دورية أكاديمية

Accelerating functional gene discovery in osteoarthritis.

التفاصيل البيبلوغرافية
العنوان: Accelerating functional gene discovery in osteoarthritis.
المؤلفون: Butterfield NC; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Curry KF; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Steinberg J; Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764, Neuherberg, Germany.; Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK.; Cancer Council NSW, Sydney, NSW, 2000, Australia., Dewhurst H; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Komla-Ebri D; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Mannan NS; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Adoum AT; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Leitch VD; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Logan JG; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Waung JA; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Ghirardello E; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK., Southam L; Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764, Neuherberg, Germany.; Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK., Youlten SE; The Garvan Institute of Medical Research and St. Vincent's Clinical School, University of New South Wales Medicine, Sydney, NSW, 2010, Australia., Wilkinson JM; Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2RX, UK.; Centre for Integrated Research into Musculoskeletal Ageing and Sheffield Healthy Lifespan Institute, University of Sheffield, Sheffield, S10 2TN, UK., McAninch EA; Division of Endocrinology and Metabolism, Rush University Medical Center, Chicago, IL, 60612, USA., Vancollie VE; Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK., Kussy F; Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK., White JK; Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK.; The Jackson Laboratory, Bar Harbor, ME, 04609, USA., Lelliott CJ; Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK., Adams DJ; Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK., Jacques R; School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, S1 4DA, UK., Bianco AC; Section of Adult and Pediatric Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA., Boyde A; Dental Physical Sciences, Queen Mary University of London, Mile End Road, London, E1 4NS, UK., Zeggini E; Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, 85764, Neuherberg, Germany.; Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK., Croucher PI; The Garvan Institute of Medical Research and St. Vincent's Clinical School, University of New South Wales Medicine, Sydney, NSW, 2010, Australia., Williams GR; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK. graham.williams@imperial.ac.uk., Bassett JHD; Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, W12 0NN, UK. d.bassett@imperial.ac.uk.
المصدر: Nature communications [Nat Commun] 2021 Jan 20; Vol. 12 (1), pp. 467. Date of Electronic Publication: 2021 Jan 20.
نوع المنشور: Journal Article; Research Support, Non-U.S. Gov't
اللغة: English
بيانات الدورية: Publisher: Nature Pub. Group Country of Publication: England NLM ID: 101528555 Publication Model: Electronic Cited Medium: Internet ISSN: 2041-1723 (Electronic) Linking ISSN: 20411723 NLM ISO Abbreviation: Nat Commun Subsets: MEDLINE
أسماء مطبوعة: Original Publication: [London] : Nature Pub. Group
مواضيع طبية MeSH: Genetic Association Studies*, Genetic Predisposition to Disease/*genetics , Osteoarthritis/*genetics, Animals ; Bone and Bones/pathology ; CRISPR-Cas Systems ; Cartilage/pathology ; Clustered Regularly Interspaced Short Palindromic Repeats ; Disease Models, Animal ; Drug Discovery ; Gene Editing ; Gonadotropin-Releasing Hormone/genetics ; Iodide Peroxidase ; Mice ; Mice, Knockout ; Osteoarthritis/pathology ; Osteoarthritis/surgery ; Paired Box Transcription Factors/genetics ; Phenotype ; Iodothyronine Deiodinase Type II
مستخلص: Osteoarthritis causes debilitating pain and disability, resulting in a considerable socioeconomic burden, yet no drugs are available that prevent disease onset or progression. Here, we develop, validate and use rapid-throughput imaging techniques to identify abnormal joint phenotypes in randomly selected mutant mice generated by the International Knockout Mouse Consortium. We identify 14 genes with functional involvement in osteoarthritis pathogenesis, including the homeobox gene Pitx1, and functionally characterize 6 candidate human osteoarthritis genes in mouse models. We demonstrate sensitivity of the methods by identifying age-related degenerative joint damage in wild-type mice. Finally, we phenotype previously generated mutant mice with an osteoarthritis-associated polymorphism in the Dio2 gene by CRISPR/Cas9 genome editing and demonstrate a protective role in disease onset with public health implications. We hope this expanding resource of mutant mice will accelerate functional gene discovery in osteoarthritis and offer drug discovery opportunities for this common, incapacitating chronic disease.
