Care System (BVAHCS), Birmingham, AL, USA
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sarah A Pinson
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Richard A Dennis
- Central Arkansas Veterans Healthcare System (CAVHS), Little Rock, AR, USA
| | - Vivek Jagadale
- Central Arkansas Veterans Healthcare System (CAVHS), Little Rock, AR, USA
| | - Cynthia J Brown
- Birmingham Veterans Affairs Health Care System (BVAHCS), Birmingham, AL, USA
- Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences (C-DACS), Veterans Affairs Boston Healthcare System (VABHS), Boston, MA, USA
| | - Hemant K Tiwari
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marcas M Bamman
- Birmingham Veterans Affairs Health Care System (BVAHCS), Birmingham, AL, USA
- Department of Cell, Developmental, and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Florida Institute for Human & Machine Cognition, Pensacola, FL, USA
| | - Jasvinder A Singh
- Birmingham Veterans Affairs Health Care System (BVAHCS), Birmingham, AL, USA
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
- Division of Rheumatology and Clinical Immunology, Department of Medicine at the School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
20
|
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: 0.5] [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
|
21
|
Yu Q, Huang Y, Chen X, Chen Y, Zhu X, Liu Y, Liu J. A neutrophil cell membrane-biomimetic nanoplatform based on L-arginine nanoparticles for early osteoarthritis diagnosis and nitric oxide therapy. NANOSCALE 2022; 14:11619-11634. [PMID: 35894521 DOI: 10.1039/d2nr02601e] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Osteoarthritis (OA) is a common debilitating disease affecting articular joints for which no effective disease-modifying early diagnosis or medical therapy tools are currently available. The inefficient delivery of drugs into inflamed chondrocytes has restricted the development of anti-OA medication. Evidence has shown that inflammatory neutrophils possess the property of targeting inflammation via inflammatory tissue recruiting. Herein, we report neutrophil-cell-membrane-based biomimetic nanoparticles (NM-LANPs@Ru) as an OA theranostic nanoplatform; they act as a NO delivery system, coating neutrophil cell membrane onto the surface of self-assembled PEGylated L-arginine nanoparticles (LANPs) to act as a NO donor and loading a Ru complex to act as a ROS inducer. NM-LANPs@Ru demonstrated the specific targeting of inflamed OA with low toxicity, good NO release, and excellent fluorescence/photoacoustic (FL/PA) imaging properties. We showed that NM-LANPs@Ru exhibited enhanced cellular association in inflamed chondrocyte cells (C28/I2), much higher than NO release from ROS oxidized LA, and it improved the inhibition of the apoptosis of inflamed C28/I2 cells compared with control treatments. In vivo studies demonstrated that NM-LANPs@Ru effectively targeted inflamed OA, based on real-time dual-modal FL/PA imaging, eventually exhibiting its excellent anti-inflammatory activity. Our study may provide a new approach for the early diagnosis and treatment of osteoarthritis using a neutrophil-cell-membrane-based biomimetic nanoplatform for NO or drug delivery.
Collapse
Affiliation(s)
- Qianqian Yu
- South China Advanced Institute for Soft Matter Science and Technology, School of Molecular Science and Engineering, South China University of Technology, Guangzhou, China
| | - Yuqin Huang
- College of Chemistry and Materials Science, Jinan University, Guangzhou, China.
| | - Xu Chen
- College of Chemistry and Materials Science, Jinan University, Guangzhou, China.
| | - Yutong Chen
- College of Chemistry and Materials Science, Jinan University, Guangzhou, China.
| | - Xufeng Zhu
- College of Chemistry and Materials Science, Jinan University, Guangzhou, China.
| | - Yanan Liu
- College of Chemistry and Materials Science, Jinan University, Guangzhou, China.
| | - Jie Liu
- College of Chemistry and Materials Science, Jinan University, Guangzhou, China.
| |
Collapse
|
22
|
Identification of candidate enhancers controlling the transcriptome during the formation of interphalangeal joints. Sci Rep 2022; 12:12835. [PMID: 35896673 PMCID: PMC9329285 DOI: 10.1038/s41598-022-16951-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/19/2022] [Indexed: 11/09/2022] Open
Abstract
The formation of the synovial joint begins with the visible emergence of a stripe of densely packed mesenchymal cells located between distal ends of the developing skeletal anlagen called the interzone. Recently the transcriptome of the early synovial joint was reported. Knowledge about enhancers would complement these data and lead to a better understanding of the control of gene transcription at the onset of joint development. Using ChIP-sequencing we have mapped the H3-signatures H3K27ac and H3K4me1 to locate regulatory elements specific for the interzone and adjacent phalange, respectively. This one-stage atlas of candidate enhancers (CEs) was used to map the association between these respective joint tissue specific CEs and biological processes. Subsequently, integrative analysis of transcriptomic data and CEs identified new putative regulatory elements of genes expressed in interzone (e.g., GDF5, BMP2 and DACT2) and phalange (e.g., MATN1, HAPLN1 and SNAI1). We also linked such CEs to genes known as crucial in synovial joint hypermobility and osteoarthritis, as well as phalange malformations. These analyses show that the CE atlas can serve as resource for identifying, and as starting point for experimentally validating, putative disease-causing genomic regulatory regions in patients with synovial joint dysfunctions and/or phalange disorders, and enhancer-controlled synovial joint and phalange formation.
Collapse
|
23
|
Aubourg G, Rice SJ, Bruce-Wootton P, Loughlin J. Genetics of osteoarthritis. Osteoarthritis Cartilage 2022; 30:636-649. [PMID: 33722698 PMCID: PMC9067452 DOI: 10.1016/j.joca.2021.03.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/17/2021] [Accepted: 03/06/2021] [Indexed: 02/02/2023]
Abstract
Osteoarthritis genetics has been transformed in the past decade through the application of large-scale genome-wide association scans. So far, over 100 polymorphic DNA variants have been associated with this common and complex disease. These genetic risk variants account for over 20% of osteoarthritis heritability and the vast majority map to non-protein coding regions of the genome where they are presumed to act by regulating the expression of target genes. Statistical fine mapping, in silico analyses of genomics data, and laboratory-based functional studies have enabled the identification of some of these targets, which encode proteins with diverse roles, including extracellular signaling molecules, intracellular enzymes, transcription factors, and cytoskeletal proteins. A large number of the risk variants correlate with epigenetic factors, in particular cartilage DNA methylation changes in cis, implying that epigenetics may be a conduit through which genetic effects on gene expression are mediated. Some of the variants also appear to have been selected as humans adapted to bipedalism, suggesting that a proportion of osteoarthritis genetic susceptibility results from antagonistic pleiotropy, with risk variants having a positive role in joint formation but a negative role in the long-term health of the joint. Although data from an osteoarthritis genetic study has not yet directly led to a novel treatment, some of the osteoarthritis associated genes code for proteins that have available therapeutics. Genetic investigations are therefore revealing fascinating fundamental insights into osteoarthritis and can expose options for translational intervention.
Collapse
Affiliation(s)
- G Aubourg
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - S J Rice
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - P Bruce-Wootton
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - J Loughlin
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
24
|
Liu W, Wen D, Liu Z, Wang K, Wang J. Erythropoiesis signature and ubiquitin‐mediated proteolysis are enriched in systematic juvenile idiopathic arthritis. Int J Immunogenet 2022; 49:193-201. [PMID: 35253998 DOI: 10.1111/iji.12573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/23/2022] [Accepted: 02/10/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Wenping Liu
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| | - Dawei Wen
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| | - Ziyi Liu
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| | - Kunyu Wang
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| | - Jibo Wang
- Department of Rheumatology & Clinical Immunology Affiliated Hospital of Qingdao University Qingdao China
| |
Collapse
|
25
|
Zhang D, Li Q, Zhang D, Yang X, Wang C, Zhang R, Yang X, Li Z, Xiong Y. An eQTL variant of ALDH1A2 is associated with Kashin-Beck disease in Chinese population. J Bone Miner Metab 2022; 40:317-326. [PMID: 35059888 DOI: 10.1007/s00774-021-01287-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 10/28/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The aims of the study were to investigate the relationship between aldehyde dehydrogenase 1 family member A2 (ALDH1A2) and Kashin-Beck disease (KBD), explore the effects of the rs3204689 polymorphism and methylation status on the expression levels of ALDH1A2, and further clarify the pathogenesis of KBD. MATERIALS AND METHODS The genotype of ALDH1A2 rs3204689 was detected by PCR-RFLP in 103 KBD patients and 109 healthy controls in the whole blood. The mRNA level of ALDH1A2 was measured by qRT-PCR, and the protein expression was detected using IHC staining and Western blotting. The MSP-PCR was used to identify the ALDH1A2 methylation level. RESULTS There were significant differences in G/G, G/C, and C/C frequencies of ALDH1A2 rs3204689 between the KBD and control groups (χ2 = 7.113, P = 0.029); the minor allele G of ALDH1A2 was associated with the risk of KBD (χ2 = 5.984, P = 0.014). The mRNA and protein levels of ALDH1A2 were increased in the whole blood and cartilage of KBD patients compared with the controls (P = 0.049, P < 0.0001, P = 0.019). Meanwhile, a statistically significant difference was observed between G/G, G/C and C/C genotype on mRNA expression (P = 0.039). The methylation level of the ALDH1A2 gene promoter region showed no significant difference between the KBD and control groups (χ2 = 0.317, P = 0.573). CONCLUSION Our case-control study indicates that the common variant rs3204689 near ALDH1A2 is associated with KBD in Chinese population. The risk allele G of rs3204689 is statistically linked to the high expression of ALDH1A2, which is up-regulated in the cartilage and whole blood of KBD patients. Our findings suggest a potential role of ALDH1A2 in the pathogenesis of KBD.