التعليقات: Erratum in: Nat Commun. 2021 May 28;12(1):3302. (PMID: 34050183)
References: Styrkarsdottir, U. et al. Meta-analysis of Icelandic and UK data sets identifies missense variants in SMO, IL11, COL11A1 and 13 more new loci associated with osteoarthritis. Nat. Genet. 50, 1681–1687 (2018). (PMID: 3037406910.1038/s41588-018-0247-0)
Tachmazidou, I. et al. Identification of new therapeutic targets for osteoarthritis through genome-wide analyses of UK Biobank data. Nat. Genet. 51, 230–236 (2019). (PMID: 30664745640026710.1038/s41588-018-0327-1)
Zengini, E. et al. Genome-wide analyses using UK Biobank data provide insights into the genetic architecture of osteoarthritis. Nat. Genet. 50, 549–558 (2018). (PMID: 29559693589673410.1038/s41588-018-0079-y)
Lories, R. J. & Luyten, F. P. The bone-cartilage unit in osteoarthritis. Nat. Rev. Rheumatol. 7, 43–49 (2011). (PMID: 2113588110.1038/nrrheum.2010.197)
Choi, W. S. et al. The CH25H-CYP7B1-RORalpha axis of cholesterol metabolism regulates osteoarthritis. Nature 566, 254–258 (2019). (PMID: 3072850010.1038/s41586-019-0920-1)
Zhang, M. et al. Induced superficial chondrocyte death reduces catabolic cartilage damage in murine posttraumatic osteoarthritis. J. Clin. Invest 126, 2893–2902 (2016). (PMID: 27427985496631610.1172/JCI83676)
Dreier, R. Hypertrophic differentiation of chondrocytes in osteoarthritis: the developmental aspect of degenerative joint disorders. Arthritis Res. Ther. 12, 216 (2010). (PMID: 20959023299099110.1186/ar3117)
Loeser, R. F., Goldring, S. R., Scanzello, C. R. & Goldring, M. B. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum. 64, 1697–1707 (2012). (PMID: 22392533336601810.1002/art.34453)
Burr, D. B. & Gallant, M. A. Bone remodelling in osteoarthritis. Nat. Rev. Rheumatol. 8, 665–673 (2012). (PMID: 2286892510.1038/nrrheum.2012.130)
Kuhn, K., D’Lima, D. D., Hashimoto, S. & Lotz, M. Cell death in cartilage. Osteoarthr. Cartil. 12, 1–16 (2004). (PMID: 10.1016/j.joca.2003.09.015)
Enomoto, H. et al. Vascular endothelial growth factor isoforms and their receptors are expressed in human osteoarthritic cartilage. Am. J. Pathol. 162, 171–181 (2003). (PMID: 12507900185111410.1016/S0002-9440(10)63808-4)
Billinghurst, R. C. et al. Enhanced cleavage of type II collagen by collagenases in osteoarthritic articular cartilage. J. Clin. Invest. 99, 1534–1545 (1997). (PMID: 911999750797310.1172/JCI119316)
Glasson, S. S. et al. Deletion of active ADAMTS5 prevents cartilage degradation in a murine model of osteoarthritis. Nature 434, 644–648 (2005). (PMID: 1580062410.1038/nature03369)
Boyden, L. M. et al. High bone density due to a mutation in LDL-receptor-related protein 5. N. Engl. J. Med. 346, 1513–1521 (2002). (PMID: 1201539010.1056/NEJMoa013444)
Farooqi, I. S. et al. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N. Engl. J. Med. 341, 879–884 (1999). (PMID: 1048641910.1056/NEJM199909163411204)
Bassett, J. H. et al. Rapid-throughput skeletal phenotyping of 100 knockout mice identifies 9 new genes that determine bone strength. PLoS Genet. 8, e1002858 (2012). (PMID: 22876197341085910.1371/journal.pgen.1002858)
Kemp, J. P. et al. Identification of 153 new loci associated with heel bone mineral density and functional involvement of GPC6 in osteoporosis. Nat. Genet. 49, 1468–1475 (2017). (PMID: 28869591562162910.1038/ng.3949)
Medina-Gomez, C. et al. Life-course genome-wide association study meta-analysis of total body bmd and assessment of age-specific effects. Am. J. Hum. Genet. 102, 88–102 (2018). (PMID: 29304378577798010.1016/j.ajhg.2017.12.005)
Morris, J. A. et al. An atlas of genetic influences on osteoporosis in humans and mice. Nat. Genet. 51, 258–266 (2019). (PMID: 3059854910.1038/s41588-018-0302-x)
Trajanoska, K. et al. Assessment of the genetic and clinical determinants of fracture risk: genome wide association and mendelian randomisation study. BMJ 362, k3225 (2018). (PMID: 30158200611377310.1136/bmj.k3225)
Butterfield, N. C., Logan, J. G., Waung, J., Williams, G. R. & Bassett, J. H. D. Quantitative X-ray imaging of mouse bone by faxitron. Methods Mol. Biol. 1914, 559–569 (2019). (PMID: 3072948610.1007/978-1-4939-8997-3_30)
Waung, J. A. et al. Quantitative X-ray microradiography for high-throughput phenotyping of osteoarthritis in mice. Osteoarthritis Cartilage 22, 1396–400 (2014).
Glasson, S. S., Chambers, M. G., Van Den Berg, W. B. & Little, C. B. The OARSI histopathology initiative - recommendations for histological assessments of osteoarthritis in the mouse. Osteoarthr. Cartil. 18, S17–S23 (2010). (PMID: 10.1016/j.joca.2010.05.025)
Glasson, S. S., Blanchet, T. J. & Morris, E. A. The surgical destabilization of the medial meniscus (DMM) model of osteoarthritis in the 129/SvEv mouse. Osteoarthr. Cartil. 15, 1061–1069 (2007). (PMID: 10.1016/j.joca.2007.03.006)
Perez-Garcia, V. et al. Placentation defects are highly prevalent in embryonic lethal mouse mutants. Nature 555, 463–468 (2018). (PMID: 29539633586671910.1038/nature26002)
Wu, C., Jin, X., Tsueng, G., Afrasiabi, C. & Su, A. I. BioGPS: building your own mash-up of gene annotations and expression profiles. Nucleic Acids Res. 44, D313–D316 (2016). (PMID: 2657858710.1093/nar/gkv1104)
Kean, T. J., Ge, Z., Li, Y., Chen, R. & Dennis, J. E. Transcriptome-wide analysis of human chondrocyte expansion on synoviocyte matrix. Cells 8, 85 (2019). (PMID: 640636210.3390/cells8020085)
Soul, J., Hardingham, T., Boot-Handford, R. & Schwartz, J. M. SkeletalVis: an exploration and meta-analysis data portal of cross-species skeletal transcriptomics data. Bioinformatics 35, 2283–2290 (2019). (PMID: 3048125710.1093/bioinformatics/bty947)
Szeto, D. P. et al. Role of the Bicoid-related homeodomain factor Pitx1 in specifying hindlimb morphogenesis and pituitary development. Genes Dev. 13, 484–494 (1999). (PMID: 1004936331647110.1101/gad.13.4.484)
Lanctot, C., Moreau, A., Chamberland, M., Tremblay, M. L. & Drouin, J. Hindlimb patterning and mandible development require the Ptx1 gene. Development 126, 1805–1810 (1999). (PMID: 1010111510.1242/dev.126.9.1805)
Spielmann, M. et al. Homeotic arm-to-leg transformation associated with genomic rearrangements at the PITX1 locus. Am. J. Hum. Genet. 91, 629–635 (2012). (PMID: 23022097348464710.1016/j.ajhg.2012.08.014)
Gurnett, C. A. et al. Asymmetric lower-limb malformations in individuals with homeobox PITX1 gene mutation. Am. J. Hum. Genet. 83, 616–622 (2008). (PMID: 18950742266804410.1016/j.ajhg.2008.10.004)
Boudjelal, M. et al. Overexpression of Stra13, a novel retinoic acid-inducible gene of the basic helix-loop-helix family, inhibits mesodermal and promotes neuronal differentiation of P19 cells. Genes Dev. 11, 2052–2065 (1997). (PMID: 928404531645410.1101/gad.11.16.2052)
MacLean, H. E. & Kronenberg, H. M. Expression of Stra13 during mouse endochondral bone development. Gene Expr. Patterns 4, 633–636 (2004). (PMID: 1546548510.1016/j.modgep.2004.04.011)
Miyazaki, K. et al. Identification of functional hypoxia response elements in the promoter region of the DEC1 and DEC2 genes. J. Biol. Chem. 277, 47014–47021 (2002). (PMID: 1235477110.1074/jbc.M204938200)
Shen, M. et al. Basic helix-loop-helix protein DEC1 promotes chondrocyte differentiation at the early and terminal stages. J. Biol. Chem. 277, 50112–50120 (2002). (PMID: 1238450510.1074/jbc.M206771200)