Collapse
Affiliation(s)
- Di Zhang
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, School of Public Health, National Health Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Qiang Li
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, School of Public Health, National Health Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Dandan Zhang
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, School of Public Health, National Health Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Xiaoli Yang
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, School of Public Health, National Health Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Chen Wang
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, School of Public Health, National Health Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Rongqiang Zhang
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, School of Public Health, National Health Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
- Shaanxi University of Chinese Medicine, Xianyang, People's Republic of China
| | - Xuena Yang
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, School of Public Health, National Health Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Zhaofang Li
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, School of Public Health, National Health Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yongmin Xiong
- Institute of Endemic Diseases and Key Laboratory of Trace Elements and Endemic Diseases, School of Public Health, National Health Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|
26
|
Mikaelsdottir E, Thorleifsson G, Stefansdottir L, Halldorsson G, Sigurdsson JK, Lund SH, Tragante V, Melsted P, Rognvaldsson S, Norland K, Helgadottir A, Magnusson MK, Ragnarsson GB, Kristinsson SY, Reykdal S, Vidarsson B, Gudmundsdottir IJ, Olafsson I, Onundarson PT, Sigurdardottir O, Sigurdsson EL, Grondal G, Geirsson AJ, Geirsson G, Gudmundsson J, Holm H, Saevarsdottir S, Jonsdottir I, Thorgeirsson G, Gudbjartsson DF, Thorsteinsdottir U, Rafnar T, Stefansson K. Genetic variants associated with platelet count are predictive of human disease and physiological markers. Commun Biol 2021; 4:1132. [PMID: 34580418 PMCID: PMC8476563 DOI: 10.1038/s42003-021-02642-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 09/07/2021] [Indexed: 12/13/2022] Open
Abstract
Platelets play an important role in hemostasis and other aspects of vascular biology. We conducted a meta-analysis of platelet count GWAS using data on 536,974 Europeans and identified 577 independent associations. To search for mechanisms through which these variants affect platelets, we applied cis-expression quantitative trait locus, DEPICT and IPA analyses and assessed genetic sharing between platelet count and various traits using polygenic risk scoring. We found genetic sharing between platelet count and counts of other blood cells (except red blood cells), in addition to several other quantitative traits, including markers of cardiovascular, liver and kidney functions, height, and weight. Platelet count polygenic risk score was predictive of myeloproliferative neoplasms, rheumatoid arthritis, ankylosing spondylitis, hypertension, and benign prostate hyperplasia. Taken together, these results advance understanding of diverse aspects of platelet biology and how they affect biological processes in health and disease. Evgenia Mikaelsdottir et al. report a study of variants associated with platelet count among European individuals where they identify 577 associations. They also report a genetic overlap between platelet count and human diseases, including myeloproliferative neoplasms, rheumatoid arthritis, and hypertension, as well as a genetic overlap between platelet count and various physiological markers.
Collapse
Affiliation(s)
| | | | | | | | | | - Sigrun H Lund
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland
| | | | - Pall Melsted
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | - Magnus K Magnusson
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | - Gunnar B Ragnarsson
- Department of Oncology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | - Sigurdur Y Kristinsson
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.,Department of Hematology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | - Sigrun Reykdal
- Department of Hematology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | - Brynjar Vidarsson
- Department of Hematology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | | | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | - Pall T Onundarson
- Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.,Laboratory Hematology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | - Olof Sigurdardottir
- Department of Clinical Biochemistry, Akureyri Hospital, 600, Akureyri, Iceland
| | | | - Gerdur Grondal
- Department of Rheumatology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | - Arni J Geirsson
- Department of Rheumatology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | - Gudmundur Geirsson
- Department of Urology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | | | - Hilma Holm
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland
| | - Saedis Saevarsdottir
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.,Department of Rheumatology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | - Ingileif Jonsdottir
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | - Gudmundur Thorgeirsson
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland.,Department of Cardiology, Landspitali-University Hospital, 101, Reykjavik, Iceland
| | - Daniel F Gudbjartsson
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland.,School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland
| | - Thorunn Rafnar
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE Genetics/Amgen, Sturlugata 8, 101, Reykjavik, Iceland. .,Faculty of Medicine, University of Iceland, 101, Reykjavik, Iceland.
| |
Collapse
|
27
|
Boer CG, Hatzikotoulas K, Southam L, Stefánsdóttir L, Zhang Y, Coutinho de Almeida R, Wu TT, Zheng J, Hartley A, Teder-Laving M, Skogholt AH, Terao C, Zengini E, Alexiadis G, Barysenka A, Bjornsdottir G, Gabrielsen ME, Gilly A, Ingvarsson T, Johnsen MB, Jonsson H, Kloppenburg M, Luetge A, Lund SH, Mägi R, Mangino M, Nelissen RRGHH, Shivakumar M, Steinberg J, Takuwa H, Thomas LF, Tuerlings M, Babis GC, Cheung JPY, Kang JH, Kraft P, Lietman SA, Samartzis D, Slagboom PE, Stefansson K, Thorsteinsdottir U, Tobias JH, Uitterlinden AG, Winsvold B, Zwart JA, Davey Smith G, Sham PC, Thorleifsson G, Gaunt TR, Morris AP, Valdes AM, Tsezou A, Cheah KSE, Ikegawa S, Hveem K, Esko T, Wilkinson JM, Meulenbelt I, Lee MTM, van Meurs JBJ, Styrkársdóttir U, Zeggini E. Deciphering osteoarthritis genetics across 826,690 individuals from 9 populations. Cell 2021; 184:4784-4818.e17. [PMID: 34450027 PMCID: PMC8459317 DOI: 10.1016/j.cell.2021.07.038] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/26/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022]
Abstract
Osteoarthritis affects over 300 million people worldwide. Here, we conduct a genome-wide association study meta-analysis across 826,690 individuals (177,517 with osteoarthritis) and identify 100 independently associated risk variants across 11 osteoarthritis phenotypes, 52 of which have not been associated with the disease before. We report thumb and spine osteoarthritis risk variants and identify differences in genetic effects between weight-bearing and non-weight-bearing joints. We identify sex-specific and early age-at-onset osteoarthritis risk loci. We integrate functional genomics data from primary patient tissues (including articular cartilage, subchondral bone, and osteophytic cartilage) and identify high-confidence effector genes. We provide evidence for genetic correlation with phenotypes related to pain, the main disease symptom, and identify likely causal genes linked to neuronal processes. Our results provide insights into key molecular players in disease processes and highlight attractive drug targets to accelerate translation.