Iwata, T. et al. Effects of overexpression of basic helix-loop-helix transcription factor Dec1 on osteogenic and adipogenic differentiation of mesenchymal stem cells. Eur. J. Cell Biol. 85, 423–431 (2006). (PMID: 1648762610.1016/j.ejcb.2005.12.007)
Hu, S. et al. Differentiated embryonic chondrocytes 1 expression of periodontal ligament tissue and gingival tissue in the patients with chronic periodontitis. Arch. Oral. Biol. 60, 517–525 (2015). (PMID: 2557529610.1016/j.archoralbio.2014.12.006)
Wu, L. et al. Insights on biology and pathology of HIF-1alpha/-2alpha, TGFbeta/BMP, Wnt/beta-catenin, and NF-kappaB pathways in osteoarthritis. Curr. Pharm. Des. 18, 3293–3312 (2012). (PMID: 2264609210.2174/1381612811209023293)
Saito, T. & Kawaguchi, H. HIF-2alpha as a possible therapeutic target of osteoarthritis. Osteoarthr. Cartil. 18, 1552–1556 (2010). (PMID: 10.1016/j.joca.2010.10.006)
Gamer, L. W. et al. The role of Bmp2 in the maturation and maintenance of the murine knee joint. J. Bone Min. Res. 33, 1708–1717 (2018). (PMID: 10.1002/jbmr.3441)
Dai, M. W. et al. Parathyroid hormone(1-34) exhibits more comprehensive effects than celecoxib in cartilage metabolism and maintaining subchondral bone micro-architecture in meniscectomized guinea pigs. Osteoarthr. Cartil. 24, 1103–1112 (2016). (PMID: 10.1016/j.joca.2016.01.007)
Tosoni, D. et al. TTP specifically regulates the internalization of the transferrin receptor. Cell 123, 875–888 (2005). (PMID: 1632558110.1016/j.cell.2005.10.021)
Francavilla, C. et al. Functional proteomics defines the molecular switch underlying FGF receptor trafficking and cellular outputs. Mol. Cell 51, 707–722 (2013). (PMID: 2401159010.1016/j.molcel.2013.08.002)
Kim, Y. M. et al. SH3BP4 is a negative regulator of amino acid-Rag GTPase-mTORC1 signaling. Mol. Cell 46, 833–846 (2012). (PMID: 22575674338927610.1016/j.molcel.2012.04.007)
Antas, P. et al. SH3BP4 regulates intestinal stem cells and tumorigenesis by modulating beta-catenin nuclear localization. Cell Rep. 26, 2266–2273 (2019). e2264. (PMID: 30811977639171110.1016/j.celrep.2019.01.110)
Rauner, M. et al. Transferrin receptor 2 controls bone mass and pathological bone formation via BMP and Wnt signaling. Nat. Metab. 1, 111–124 (2019). (PMID: 30886999642007410.1038/s42255-018-0005-8)
Tang, J. et al. Fibroblast growth factor receptor 3 inhibits osteoarthritis progression in the knee joints of adult mice. Arthritis Rheum. 68, 2432–2443 (2016). (PMID: 10.1002/art.39739)
Zhang, Y. et al. Cartilage-specific deletion of mTOR upregulates autophagy and protects mice from osteoarthritis. Ann. Rheum. Dis. 74, 1432–1440 (2015). (PMID: 2465162110.1136/annrheumdis-2013-204599)
Monteagudo, S. & Lories, R. J. Cushioning the cartilage: a canonical Wnt restricting matter. Nat. Rev. Rheum. 13, 670–681 (2017). (PMID: 10.1038/nrrheum.2017.171)
Steinberg, J. et al. Decoding the genomic basis of osteoarthritis. bioRxiv, https://doi.org/10.1101/835850 (2019).
Murn, J. et al. Control of a neuronal morphology program by an RNA-binding zinc finger protein, Unkempt. Genes Dev. 29, 501–512 (2015). (PMID: 25737280435840310.1101/gad.258483.115)
Seki, T. et al. JosD1, a membrane-targeted deubiquitinating enzyme, is activated by ubiquitination and regulates membrane dynamics, cell motility, and endocytosis. J. Biol. Chem. 288, 17145–17155 (2013). (PMID: 23625928368252010.1074/jbc.M113.463406)
Wang, X. et al. JOSD1 negatively regulates type-I interferon antiviral activity by deubiquitinating and stabilizing SOCS1. Viral Immunol. 30, 342–349 (2017). (PMID: 2835510510.1089/vim.2017.0015)
Kapoor, M., Martel-Pelletier, J., Lajeunesse, D., Pelletier, J. P. & Fahmi, H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat. Rev. Rheum. 7, 33–42 (2011). (PMID: 10.1038/nrrheum.2010.196)
Rogers, C. et al. Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation. Nat. Commun. 10, 1689 (2019). (PMID: 30976076645983610.1038/s41467-019-09397-2)
Zhang, L. et al. Increased HIF-1alpha in knee osteoarthritis aggravate synovial fibrosis via fibroblast-like synoviocyte pyroptosis. Oxid. Med. Cell Longev. 2019, 6326517 (2019). (PMID: 307557876348923)
Zhao, L. R. et al. NLRP1 and NLRP3 inflammasomes mediate LPS/ATPinduced pyroptosis in knee osteoarthritis. Mol. Med. Rep. 17, 5463–5469 (2018). (PMID: 29393464)
Mao, X. & Tong, J. ARHGAP30 suppressed lung cancer cell proliferation, migration, and invasion through inhibition of the Wnt/beta-catenin signaling pathway. Onco Targets Ther. 11, 7447–7457 (2018). (PMID: 30425532620487610.2147/OTT.S175255)
Thanasopoulou, A., Stravopodis, D. J., Dimas, K. S., Schwaller, J. & Anastasiadou, E. Loss of CCDC6 affects cell cycle through impaired intra-S-phase checkpoint control. PLoS ONE 7, e31007 (2012). (PMID: 22363533328190010.1371/journal.pone.0031007)
Fei, Q. et al. Identification of upstream regulators for synovial expression signature genes in osteoarthritis. Jt. Bone Spine 83, 545–551 (2016). (PMID: 10.1016/j.jbspin.2015.09.001)
Khoshnoodi, J., Pedchenko, V. & Hudson, B. G. Mammalian collagen IV. Microsc. Res. Tech. 71, 357–370 (2008). (PMID: 18219669478809610.1002/jemt.20564)
Yang, X., Zhao, J., He, Y. & Huangfu, X. Screening for characteristic genes in osteoarthritis induced by destabilization of the medial meniscus utilizing bioinformatics approach. J. Musculoskelet. Neuronal Interact. 14, 343–348 (2014). (PMID: 25198230)
Olex, A. L., Turkett, W. H., Fetrow, J. S. & Loeser, R. F. Integration of gene expression data with network-based analysis to identify signaling and metabolic pathways regulated during the development of osteoarthritis. Gene 542, 38–45 (2014). (PMID: 24630964403174610.1016/j.gene.2014.03.022)
Karlsson, C. et al. Genome-wide expression profiling reveals new candidate genes associated with osteoarthritis. Osteoarthr. Cartil. 18, 581–592 (2010). (PMID: 10.1016/j.joca.2009.12.002)
Rushton, M. D. et al. Characterization of the cartilage DNA methylome in knee and hip osteoarthritis. Arthritis Rheum. 66, 2450–2460 (2014). (PMID: 10.1002/art.38713)
Peeters, R. P. et al. Polymorphisms in thyroid hormone pathway genes are associated with plasma TSH and iodothyronine levels in healthy subjects. J. Clin. Endocrinol. Metab. 88, 2880–2888 (2003). (PMID: 1278890210.1210/jc.2002-021592)
Meulenbelt, I. et al. Identification of DIO2 as a new susceptibility locus for symptomatic osteoarthritis. Hum. Mol. Genet. 17, 1867–1875 (2008). (PMID: 1833457810.1093/hmg/ddn082)
Kerkhof, H. J. et al. A genome-wide association study identifies an osteoarthritis susceptibility locus on chromosome 7q22. Arthritis Rheum. 62, 499–510 (2010). (PMID: 201123603354739)
Evangelou, E. et al. A meta-analysis of genome-wide association studies identifies novel variants associated with osteoarthritis of the hip. Ann. Rheum. Dis. 73, 2130–2136 (2014). (PMID: 2398998610.1136/annrheumdis-2012-203114)