Collapse
Affiliation(s)
- Cindy G Boer
- Department of Internal Medicine, Erasmus MC, Medical Center, 3015CN Rotterdam, the Netherlands
| | - Konstantinos Hatzikotoulas
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Lorraine Southam
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger Health System, Danville, PA 17822, USA
| | - Rodrigo Coutinho de Almeida
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - Tian T Wu
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jie Zheng
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - April Hartley
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; Musculoskeletal Research Unit, Translation Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK
| | - Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Anne Heidi Skogholt
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa 230-0045, Japan
| | - Eleni Zengini
- 4(th) Psychiatric Department, Dromokaiteio Psychiatric Hospital, 12461 Athens, Greece
| | - George Alexiadis
- 1(st) Department of Orthopaedics, KAT General Hospital, 14561 Athens, Greece
| | - Andrei Barysenka
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | | | - Maiken E Gabrielsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Thorvaldur Ingvarsson
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland; Department of Orthopedic Surgery, Akureyri Hospital, 600 Akureyri, Iceland
| | - Marianne B Johnsen
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0316 Oslo, Norway; Research and Communication Unit for Musculoskeletal Health (FORMI), Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, 0424 Oslo, Norway
| | - Helgi Jonsson
- Department of Medicine, Landspitali The National University Hospital of Iceland, 108 Reykjavik, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Margreet Kloppenburg
- Departments of Rheumatology and Clinical Epidemiology, Leiden University Medical Center, 9600, 23OORC Leiden, the Netherlands
| | - Almut Luetge
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | | | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London SE1 7EH, UK
| | - Rob R G H H Nelissen
- Department of Orthopaedics, Leiden University Medical Center, 9600, 23OORC Leiden, the Netherlands
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julia Steinberg
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW 1340, Australia
| | - Hiroshi Takuwa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo 108-8639, Japan; Department of Orthopedic Surgery, Shimane University, Shimane 693-8501, Japan
| | - Laurent F Thomas
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway; BioCore-Bioinformatics Core Facility, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Margo Tuerlings
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - George C Babis
- 2(nd) Department of Orthopaedics, National and Kapodistrian University of Athens, Medical School, Nea Ionia General Hospital Konstantopouleio, 14233 Athens, Greece
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jae Hee Kang
- Department of Medicine, Brigham and Women's Hospital, 181 Longwood Ave, Boston, MA 02115, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Steven A Lietman
- Musculoskeletal Institute, Geisinger Health System, Danville, PA 17822, USA
| | - Dino Samartzis
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Pokfulam, Hong Kong, China; Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - Kari Stefansson
- deCODE Genetics/Amgen Inc., 102 Reykjavik, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen Inc., 102 Reykjavik, Iceland; Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Jonathan H Tobias
- Musculoskeletal Research Unit, Translation Health Sciences, Bristol Medical School, University of Bristol, Southmead Hospital, Bristol BS10 5NB, UK; MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, Medical Center, 3015CN Rotterdam, the Netherlands
| | - Bendik Winsvold
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway; Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | - John-Anker Zwart
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, 0450 Oslo, Norway
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Pak Chung Sham
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | - Tom R Gaunt
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, University of Manchester, Manchester M13 9LJ, UK
| | - Ana M Valdes
- Faculty of Medicine and Health Sciences, School of Medicine, University of Nottingham, Nottingham, Nottinghamshire NG5 1PB, UK
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa 411 10, Greece
| | - Kathryn S E Cheah
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo 108-8639, Japan
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7491 Trondheim, Norway; HUNT Research Center, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - J Mark Wilkinson
- Department of Oncology and Metabolism and Healthy Lifespan Institute, University of Sheffield, Sheffield S10 2RX, UK
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Postzone S05-P Leiden University Medical Center, 2333ZC Leiden, the Netherlands
| | - Ming Ta Michael Lee
- Genomic Medicine Institute, Geisinger Health System, Danville, PA 17822, USA; Institute of Biomedical Sciences, Academia Sinica, 115 Taipei, Taiwan
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus MC, Medical Center, 3015CN Rotterdam, the Netherlands
| | | | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany; TUM School of Medicine, Technical University of Munich and Klinikum Rechts der Isar, 81675 Munich, Germany.
| |
Collapse
|
28
|
Wilkinson JM, Zeggini E. The Genetic Epidemiology of Joint Shape and the Development of Osteoarthritis. Calcif Tissue Int 2021; 109:257-276. [PMID: 32393986 PMCID: PMC8403114 DOI: 10.1007/s00223-020-00702-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023]
Abstract
Congruent, low-friction relative movement between the articulating elements of a synovial joint is an essential pre-requisite for sustained, efficient, function. Where disorders of joint formation or maintenance exist, mechanical overloading and osteoarthritis (OA) follow. The heritable component of OA accounts for ~ 50% of susceptible risk. Although almost 100 genetic risk loci for OA have now been identified, and the epidemiological relationship between joint development, joint shape and osteoarthritis is well established, we still have only a limited understanding of the contribution that genetic variation makes to joint shape and how this modulates OA risk. In this article, a brief overview of synovial joint development and its genetic regulation is followed by a review of current knowledge on the genetic epidemiology of established joint shape disorders and common shape variation. A summary of current genetic epidemiology of OA is also given, together with current evidence on the genetic overlap between shape variation and OA. Finally, the established genetic risk loci for both joint shape and osteoarthritis are discussed.
Collapse
Affiliation(s)
- J Mark Wilkinson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, UK.
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
29
|
Steinberg J, Southam L, Fontalis A, Clark MJ, Jayasuriya RL, Swift D, Shah KM, Brooks RA, McCaskie AW, Wilkinson JM, Zeggini E. Linking chondrocyte and synovial transcriptional profile to clinical phenotype in osteoarthritis. Ann Rheum Dis 2021; 80:1070-1074. [PMID: 33903094 PMCID: PMC8292595 DOI: 10.1136/annrheumdis-2020-219760] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 03/30/2021] [Accepted: 04/11/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To determine how gene expression profiles in osteoarthritis joint tissues relate to patient phenotypes and whether molecular subtypes can be reproducibly captured by a molecular classification algorithm. METHODS We analysed RNA sequencing data from cartilage and synovium in 113 osteoarthritis patients, applying unsupervised clustering and Multi-Omics Factor Analysis to characterise transcriptional profiles. We tested the association of the molecularly defined patient subgroups with clinical characteristics from electronic health records. RESULTS We detected two patient subgroups in low-grade cartilage (showing no/minimal degeneration, cartilage normal/softening only), with differences associated with inflammation, extracellular matrix-related and cell adhesion pathways. The high-inflammation subgroup was associated with female sex (OR 4.12, p=0.0024) and prescription of proton pump inhibitors (OR 4.21, p=0.0040). We identified two independent patient subgroupings in osteoarthritis synovium: one related to inflammation and the other to extracellular matrix and cell adhesion processes. A seven-gene classifier including MMP13, APOD, MMP2, MMP1, CYTL1, IL6 and C15orf48 recapitulated the main axis of molecular heterogeneity in low-grade knee osteoarthritis cartilage (correlation ρ=-0.88, p<10-10) and was reproducible in an independent patient cohort (ρ=-0.85, p<10-10). CONCLUSIONS These data support the reproducible stratification of osteoarthritis patients by molecular subtype and the exploration of new avenues for tailored treatments.
Collapse
Affiliation(s)
- Julia Steinberg
- Institute for Translational Genomics, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
- Daffodil Centre, The University of Sydney, a joint venture with Cancer Council NSW, Sydney, New South Wales, Australia
- Wellcome Sanger Institute, Hinxton, UK
| | - Lorraine Southam
- Institute for Translational Genomics, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
- Wellcome Sanger Institute, Hinxton, UK
| | - Andreas Fontalis
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Matthew J Clark
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Raveen L Jayasuriya
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Diane Swift
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Karan M Shah
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
| | - Roger A Brooks
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Cambridge, UK
| | - Andrew W McCaskie
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Cambridge, UK
| | - Jeremy Mark Wilkinson
- Department of Oncology and Metabolism, The University of Sheffield, Sheffield, UK
- Centre for Integrated Research into Musculoskeletal Ageing and Sheffield Healthy Lifespan Institute, University of Sheffield, Sheffield, UK
| | - Eleftheria Zeggini
- Institute for Translational Genomics, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
- Wellcome Sanger Institute, Hinxton, UK
- Translational Genomics, Klinikum rechts der Isar der Technischen Universitat Munchen, Munchen, Germany
| |
Collapse
|
30
|
Abstract
Pain is an immense clinical and societal challenge, and the key to understanding and treating it is variability. Robust interindividual differences are consistently observed in pain sensitivity, susceptibility to developing painful disorders, and response to analgesic manipulations. This review examines the causes of this variability, including both organismic and environmental sources. Chronic pain development is a textbook example of a gene-environment interaction, requiring both chance initiating events (e.g., trauma, infection) and more immutable risk factors. The focus is on genetic factors, since twin studies have determined that a plurality of the variance likely derives from inherited genetic variants, but sex, age, ethnicity, personality variables, and environmental factors are also considered.
Collapse
Affiliation(s)
- Jeffrey S Mogil
- Departments of Psychology and Anesthesia, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec H3A 1B1, Canada;
| |
Collapse
|
31
|
Current Epidemiology and Risk Factors for the Development of Hand Osteoarthritis. Curr Rheumatol Rep 2021; 23:61. [PMID: 34216294 DOI: 10.1007/s11926-021-01025-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW Hand osteoarthritis (hand OA), the most common peripheral arthritis in the world, is less studied than osteoarthritis (OA) of the knee and hip. However, it is uniquely situated to offer novel insight into OA as a disease process by removing weight-bearing as a confounder of systemic disease mechanisms. Here we review the epidemiology of hand OA and key risk factors for its development. RECENT FINDINGS Mounting evidence points to obesity as an important risk factor for hand OA development, with new evidence implicating a role for leptin and serum fatty acids. Disease progression in hand OA and specifically the erosive OA subtype may be associated with diabetes. New evidence supports an association between cardiovascular disease progression and symptomatic hand OA. Alcohol use may be associated with increased synovitis and erosive hand OA. Differences in ethnical distributions of hand OA have become more apparent, with a lower prevalence in Black patients compared to White patients. Novel genetic insights implicating the WNT gene pathway and IL-1β have led to novel potential targets in hand OA pathogenesis. Hand OA is a heterogeneous disease with many modifiable and non-modifiable risk factors that can determine disease severity and shed light on disease pathogenesis.