Waarsing, J. H. et al. Osteoarthritis susceptibility genes influence the association between hip morphology and osteoarthritis. Arthritis Rheum. 63, 1349–1354 (2011). (PMID: 2140047310.1002/art.30288)
Robson, H., Siebler, T., Stevens, D. A., Shalet, S. M. & Williams, G. R. Thyroid hormone acts directly on growth plate chondrocytes to promote hypertrophic differentiation and inhibit clonal expansion and cell proliferation. Endocrinology 141, 3887–3897 (2000). (PMID: 1101424610.1210/endo.141.10.7733)
Makihira, S. et al. Thyroid hormone enhances aggrecanase-2/ADAM-TS5 expression and proteoglycan degradation in growth plate cartilage. Endocrinology 144, 2480–2488 (2003). (PMID: 1274631010.1210/en.2002-220746)
Jo, S. et al. Type 2 deiodinase polymorphism causes ER stress and hypothyroidism in the brain. J. Clin. Invest 129, 230–245 (2019). (PMID: 3035204610.1172/JCI123176)
Hernandez, A. Cognitive function in hypothyroidism: what is that deiodinase again? J. Clin. Invest 129, 55–57 (2019). (PMID: 3050761110.1172/JCI125203)
Gereben, B., McAninch, E. A., Ribeiro, M. O. & Bianco, A. C. Scope and limitations of iodothyronine deiodinases in hypothyroidism. Nat. Rev. Endocrinol. 11, 642–652 (2015). (PMID: 26416219500378110.1038/nrendo.2015.155)
Waung, J. A., Bassett, J. H. & Williams, G. R. Adult mice lacking the type 2 iodothyronine deiodinase have increased subchondral bone but normal articular cartilage. Thyroid 25, 269–277 (2015). (PMID: 25549200436141010.1089/thy.2014.0476)
Bomer, N. et al. Aberrant calreticulin expression in articular cartilage of Dio2 deficient mice. PLoS ONE 11, e0154999 (2016). (PMID: 27163789486266710.1371/journal.pone.0154999)
Li, H. et al. Endoplasmic reticulum stress regulates rat mandibular cartilage thinning under compressive mechanical stress. J. Biol. Chem. 288, 18172–18183 (2013). (PMID: 23603905368996010.1074/jbc.M112.407296)
Bomer, N. et al. Underlying molecular mechanisms of DIO2 susceptibility in symptomatic osteoarthritis. Ann. Rheum. Dis. 74, 1571–1579 (2015). (PMID: 2469500910.1136/annrheumdis-2013-204739)
Bos, S. D. et al. Increased type II deiodinase protein in OA-affected cartilage and allelic imbalance of OA risk polymorphism rs225014 at DIO2 in human OA joint tissues. Ann. Rheum. Dis. 71, 1254–1258 (2012). (PMID: 2249278010.1136/annrheumdis-2011-200981)
Nagase, H. et al. Deiodinase 2 upregulation demonstrated in osteoarthritis patients cartilage causes cartilage destruction in tissue-specific transgenic rats. Osteoarthr. Cartil. 21, 514–523 (2013). (PMID: 10.1016/j.joca.2012.12.013)
Stok, K. S. et al. Three-dimensional quantitative morphometric analysis (QMA) for in situ joint and tissue assessment of osteoarthritis in a preclinical rabbit disease model. PLoS ONE 11, e0147564 (2016). (PMID: 26808542472651210.1371/journal.pone.0147564)
Lee, Y. S. et al. Articular cartilage imaging by the use of phase-contrast tomography in a collagen-induced arthritis mouse model. Acad. Radio. 17, 244–250 (2010). (PMID: 10.1016/j.acra.2009.09.015)
Moodie, J. P., Stok, K. S., Muller, R., Vincent, T. L. & Shefelbine, S. J. Multimodal imaging demonstrates concomitant changes in bone and cartilage after destabilisation of the medial meniscus and increased joint laxity. Osteoarthr. Cartil. 19, 163–170 (2011). (PMID: 10.1016/j.joca.2010.11.006)
Kotwal, N., Li, J., Sandy, J., Plaas, A. & Sumner, D. R. Initial application of EPIC-muCT to assess mouse articular cartilage morphology and composition: effects of aging and treadmill running. Osteoarthr. Cartil. 20, 887–895 (2012). (PMID: 10.1016/j.joca.2012.04.012)
Kerckhofs, G., Sainz, J., Wevers, M., Van de Putte, T. & Schrooten, J. Contrast-enhanced nanofocus computed tomography images the cartilage subtissue architecture in three dimensions. Eur. Cells Mater. 25, 179–189 (2013). (PMID: 10.22203/eCM.v025a13)