Collapse
|
32
|
Kehayova YS, Watson E, Wilkinson JM, Loughlin J, Rice SJ. Genetic and Epigenetic Interplay Within a COLGALT2 Enhancer Associated With Osteoarthritis. Arthritis Rheumatol 2021; 73:1856-1865. [PMID: 33760386 DOI: 10.1002/art.41738] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/11/2021] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The osteoarthritis (OA)-associated single-nucleotide polymorphism (SNP) rs11583641 is located in COLGALT2, encoding a posttranslational modifier of collagen. In cartilage, the SNP genotype correlates with DNA methylation in a putative enhancer. This study was undertaken to characterize the mechanistic relationship between rs11583641, the putative enhancer, and COLGALT2 expression using cartilage samples from human patients and a chondrocyte cell model. METHODS Nucleic acids were extracted from articular cartilage samples obtained from patients with OA (n = 137). Samples were genotyped, and DNA methylation was quantified at 12 CpGs using pyrosequencing. The putative enhancer was deleted in Tc28a2 chondrocytes using clustered regularly interspaced short palindromic repeat/Cas9, and the impact on nearby gene expression was determined using real-time quantitative polymerase chain reaction. Targeted modulation of the epigenome using catalytically dead Cas9 (dCas9) constructs fused to DNA methyltransferase 3a or ten-eleven translocase 1 allowed for the investigation of a causal relationship between DNA methylation and enhancer activity. RESULTS The genotype at rs11583641 correlated with DNA methylation at 3 CpGs, and the presence of the OA risk allele, C, corresponded to reduced levels of methylation. Deletion of the enhancer resulted in a 2.7-fold reduction in COLGALT2 expression. Targeted methylation and demethylation of the CpGs had antagonistic effects on COLGALT2 expression. An allelic imbalance in the expression of COLGALT2 was identified in the cartilage from patients with OA, with relative overexpression of the OA risk allele. Allelic expression ratios correlated with DNA methylation at 4 CpGs. CONCLUSION COLGALT2 is a target of OA genetic risk at this locus. The genotype at rs11583641 impacts DNA methylation in a gene enhancer, which, in turn, modulates COLGALT2 expression. COLGALT2 encodes an enzyme that initiates posttranslational glycosylation of collagens and is therefore a compelling OA susceptibility target.
Collapse
Affiliation(s)
- Yulia S Kehayova
- International Centre for Life and Newcastle University, Newcastle-upon-Tyne, UK, and MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing and University of Liverpool, Liverpool, UK
| | - Emily Watson
- International Centre for Life and Newcastle University, Newcastle-upon-Tyne, UK, and MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing and University of Liverpool, Liverpool, UK
| | - J Mark Wilkinson
- University of Sheffield, Sheffield, UK, and MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing and University of Liverpool, Liverpool, UK
| | - John Loughlin
- International Centre for Life and Newcastle University, Newcastle-upon-Tyne, UK, and MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing and University of Liverpool, Liverpool, UK
| | - Sarah J Rice
- International Centre for Life and Newcastle University, Newcastle-upon-Tyne, UK, and MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing and University of Liverpool, Liverpool, UK
| |
Collapse
|
33
|
Kazmers NH, Meeks HD, Novak KA, Yu Z, Fulde GL, Thomas JL, Barker T, Jurynec MJ. Familial Clustering of Erosive Hand Osteoarthritis in a Large Statewide Cohort. Arthritis Rheumatol 2021; 73:440-447. [PMID: 32940959 PMCID: PMC7914133 DOI: 10.1002/art.41520] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/04/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Erosive hand osteoarthritis (OA) is a severe and rapidly progressing subset of hand OA. Its etiology remains largely unknown, which has hindered development of successful treatments. This study was undertaken to test the hypothesis that erosive hand OA demonstrates familial clustering in a large statewide population linked to genealogical records, and to determine the association of potential risk factors with erosive hand OA. METHODS Patients diagnosed as having erosive hand OA were identified by searching 4,741,840 unique medical records from a comprehensive statewide database, the Utah Population Database (UPDB). Affected individuals were mapped to pedigrees to identify high-risk families with excess clustering of erosive hand OA as defined by a familial standardized incidence ratio (FSIR) of ≥2.0. The magnitude of familial risk of erosive hand OA in related individuals was calculated using Cox regression models. Association of potential erosive hand OA risk factors was analyzed using multivariate conditional logistic regression and logistic regression models. RESULTS We identified 703 affected individuals linked to 240 unrelated high-risk pedigrees with excess clustering of erosive hand OA (FSIR ≥2.0, P < 0.05). The relative risk of developing erosive hand OA was significantly elevated in first-degree relatives (P < 0.001). There were significant associations between a diagnosis of erosive hand OA and age, sex, diabetes, and obesity (all P < 0.05). CONCLUSION Familial clustering of erosive hand OA observed in a statewide database indicates a potential genetic contribution to the etiology of the disease. Age, sex, diabetes, and obesity are risk factors for erosive hand OA. Identification of causal gene variants in these high-risk families may provide insight into the genes and pathways that contribute to erosive hand OA onset and progression.
Collapse
Affiliation(s)
- Nikolas H. Kazmers
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108
| | - Huong D. Meeks
- Population Science, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, UT 84112
| | - Kendra A. Novak
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108
| | - Zhe Yu
- Population Science, Huntsman Cancer Institute, University of Utah Health, Salt Lake City, UT 84112
| | - Gail L. Fulde
- Intermountain Healthcare, Precision Genomics, St. George, UT 84790
| | - Joy L. Thomas
- Intermountain Healthcare, Precision Genomics, St. George, UT 84790
| | - Tyler Barker
- Intermountain Healthcare, Precision Genomics, Murray, UT 84107
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112
| | - Michael J. Jurynec
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108
| |
Collapse
|
34
|
Jones G, Trajanoska K, Santanasto AJ, Stringa N, Kuo CL, Atkins JL, Lewis JR, Duong T, Hong S, Biggs ML, Luan J, Sarnowski C, Lunetta KL, Tanaka T, Wojczynski MK, Cvejkus R, Nethander M, Ghasemi S, Yang J, Zillikens MC, Walter S, Sicinski K, Kague E, Ackert-Bicknell CL, Arking DE, Windham BG, Boerwinkle E, Grove ML, Graff M, Spira D, Demuth I, van der Velde N, de Groot LCPGM, Psaty BM, Odden MC, Fohner AE, Langenberg C, Wareham NJ, Bandinelli S, van Schoor NM, Huisman M, Tan Q, Zmuda J, Mellström D, Karlsson M, Bennett DA, Buchman AS, De Jager PL, Uitterlinden AG, Völker U, Kocher T, Teumer A, Rodriguéz-Mañas L, García FJ, Carnicero JA, Herd P, Bertram L, Ohlsson C, Murabito JM, Melzer D, Kuchel GA, Ferrucci L, Karasik D, Rivadeneira F, Kiel DP, Pilling LC. Genome-wide meta-analysis of muscle weakness identifies 15 susceptibility loci in older men and women. Nat Commun 2021; 12:654. [PMID: 33510174 PMCID: PMC7844411 DOI: 10.1038/s41467-021-20918-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023] Open
Abstract
Low muscle strength is an important heritable indicator of poor health linked to morbidity and mortality in older people. In a genome-wide association study meta-analysis of 256,523 Europeans aged 60 years and over from 22 cohorts we identify 15 loci associated with muscle weakness (European Working Group on Sarcopenia in Older People definition: n = 48,596 cases, 18.9% of total), including 12 loci not implicated in previous analyses of continuous measures of grip strength. Loci include genes reportedly involved in autoimmune disease (HLA-DQA1 p = 4 × 10-17), arthritis (GDF5 p = 4 × 10-13), cell cycle control and cancer protection, regulation of transcription, and others involved in the development and maintenance of the musculoskeletal system. Using Mendelian randomization we report possible overlapping causal pathways, including diabetes susceptibility, haematological parameters, and the immune system. We conclude that muscle weakness in older adults has distinct mechanisms from continuous strength, including several pathways considered to be hallmarks of ageing.
Collapse
Affiliation(s)
- Garan Jones
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adam J Santanasto
- University of Pittsburgh, Department of Epidemiology, Pittsburgh, PA, USA
| | - Najada Stringa
- Department of Epidemiology and Biostatistics, Amsterdam UMC- Vrije Universiteit, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Chia-Ling Kuo
- Biostatistics Center, Connecticut Convergence Institute for Translation in Regenerative Engineering, UConn Health, Farmington, CT, USA
| | - Janice L Atkins
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Joshua R Lewis
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
- School fo Public Health University of Sydney, Sydney, NSW, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
| | - ThuyVy Duong
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shengjun Hong
- Lübeck Interdisciplinary Plattform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Mary L Biggs
- Cardiovascular Health Research Unit, Department of Medicine, and Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Chloe Sarnowski
- Biostatistics Department, Boston University School of Public Health, Boston, MA, USA
| | - Kathryn L Lunetta
- Biostatistics Department, Boston University School of Public Health, Boston, MA, USA
| | - Toshiko Tanaka
- Longitudinal Study Section, Translational Gerontology branch, National Institute on Aging, Baltimore, MD, USA
| | - Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ryan Cvejkus
- University of Pittsburgh, Department of Epidemiology, Pittsburgh, PA, USA
| | - Maria Nethander
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sahar Ghasemi
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Jingyun Yang
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Stefan Walter
- Department of Medicine and Public Health, Rey Juan Carlos University, Madrid, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain
| | - Kamil Sicinski
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, WI, USA
| | - Erika Kague
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | | | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - B Gwen Windham
- Department of Medicine/Geriatrics, University of Mississippi School of Medicine, Jackson, MS, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Misa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, 27516, USA
| | - Dominik Spira
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Department of Endocrinology and Metabolism, Berlin, Germany
| | - Ilja Demuth
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health, Department of Endocrinology and Metabolism, Berlin, Germany
- Charité - Universitätsmedizin Berlin, BCRT - Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Nathalie van der Velde
- Department of Internal Medicine, Section of Geriatric Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Lisette C P G M de Groot
- Wageningen University, Division of Human Nutrition, PO-box 17, 6700 AA, Wageningen, The Netherlands
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Michelle C Odden
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA, USA
| | - Alison E Fohner
- Department of Epidemiology and Institute of Public Genetics, University of Washington, Seattle, WA, USA
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | | | - Natasja M van Schoor
- Department of Epidemiology and Biostatistics, Amsterdam UMC- Vrije Universiteit, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Martijn Huisman
- Department of Epidemiology and Biostatistics, Amsterdam UMC- Vrije Universiteit, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Qihua Tan
- Epidemiology and Biostatistics, Department of Public Health, Faculty of Health Science, University of Southern Denmark, Odense, Denmark
| | - Joseph Zmuda
- University of Pittsburgh, Department of Epidemiology, Pittsburgh, PA, USA
| | - Dan Mellström
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Geriatric Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department of Orthopedics and Clinical Sciences, Lund University, Skåne University Hospital, Malmö, Sweden
| | - David A Bennett
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center & Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Philip L De Jager
- Center for Translational and Systems Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Andre G Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Thomas Kocher
- Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, University Medicine Greifswald, Greifswald, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Leocadio Rodriguéz-Mañas
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain
- Department of Geriatrics, Getafe University Hospital, Getafe, Spain
| | - Francisco J García
- CIBER of Frailty and Healthy Aging (CIBERFES), Madrid, Spain
- Department of Geriatrics, Hospital Virgen del Valle, Complejo Hospitalario de Toledo, Toledo, Spain
| | | | - Pamela Herd
- Professor of Public Policy, Georgetown University, Washington, DC, USA
| | - Lars Bertram
- Lübeck Interdisciplinary Plattform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska University Hospital, Department of Drug Treatment, Gothenburg, Sweden
| | - Joanne M Murabito
- Section of General Internal Medicine, Boston University School of Medicine, Boston, MA, USA
| | - David Melzer
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - George A Kuchel
- Center on Aging, University of Connecticut Health, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | | | - David Karasik
- Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA, USA
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Douglas P Kiel
- Marcus Institute for Aging Research, Hebrew SeniorLife and Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Broad Institute of MIT & Harvard, Boston, MA, USA
| | - Luke C Pilling
- Epidemiology and Public Health Group, Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Osteoarthritis is a heterogeneous, multifactorial condition regulated by complex biological interactions at multiple levels. Comprehensive understanding of these regulatory interactions is required to develop feasible advances to improve patient outcomes. Improvements in technology have made extensive genomic, transcriptomic, epigenomic, proteomic, and metabolomic profiling possible. This review summarizes findings over the past 20 months related to omics technologies in osteoarthritis and examines how using a multiomics approach is necessary for advancing our understanding of osteoarthritis as a disease to improve precision osteoarthritis treatments. RECENT FINDINGS Using the search terms 'genomics' or 'transcriptomics' or 'epigenomics' or 'proteomics' or 'metabolomics' and 'osteoarthritis' from January 1, 2018 to August 31, 2019, we identified advances in omics approaches applied to osteoarthritis. Trends include untargeted whole genome, transcriptome, proteome, and metabolome analyses leading to identification of novel molecular signatures, cell subpopulations and multiomics validation approaches. SUMMARY To address the complexity of osteoarthritis, integration of multitissue analyses by multiomics approaches with the inclusion of longitudinal clinical data is necessary for a comprehensive understanding of the disease process, and for appropriate development of efficacious diagnostics, prognostics, and biotherapeutics.
Collapse
|
36
|
Sutter PA, Karki S, Crawley I, Singh V, Bernt KM, Rowe DW, Crocker SJ, Bayarsaihan D, Guzzo RM. Mesenchyme-specific loss of Dot1L histone methyltransferase leads to skeletal dysplasia phenotype in mice. Bone 2021; 142:115677. [PMID: 33022452 PMCID: PMC7744341 DOI: 10.1016/j.bone.2020.115677] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/20/2020] [Accepted: 10/01/2020] [Indexed: 12/17/2022]
Abstract
Chromatin modifying enzymes play essential roles in skeletal development and bone maintenance, and deregulation of epigenetic mechanisms can lead to skeletal growth and malformation disorders. Here, we report a novel skeletal dysplasia phenotype in mice with conditional loss of Disruptor of telomeric silencing 1-like (Dot1L) histone methyltransferase in limb mesenchymal progenitors and downstream descendants. Phenotypic characterizations of mice with Dot1L inactivation by Prrx1-Cre (Dot1L-cKOPrrx1) revealed limb shortening, abnormal bone morphologies, and forelimb dislocations. Our in vivo and in vitro data support a crucial role for Dot1L in regulating growth plate chondrocyte proliferation and differentiation, extracellular matrix production, and secondary ossification center formation. Micro-computed tomography analysis of femurs revealed that partial loss of Dot1L expression is sufficient to impair trabecular bone formation and microarchitecture in young mice. Moreover, RNAseq analysis of Dot1L deficient chondrocytes implicated Dot1L in the regulation of key genes and pathways necessary to promote cell cycle regulation and skeletal growth. Collectively, our data show that early expression of Dot1L in limb mesenchyme provides essential regulatory control of endochondral bone morphology, growth, and stability.
Collapse
Affiliation(s)
- Pearl A Sutter
- Department of Neuroscience, School of Medicine, University of Connecticut Health, Farmington, CT, United States of America
| | - Sangita Karki
- Department of Neuroscience, School of Medicine, University of Connecticut Health, Farmington, CT, United States of America
| | - Ilan Crawley
- Department of Neuroscience, School of Medicine, University of Connecticut Health, Farmington, CT, United States of America
| | - Vijender Singh
- Bioinformatics, University of Connecticut, Storrs, CT, United States of America
| | - Kathrin M Bernt
- Division of Pediatric Oncology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania and Abramson Cancer Center, Philadelphia, PA, United States of America
| | - David W Rowe
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health, Farmington, CT, United States of America; Center for Regenerative Medicine and Skeletal Development, Farmington, CT, United States of America
| | - Stephen J Crocker
- Department of Neuroscience, School of Medicine, University of Connecticut Health, Farmington, CT, United States of America
| | - Dashzeveg Bayarsaihan
- Department of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health, Farmington, CT, United States of America; Center for Regenerative Medicine and Skeletal Development, Farmington, CT, United States of America
| | - Rosa M Guzzo
- Department of Neuroscience, School of Medicine, University of Connecticut Health, Farmington, CT, United States of America.
| |
Collapse
|
37
|
Boer CG, Yau MS, Rice SJ, Coutinho de Almeida R, Cheung K, Styrkarsdottir U, Southam L, Broer L, Wilkinson JM, Uitterlinden AG, Zeggini E, Felson D, Loughlin J, Young M, Capellini TD, Meulenbelt I, van Meurs JB. Genome-wide association of phenotypes based on clustering patterns of hand osteoarthritis identify WNT9A as novel osteoarthritis gene. Ann Rheum Dis 2020; 80:367-375. [PMID: 33055079 PMCID: PMC7892373 DOI: 10.1136/annrheumdis-2020-217834] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
Background Despite recent advances in the understanding of the genetic architecture of osteoarthritis (OA), only two genetic loci have been identified for OA of the hand, in part explained by the complexity of the different hand joints and heterogeneity of OA pathology. Methods We used data from the Rotterdam Study (RSI, RSII and RSIII) to create three hand OA phenotypes based on clustering patterns of radiographic OA severity to increase power in our modest discovery genome-wide association studies in the RS (n=8700), and sought replication in an independent cohort, the Framingham Heart Study (n=1203). We used multiple approaches that leverage different levels of information and functional data to further investigate the underlying biological mechanisms and candidate genes for replicated loci. We also attempted to replicate known OA loci at other joint sites, including the hips and knees. Results We found two novel genome-wide significant loci for OA in the thumb joints. We identified WNT9A as a possible novel causal gene involved in OA pathogenesis. Furthermore, several previously identified genetic loci for OA seem to confer risk for OA across multiple joints: TGFa, RUNX2, COL27A1, ASTN2, IL11 and GDF5 loci. Conclusions We identified a robust novel genetic locus for hand OA on chromosome 1, of which WNT9A is the most likely causal gene. In addition, multiple genetic loci were identified to be associated with OA across multiple joints. Our study confirms the potential for novel insight into the genetic architecture of OA by using biologically meaningful stratified phenotypes.