Ruan, M. Z. et al. Quantitative imaging of murine osteoarthritic cartilage by phase-contrast micro-computed tomography. Arthritis Rheum. 65, 388–396 (2013). (PMID: 23124630377960210.1002/art.37766)
Das Neves Borges, P., Forte, A. E., Vincent, T. L., Dini, D. & Marenzana, M. Rapid, automated imaging of mouse articular cartilage by microCT for early detection of osteoarthritis and finite element modelling of joint mechanics. Osteoarthr. Cartil. 22, 1419–1428 (2014). (PMID: 10.1016/j.joca.2014.07.014)
Lakin, B. A. et al. Contrast-enhanced CT using a cationic contrast agent enables non-destructive assessment of the biochemical and biomechanical properties of mouse tibial plateau cartilage. J. Orthop. Res 34, 1130–1138 (2016). (PMID: 26697956555638610.1002/jor.23141)
Mashiatulla, M. et al. Murine articular cartilage morphology and compositional quantification with high resolution cationic contrast-enhanced muCT. J. Orthop. Res 35, 2740–2748 (2017). (PMID: 28471533567136610.1002/jor.23595)
Staines, K. A., Madi, K., Javaheri, B., Lee, P. D. & Pitsillides, A. A. A computed microtomography method for understanding epiphyseal growth plate. Fusion. Front. Mater. 4, 48 (2018). (PMID: 2941704710.3389/fmats.2017.00048)
de Bournonville, S., Vangrunderbeeck, S. & Kerckhofs, G. Contrast-enhanced MicroCT for virtual 3D anatomical pathology of biological tissues: a literature review. Contrast Media Mol. Imaging 2019, 8617406 (2019). (PMID: 30944550642176410.1155/2019/8617406)
Gabner, S., Bock, P., Fink, D., Glosmann, M., Handschuh, S. The visible skeleton 2.0: phenotyping of cartilage and bone in fixed vertebrate embryos and foetuses based on X-ray microCT. Development 147, https://doi.org/10.1242/dev.187633 (2020).
Picard, C., Azeddine, B., Moldovan, F., Martel-Pelletier, J. & Moreau, A. New emerging role of pitx1 transcription factor in osteoarthritis pathogenesis. Clin. Orthop. Relat. Res. 462, 59–66 (2007). (PMID: 1754902910.1097/BLO.0b013e3180d09d9c)
Wilhelmi, G. & Faust, R. Suitability of the C57 black mouse as an experimental animal for the study of skeletal changes due to ageing, with special reference to osteo-arthrosis and its response to tribenoside. Pharmacology 14, 289–296 (1976). (PMID: 94709310.1159/000136607)
Bianco, A. C. & Kim, B. S. Pathophysiological relevance of deiodinase polymorphism. Curr. Opin. Endocrinol. Diabetes Obes. 25, 341–346 (2018). (PMID: 30063552657102310.1097/MED.0000000000000428)
Rodriguez-Gutierrez, R., Maraka, S., Ospina, N. S., Montori, V. M. & Brito, J. P. Levothyroxine overuse: time for an about face? Lancet Diabetes Endocrinol. 5, 246–248 (2017). (PMID: 2802953610.1016/S2213-8587(16)30276-5)
Ma, H. L. et al. Osteoarthritis severity is sex dependent in a surgical mouse model. Osteoarthr. Cartil. 15, 695–700 (2007). (PMID: 10.1016/j.joca.2006.11.005)
Fang, H. & Beier, F. Mouse models of osteoarthritis: modelling risk factors and assessing outcomes. Nat. Rev. Rheumatol. 10, 413–421 (2014). (PMID: 2466264510.1038/nrrheum.2014.46)
Kilkenny, C., Browne, W. J., Cuthill, I. C., Emerson, M. & Altman, D. G. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 8, e1000412 (2010). (PMID: 20613859289395110.1371/journal.pbio.1000412)
Skarnes, W. C. et al. A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474, 337–342 (2011). (PMID: 21677750357241010.1038/nature10163)
White, J. K. et al. Genome-wide generation and systematic phenotyping of knockout mice reveals new roles for many genes. Cell 154, 452–464 (2013). (PMID: 23870131371720710.1016/j.cell.2013.06.022)
Gauthier, K. et al. Genetic analysis reveals different functions for the products of the thyroid hormone receptor alpha locus. Mol. Cell Biol. 21, 4748–4760 (2001). (PMID: 114161508715710.1128/MCB.21.14.4748-4760.2001)