Collapse
Affiliation(s)
- Cindy Germaine Boer
- Department of Internal Medicine, Genetic Laboratories, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Michelle S Yau
- Hebrew SeniorLife, Beth Israel Deaconess Medical Center. Harvard Medical School, Hinda and Arthur Marcus Institute for Aging Research, Boston, Massachusetts, USA.,Department of Rheumatology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Sarah J Rice
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Rodrigo Coutinho de Almeida
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kathleen Cheung
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.,Newcastle University, Bioinformatics Support Unit, Newcastle upon Tyne, UK
| | | | - Lorraine Southam
- Institute of Translational Genomics, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - Linda Broer
- Department of Internal Medicine, Genetic Laboratories, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | | | - André G Uitterlinden
- Department of Internal Medicine, Genetic Laboratories, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
| | - David Felson
- Arthritis Research UK Epidemiology Unit, The University of Manchester, Manchester, UK
| | - John Loughlin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Mariel Young
- Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| | | | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Joyce Bj van Meurs
- Department of Internal Medicine, Genetic Laboratories, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
38
|
Liu L, Wang S, Wen Y, Li P, Cheng S, Ma M, Zhang L, Cheng B, Qi X, Liang C, Zhang F. Assessing the genetic relationships between osteoarthritis and human plasma proteins: a large scale genetic correlation scan. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:677. [PMID: 32617297 PMCID: PMC7327363 DOI: 10.21037/atm-19-4643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Osteoarthritis (OA) is a multifactorial complex disease. The impact of plasma proteins on OA remains elusive now. Methods The UK Biobank genome-wide association study data of OA was used here. Genome-wide SNP genotyping was performed using the Affymetrix UK BiLEVE Axiom or UK Biobank Axiom array. Equally, the GWAS summary data of 3,622 plasma proteins was derived from a recently published study. Consequently, linkage disequilibrium score regression (LD score regression) analysis was performed to evaluate the genetic correlation between each plasma protein and different sites of OA. Results Several suggestive plasma proteins were identified for OA. For hand OA, evidence of genetic correlation was observed for inter-alpha-trypsin inhibitor heavy chain H1 (coefficient =−0.3854, P value =0.0198), multiple inositol polyphosphate phosphatase 1 (coefficient =−1.1721, P value =0.0303). For hip OA, 7 suggestive genetic correlation signals were observed, such as Transmembrane glycoprotein NMB (coefficient =0.6944, P value =0.0098), Endothelial cell-specific molecule 1 (coefficient =0.6337, P value =0.03). For Knee OA, 12 suggestive genetic correlation signals were identified, including Elafin (coefficient =−0.5562, P value =0.0092), Interleukin-16 (coefficient =0.3949, P value =0.0435). Conclusions We investigated the genetic correlations between plasma proteins and different sites of OA in a systematic way. Our results provide novel evidence that OA is a heterogeneous disease.
Collapse
Affiliation(s)
- Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Sen Wang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Ping Li
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Mei Ma
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Lu Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Bolun Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xin Qi
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Chujun Liang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
39
|
Johnsen MB, Magnusson K, Børte S, Gabrielsen ME, Winsvold BS, Skogholt AH, Thomas L, Storheim K, Hveem K, Zwart JA. Development and validation of a prediction model for incident hand osteoarthritis in the HUNT study. Osteoarthritis Cartilage 2020; 28:932-940. [PMID: 32360252 DOI: 10.1016/j.joca.2020.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/01/2020] [Accepted: 04/21/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To develop and externally validate prediction models for incident hand osteoarthritis (OA) in a large population-based cohort of middle aged and older men and women. DESIGN We included 17,153 men and 18,682 women from a population-based cohort, aged 35-70 years at baseline (1995-1997). Incident hand OA were obtained from diagnostic codes in the Norwegian National Patient Register (1995-2018). We studied whether a range of self-reported and clinically measured predictors could predict hand OA, using the Area Under the receiver-operating Curve (AUC) from logistic regression. External validation of an existing prediction model for male hand OA was tested on discrimination in a sample of men. Bootstrapping was used to avoid overfitting. RESULTS The model for men showed modest discriminatory ability (AUC = 0.67, 95% CI 0.62-0.71). Adding a genetic risk score did not improve prediction. Similar discrimination was observed in the model for women (AUC = 0.62, 95% CI 0.59-0.64). Prediction was not improved by adding a genetic risk score or hormonal and reproductive factors. Applying external validation, similar results were observed among men in HUNT (The Nord-Trøndelag Health Study) as in the developmental sample (AUC = 0.62, 95% CI 0.57-0.65). CONCLUSION We developed prediction models for incident hand OA in men and women. For women, the model included body mass index (BMI), heavy physical work, high physical activity and perceived poor health. The model showed moderate discrimination. For men, we have shown that a prediction model including BMI, education and information on sleep can predict incident hand OA in several populations with moderate discriminative ability.
Collapse
Affiliation(s)
- M B Johnsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway; K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.
| | - K Magnusson
- Lund University, Faculty of Medicine, Department of Clinical Sciences, Clinical Epidemiology Unit, Lund, Orthopaedics, Lund, Sweden; National Advisory Unit on Rehabilitation in Rheumatology, Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway.
| | - S Børte
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway; K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.
| | - M E Gabrielsen
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.
| | - B S Winsvold
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway; K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.
| | - A H Skogholt
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.
| | - L Thomas
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
| | - K Storheim
- Research and Communication Unit for Musculoskeletal Health, Oslo University Hospital, Oslo, Norway.
| | - K Hveem
- K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.
| | - J-A Zwart
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway; K. G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
40
|
Richard D, Liu Z, Cao J, Kiapour AM, Willen J, Yarlagadda S, Jagoda E, Kolachalama VB, Sieker JT, Chang GH, Muthuirulan P, Young M, Masson A, Konrad J, Hosseinzadeh S, Maridas DE, Rosen V, Krawetz R, Roach N, Capellini TD. Evolutionary Selection and Constraint on Human Knee Chondrocyte Regulation Impacts Osteoarthritis Risk. Cell 2020; 181:362-381.e28. [PMID: 32220312 PMCID: PMC7179902 DOI: 10.1016/j.cell.2020.02.057] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 12/10/2019] [Accepted: 02/26/2020] [Indexed: 02/06/2023]
Abstract
During human evolution, the knee adapted to the biomechanical demands of bipedalism by altering chondrocyte developmental programs. This adaptive process was likely not without deleterious consequences to health. Today, osteoarthritis occurs in 250 million people, with risk variants enriched in non-coding sequences near chondrocyte genes, loci that likely became optimized during knee evolution. We explore this relationship by epigenetically profiling joint chondrocytes, revealing ancient selection and recent constraint and drift on knee regulatory elements, which also overlap osteoarthritis variants that contribute to disease heritability by tending to modify constrained functional sequence. We propose a model whereby genetic violations to regulatory constraint, tolerated during knee development, lead to adult pathology. In support, we discover a causal enhancer variant (rs6060369) present in billions of people at a risk locus (GDF5-UQCC1), showing how it impacts mouse knee-shape and osteoarthritis. Overall, our methods link an evolutionarily novel aspect of human anatomy to its pathogenesis.
Collapse
Affiliation(s)
- Daniel Richard
- Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Zun Liu
- Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Jiaxue Cao
- Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Ata M Kiapour
- Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica Willen
- Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | | | - Evelyn Jagoda
- Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Vijaya B Kolachalama
- Department of Medicine, Boston University School of Medicine, Boston, MA 02115, USA; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02115, USA; Hariri Institute for Computing and Computational Science and Engineering, Boston University, Boston, MA 02115, USA
| | - Jakob T Sieker
- Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Pathology and Laboratory Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Gary H Chang
- Department of Medicine, Boston University School of Medicine, Boston, MA 02115, USA
| | | | - Mariel Young
- Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Anand Masson
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Johannes Konrad
- Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shayan Hosseinzadeh
- Orthopaedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David E Maridas
- Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Vicki Rosen
- Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Roman Krawetz
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Neil Roach
- Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Terence D Capellini
- Human Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
41
|
Rice SJ, Beier F, Young DA, Loughlin J. Interplay between genetics and epigenetics in osteoarthritis. Nat Rev Rheumatol 2020; 16:268-281. [PMID: 32273577 DOI: 10.1038/s41584-020-0407-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 12/15/2022]
Abstract
Research into the molecular genetics of osteoarthritis (OA) has been substantially bolstered in the past few years by the implementation of powerful genome-wide scans that have revealed a large number of novel risk loci associated with the disease. This refreshing wave of discovery has occurred concurrently with epigenetic studies of joint tissues that have examined DNA methylation, histone modifications and regulatory RNAs. These epigenetic analyses have involved investigations of joint development, homeostasis and disease and have used both human samples and animal models. What has become apparent from a comparison of these two complementary approaches is that many OA genetic risk signals interact with, map to or correlate with epigenetic mediators. This discovery implies that epigenetic mechanisms, and their effect on gene expression, are a major conduit through which OA genetic risk polymorphisms exert their functional effects. This observation is particularly exciting as it provides mechanistic insight into OA susceptibility. Furthermore, this knowledge reveals avenues for attenuating the negative effect of risk-conferring alleles by exposing the epigenome as an exploitable target for therapeutic intervention in OA.
Collapse
Affiliation(s)
- Sarah J Rice
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Frank Beier
- Department of Physiology and Pharmacology, The University of Western Ontario, London, ON, Canada.,Western Bone and Joint Institute, The University of Western Ontario, London, ON, Canada
| | - David A Young
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - John Loughlin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
42
|
Generation and characterization of human induced pluripotent stem cells (iPSCs) from hand osteoarthritis patient-derived fibroblasts. Sci Rep 2020; 10:4272. [PMID: 32144293 PMCID: PMC7060311 DOI: 10.1038/s41598-020-61071-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/17/2020] [Indexed: 12/21/2022] Open
Abstract
Knowledge and research results about hand osteoarthritis (hOA) are limited due to the lack of samples and animal models of the disease. Here, we report the generation of two induced pluripotent stem cell (iPSC)-lines from patients with radiographic hOA. Furthermore, we wondered whether these iPSC-lines carried single nucleotide polymorphisms (SNPs) within genes that have been associated with hOA. Finally, we performed chondrogenic differentiation of the iPSCs in order to prove their usefulness as cellular models of the disease. We performed a non-integrative reprogramming of dermal fibroblasts obtained from two patients with radiographic rhizarthrosis and non-erosive hOA by introducing the transcriptional factors Oct4, Sox2, Klf4 and c-Myc using Sendai virus. After reprogramming, embryonic stem cell-like colonies emerged in culture, which fulfilled all the criteria to be considered iPSCs. Both iPSC-lines carried variants associated with hOA in the four studied genes and showed differences in their chondrogenic capacity when compared with a healthy control iPSC-line. To our knowledge this is the first time that the generation of iPSC-lines from patients with rhizarthrosis and non-erosive hOA is reported. The obtained iPSC-lines might enable us to model the disease in vitro, and to deeper study both the molecular and cellular mechanisms underlying hOA.
Collapse
|
43
|
Osteoarthritis year in review 2019: genetics, genomics and epigenetics. Osteoarthritis Cartilage 2020; 28:275-284. [PMID: 31874234 DOI: 10.1016/j.joca.2019.11.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 02/02/2023]
Abstract
Although osteoarthritis (OA) aetiology is complex, genetic, genomic and epigenetic studies published within the last decade have advanced our understanding of the molecular processes underlying this common musculoskeletal disease. The purpose of this narrative review is to highlight the key research articles within the OA genetics, genomics and epigenetics fields that were published between April 2018 and April 2019. The review focuses on the identification of new OA genetic risk loci, genomics techniques that have been used for the first time in human cartilage and new publicly available databases, and datasets that will aid OA functional studies. Fifty-six new OA susceptibility loci were identified by two large scale genome wide association study meta-analyses, increasing the number of genome-wide significant risk loci to 90. OA risk variants are enriched near genes involved in skeletal development and morphology, and show genetic overlap with height, hip shape, bone area and developmental dysplasia of the hip. Several functional studies of OA loci were published, including a genome-wide analysis of genetic variation on cartilage gene expression. A specialised data portal for exploring cross-species skeletal transcriptomic datasets has been developed, and the first use of cartilage single cell RNAseq analysis reported. This year also saw the systematic identification of all microRNAs, long non-coding RNAs and circular RNAs expressed in human OA cartilage. Putative transcriptional regulatory regions have been mapped in human chondrocytes genome-wide, providing a dataset that will facilitate the prioritisation and characterisation of OA genetic and epigenetic loci.
Collapse
|
44
|
Attur M, Zhou H, Samuels J, Krasnokutsky S, Yau M, Scher JU, Doherty M, Wilson AG, Bencardino J, Hochberg M, Jordan JM, Mitchell B, Kraus VB, Abramson SB. Interleukin 1 receptor antagonist ( IL1RN) gene variants predict radiographic severity of knee osteoarthritis and risk of incident disease. Ann Rheum Dis 2020; 79:400-407. [PMID: 31852669 PMCID: PMC7034355 DOI: 10.1136/annrheumdis-2019-216055] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 11/12/2019] [Accepted: 12/01/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE In these studies, we examined the association of single nucleotide polymorphisms (SNPs) of the IL1RN gene with radiographic severity of symptomatic knee osteoarthritis (SKOA) and the risk of incident OA. We also explored these genetic polymorphisms in patients with new onset rheumatoid arthritis (RA). METHODS Over 1000 subjects who met American College of Rheumatology criteria for tibiofemoral OA were selected from three independent, National Institute of Health (NIH)-funded cohorts. CTA and TTG haplotypes formed from three SNPs of the IL1RN gene (rs419598, rs315952, rs9005) were assessed for association with radiographic severity, and risk for incident radiographic OA (rOA) in a nested case-control cohort. These IL1RN haplotypes were also assessed for association with disease activity (DAS28) and plasma inflammatory markers in patients with RA. RESULTS Carriage of the IL1RN TTG haplotype was associated with increased odds of more severe rOA compared with age-matched, sex-matched and body mass index-matched individuals. Examination of the osteoarthritis initiative Incidence Subcohort demonstrated that carriage of the TTG haplotype was associated with 4.1-fold (p=0.001) increased odds of incident rOA. Plasma IL-1Ra levels were lower in TTG carriers, while chondrocytes from TTG carriers exhibited decreased secretion of IL-1Ra. In patients with RA, the TTG haplotype was associated with increased DAS28, decreased plasma IL-1Ra and elevations of plasma inflammatory markers (hsCRP, interleukin 6 (IL-6)). CONCLUSION Carriage of the IL1RN TTG haplotype is associated with more severe rOA, increased risk for incident OA, and increased evidence of inflammation in RA. These data suggest that the IL1RN TTG risk haplotype, associated with decreased IL-1Ra plasma levels, impairs endogenous 'anti-inflammatory' mechanisms.
Collapse
Affiliation(s)
- Mukundan Attur
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, New York, USA
| | - Hua Zhou
- Applied Bioinformatics Laboratories, New York University School of Medicine, New York, New York, USA
| | - Johathan Samuels
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, New York, USA
| | - Svetlana Krasnokutsky
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, New York, USA
| | - Michelle Yau
- Department of Epidemiology and Public Health, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Jose U Scher
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, New York, USA
| | - Michael Doherty
- Academic Rheumatology, University of Nottingham, Nottingham, UK
| | - Anthony G Wilson
- University College Dublin, UCD School of Medicine and Medical Science, Conway Institute, Dublin, Ireland
| | - Jenny Bencardino
- Radiology, New York University School of Medicine, New York, New York, USA
| | - Marc Hochberg
- Medicine/Epidemiology and Public Health, University of North Carolina, Chaple Hill, North Carolina, USA
| | - Joanne M Jordan
- Departments of Medicine and Epidemiology, Thurston Arthritis Research Center, Chapel Hill, North Carolina, USA
| | - Braxton Mitchell
- Medicine/Epidemiology and Public Health, University of North Carolina, Chaple Hill, North Carolina, USA
- Geriatrics Research and Education Clinical Center, Baltimore VA Medical Center, Baltimore, Maryland, USA
| | - Virginia B Kraus
- Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Steven B Abramson
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
45
|
Gao F, Yao Y, Zhang Y, Tian J. Integrating Genome-Wide Association Studies With Pathway Analysis and Gene Expression Analysis Highlights Novel Osteoarthritis Risk Pathways and Genes. Front Genet 2019; 10:827. [PMID: 31572443 PMCID: PMC6753977 DOI: 10.3389/fgene.2019.00827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/12/2019] [Indexed: 01/17/2023] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disorder worldwide. To identify more genetic signals, genome-wide association study (GWAS) has been widely used and elucidated some OA susceptibility genes. However, these susceptibility genes could only explain only a small part of heritability of OA. It is suggested that the identification of disease-related pathways may contribute to understand the genomic etiology of OA. Here, we integrated the GWAS into pathway analysis to identify novel OA risk pathways. In this study, we first selected 187 independent genetic variants identified by GWAS (P < 1.00E−05) and found that most of these genetic variants are noncoding mutations. We then conducted an expression quantitative trait loci analysis and found that 165 of these 187 genetic variants could significantly regulate the expression of nearby genes. Third, we identified OA susceptibility genes corresponding to these genetic variants, conducted a pathway analysis, and identified novel OA-related KEGG pathways, GO biological processes, GO molecular functions, and GO cellular components. In KEGG database, transforming growth factor β signaling pathway is the most significant signal (P = 5.98E−05) and is the only pathway after the BH multiple-test adjustment with false discovery rate (FDR) = 0.02. In GO database, we identified 24 statistically significant GO biological processes, one statistically significant GO molecular function, and five statistically significant GO cellular components (FDR < 0.05). These signals are related with chondrocyte differentiation and development, which are all known biological pathways associated with OA. Finally, we conducted an OA case–control gene expression analysis to evaluate the differential expression of these OA risk genes. Using an OA case–control gene expression analysis, we showed that 44 risk genes were suggestively differentially expressed in OA cases compared with controls (P < 0.05). Three genes, WWP2, COG5, and MAPT, were statistically differentially expressed in OA cases compared with controls (P < 0.05/122 = 4.10E−04). Hence, our findings may contribute to understanding the genomic etiology of OA.