Bouxsein, M. L. et al. Guidelines for assessment of bone microstructure in rodents using micro-computed tomography. J. Bone Miner. Res. 25, 1468–1486 (2010). (PMID: 2053330910.1002/jbmr.141)
Giavarina, D. Understanding bland altman analysis. Biochem Med (Zagreb) 25, 141–151 (2015). (PMID: 10.11613/BM.2015.015)
Gluer, C. C. et al. Accurate assessment of precision errors: how to measure the reproducibility of bone densitometry techniques. Osteoporos. Int 5, 262–270 (1995). (PMID: 749286510.1007/BF01774016)
Shrout, P. E. & Fleiss, J. L. Intraclass correlations: uses in assessing rater reliability. Psychol. Bull. 86, 420–428 (1979). (PMID: 1883948410.1037/0033-2909.86.2.420)
Schmitz, N., Laverty, S., Kraus, V. B. & Aigner, T. Basic methods in histopathology of joint tissues. Osteoarthr. Cartil. 18, S113–S116 (2010). (PMID: 10.1016/j.joca.2010.05.026)
Huesa, C. et al. Proteinase-activated receptor 2 modulates OA-related pain, cartilage and bone pathology. Ann. Rheum. Dis. 75, 1989–1997 (2016). (PMID: 2669884610.1136/annrheumdis-2015-208268)
Sophocleous, A. & Huesa, C. Osteoarthritis mouse model of destabilization of the medial meniscus. Methods Mol. Biol. 1914, 281–293 (2019). (PMID: 3072947110.1007/978-1-4939-8997-3_15)
Jackson, M. T. et al. Depletion of protease-activated receptor 2 but not protease-activated receptor 1 may confer protection against osteoarthritis in mice through extracartilaginous mechanisms. Arthritis Rheum. 66, 3337–3348 (2014). (PMID: 10.1002/art.38876)
Steinberg, J. et al. Widespread epigenomic, transcriptomic and proteomic differences between hip osteophytic and articular chondrocytes in osteoarthritis. Rheumatology 57, 1481–1489 (2018). (PMID: 29741735605558310.1093/rheumatology/key101)
Mitchell, A. F. S. & Krzanowski, W. J. The Mahalanobis distance and elliptic distributions. Biometrika 72, 464–467 (1985). (PMID: 10.1093/biomet/72.2.464)
Rousseeuw, P. J. & Van Zomeren, B. C. Unmasking multivariate outliers and leverage points. J. Am. Stat. Assoc. 85, 633–639 (1990). (PMID: 10.1080/01621459.1990.10474920)
Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017). (PMID: 28263959560014810.1038/nmeth.4197)
Soneson, C., Love, M. I. & Robinson, M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res 4, 1521 (2015). (PMID: 2692522710.12688/f1000research.7563.1)
Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015). (PMID: 25605792440251010.1093/nar/gkv007)
Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014). (PMID: 24485249405372110.1186/gb-2014-15-2-r29)
Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014). (PMID: 25516281430204910.1186/s13059-014-0550-8)
McCarthy, D. J., Chen, Y. & Smyth, G. K. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012). (PMID: 22287627337888210.1093/nar/gks042)
Steinberg, J. et al. Integrative epigenomics, transcriptomics and proteomics of patient chondrocytes reveal genes and pathways involved in osteoarthritis. Sci. Rep. 7, 8935 (2017). (PMID: 28827734556645410.1038/s41598-017-09335-6)
معلومات مُعتمدة: 110141/Z/15/Z United Kingdom WT_ Wellcome Trust; 110140 United Kingdom WT_ Wellcome Trust; 110141 United Kingdom WT_ Wellcome Trust; 206194 United Kingdom WT_ Wellcome Trust; R01 DK065055 United States DK NIDDK NIH HHS; United Kingdom WT_ Wellcome Trust; 098051 United Kingdom WT_ Wellcome Trust; 101123 United Kingdom WT_ Wellcome Trust
المشرفين على المادة: 0 (Paired Box Transcription Factors)
0 (homeobox protein PITX1)
33515-09-2 (Gonadotropin-Releasing Hormone)
EC 1.11.1.8 (Iodide Peroxidase)
تواريخ الأحداث: Date Created: 20210121 Date Completed: 20210201 Latest Revision: 20240117
رمز التحديث: 20240117
مُعرف محوري في PubMed: PMC7817695
DOI: 10.1038/s41467-020-20761-5
PMID: 33473114
قاعدة البيانات: MEDLINE
الوصف
تدمد:2041-1723
DOI:10.1038/s41467-020-20761-5