Collapse
Affiliation(s)
- Feng Gao
- Department of Trauma and Emergency Surgeon, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yu Yao
- Department of Trauma and Emergency Surgeon, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yiwei Zhang
- Department of Trauma and Emergency Surgeon, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jun Tian
- Department of Trauma and Emergency Surgeon, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
46
|
Hand osteoarthritis: clinical phenotypes, molecular mechanisms and disease management. Nat Rev Rheumatol 2019; 14:641-656. [PMID: 30305701 DOI: 10.1038/s41584-018-0095-4] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Osteoarthritis (OA) is a highly prevalent condition, and the hand is the most commonly affected site. Patients with hand OA frequently report symptoms of pain, functional limitations and frustration in undertaking everyday activities. The condition presents clinically with changes to the bone, ligaments, cartilage and synovial tissue, which can be observed using radiography, ultrasonography or MRI. Hand OA is a heterogeneous disorder and is considered to be multifactorial in aetiology. This Review provides an overview of the epidemiology, presentation and burden of hand OA, including an update on hand OA imaging (including the development of novel techniques), disease mechanisms and management. In particular, areas for which new evidence has substantially changed the way we understand, consider and treat hand OA are highlighted. For example, genetic studies, clinical trials and careful prospective imaging studies from the past 5 years are beginning to provide insights into the pathogenesis of hand OA that might uncover new therapeutic targets in the disease.
Collapse
|
47
|
Choi YR, Collins KH, Lee JW, Kang HJ, Guilak F. Genome Engineering for Osteoarthritis: From Designer Cells to Disease-Modifying Drugs. Tissue Eng Regen Med 2019; 16:335-343. [PMID: 31413938 PMCID: PMC6675820 DOI: 10.1007/s13770-018-0172-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/27/2018] [Accepted: 12/01/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a highly prevalent degenerative joint disease involving joint cartilage and its surrounding tissues. OA is the leading cause of pain and disability worldwide. At present, there are no disease-modifying OA drugs, and the primary therapies include exercise and nonsteroidal anti-inflammatory drugs until total joint replacement at the end-stage of the disease. METHODS In this review, we summarized the current state of knowledge in genetic and epigenetic associations and risk factors for OA and their potential diagnostic and therapeutic applications. RESULTS Genome-wide association studies and analysis of epigenetic modifications (such as miRNA expression, DNA methylation and histone modifications) conducted across various populations support the notion that there is a genetic basis for certain subsets of OA pathogenesis. CONCLUSION With recent advances in the development of genome editing technologies such as the CRISPR-Cas9 system, these genetic and epigenetic alternations in OA can be used as platforms from which potential biomarkers for the diagnosis, prognosis, drug response, and development of potential personalized therapeutic targets for OA can be approached. Furthermore, genome editing has allowed the development of "designer" cells, whereby the receptors, gene regulatory networks, or transgenes can be modified as a basis for new cell-based therapies.
Collapse
Affiliation(s)
- Yun-Rak Choi
- Department of Orthopaedic Surgery, Washington University in St. Louis, 1 Brookings Dr, St. Louis, MO 63130 USA
- Shriners Hospitals for Children – St. Louis, 4400 Clayton Ave, St. Louis, MO 63110 USA
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 Republic of Korea
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University in St. Louis, 1 Brookings Dr, St. Louis, MO 63130 USA
- Shriners Hospitals for Children – St. Louis, 4400 Clayton Ave, St. Louis, MO 63110 USA
| | - Jin-Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 Republic of Korea
| | - Ho-Jung Kang
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 Republic of Korea
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, 1 Brookings Dr, St. Louis, MO 63130 USA
- Shriners Hospitals for Children – St. Louis, 4400 Clayton Ave, St. Louis, MO 63110 USA
- Center of Regenerative Medicine, Campus Box 8233, McKinley Research Bldg, Room 3121, St. Louis, MO 63110 USA
| |
Collapse
|
48
|
Rice SJ, Tselepi M, Sorial AK, Aubourg G, Shepherd C, Almarza D, Skelton AJ, Pangou I, Deehan D, Reynard LN, Loughlin J. Prioritization of PLEC and GRINA as Osteoarthritis Risk Genes Through the Identification and Characterization of Novel Methylation Quantitative Trait Loci. Arthritis Rheumatol 2019; 71:1285-1296. [PMID: 30730609 PMCID: PMC6790675 DOI: 10.1002/art.40849] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/30/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To identify methylation quantitative trait loci (mQTLs) correlating with osteoarthritis (OA) risk alleles and to undertake mechanistic characterization as a means of target gene prioritization. METHODS We used genome-wide genotyping and cartilage DNA methylation array data in a discovery screen of novel OA risk loci. This was followed by methylation, gene expression analysis, and genotyping studies in additional cartilage samples, accompanied by in silico analyses. RESULTS We identified 4 novel OA mQTLs. The most significant mQTL contained 9 CpG sites where methylation correlated with OA risk genotype, with 5 of the CpG sites having P values <1 × 10-10 . The 9 CpG sites reside in an interval of only 7.7 kb within the PLEC gene and form 2 distinct clusters. We were able to prioritize PLEC and the adjacent gene GRINA as independent targets of the OA risk. We identified PLEC and GRINA expression QTLs operating in cartilage, as well as methylation-expression QTLs operating on the 2 genes. GRINA and PLEC also demonstrated differential expression between OA hip and non-OA hip cartilage. CONCLUSION PLEC encodes plectin, a cytoskeletal protein that maintains tissue integrity by regulating intracellular signaling in response to mechanical stimuli. GRINA encodes the ionotropic glutamate receptor TMBIM3 (transmembrane BAX inhibitor 1 motif-containing protein family member 3), which regulates cell survival. Based on our results, we hypothesize that in a joint predisposed to OA, expression of these genes alters in order to combat aberrant biomechanics, and that this is epigenetically regulated. However, carriage of the OA risk-conferring allele at this locus hinders this response and contributes to disease development.
Collapse
Affiliation(s)
- Sarah J Rice
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - Maria Tselepi
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - Antony K Sorial
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - Guillaume Aubourg
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - Colin Shepherd
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - David Almarza
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew J Skelton
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - Ioanna Pangou
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | | | - Louise N Reynard
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| | - John Loughlin
- International Centre for Life, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
49
|
Abstract
The concept of interleukin-1 (IL-1) as a target in osteoarthritis (OA) has been an attractive one for many years. It is a highly potent inducer of cartilage degradation, causing the induction of mRNA and controlling the bioavailability of disease-relevant proteases such as ADAMTS5 and MMP13. It drives synovitis and can induce other disease-relevant genes such as nerve growth factor, a key pain sensitiser in OA. However, the quality of evidence for its involvement in disease is modest. Descriptive studies have demonstrated expression of IL-1α and β in OA cartilage and elevated levels in the synovial fluid of some patients. Agnostic transcriptomic and genomic analyses do not identify IL-1 as a key pathway.
In vivo models show a conflicting role for this molecule; early studies using therapeutic approaches in large animal models show a benefit, but most murine studies fail to demonstrate protection where the ligands (IL-1α/β), the cytokine activator (IL-1–converting enzyme), or the receptor (IL-1R) have been knocked out. Recently, a number of large double-blind randomised controlled clinical studies targeting IL-1 have failed. Enthusiasm for IL-1 as a target in OA is rapidly dwindling.
Collapse
Affiliation(s)
- Tonia L Vincent
- Centre for OA Pathogenesis Versus Arthritis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY, UK
| |
Collapse
|
50
|
Klein JC, Keith A, Rice SJ, Shepherd C, Agarwal V, Loughlin J, Shendure J. Functional testing of thousands of osteoarthritis-associated variants for regulatory activity. Nat Commun 2019; 10:2434. [PMID: 31164647 PMCID: PMC6547687 DOI: 10.1038/s41467-019-10439-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 04/29/2019] [Indexed: 12/19/2022] Open
Abstract
To date, genome-wide association studies have implicated at least 35 loci in osteoarthritis but, due to linkage disequilibrium, the specific variants underlying these associations and the mechanisms by which they contribute to disease risk have yet to be pinpointed. Here, we functionally test 1,605 single nucleotide variants associated with osteoarthritis for regulatory activity using a massively parallel reporter assay. We identify six single nucleotide polymorphisms (SNPs) with differential regulatory activity between the major and minor alleles. We show that the most significant SNP, rs4730222, exhibits differential nuclear protein binding in electrophoretic mobility shift assays and drives increased expression of an alternative isoform of HBP1 in a heterozygote chondrosarcoma cell line, in a CRISPR-edited osteosarcoma cell line, and in chondrocytes derived from osteoarthritis patients. This study provides a framework for prioritization of GWAS variants and highlights a role of HBP1 and Wnt signaling in osteoarthritis pathogenesis.
Collapse
Affiliation(s)
- Jason C Klein
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Aidan Keith
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Sarah J Rice
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle-upon-Tyne, NE1 3BZ, UK
| | - Colin Shepherd
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle-upon-Tyne, NE1 3BZ, UK
| | - Vikram Agarwal
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - John Loughlin
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle-upon-Tyne, NE1 3BZ, UK
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, 98195, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|