Genetically inspired organoids prevent joint degeneration and alleviate chondrocyte senescence via Col11a1-HIF1α-mediated glycolysis-OXPHOS metabolism shift

Ye Sun , Yongqing You , Qiang Wu , Rui Hu , Kerong Dai

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (2) : e1574

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (2) : e1574 DOI: 10.1002/ctm2.1574
RESEARCH ARTICLE

Genetically inspired organoids prevent joint degeneration and alleviate chondrocyte senescence via Col11a1-HIF1α-mediated glycolysis-OXPHOS metabolism shift

Author information +
History +
PDF

Abstract

Introduction: Developmental dysplasia of hip (DDH) is a hip joint disorder leading to subsequent osteoarthritis. Previous studies suggested collagen XI alpha 1 (COL11A1) as a potential gene in hip dysplasia and chondrocyte degeneration. However, no genetic association has reported COL11A1-related cellular therapy as treatment of DDH and joint degeneration.

Methods and Results: We report identified genetic association between COL11A1 locus and DDH with genome-wide association study (GWAS). Further exome sequencing for familial DDH patients was conducted in different populations to identify potential pathogenic Col11A1 variants for familiar DDH. Further studies demonstrated involvement of COL11A1 expression was down-regulated in femoral head cartilage of DDH patients and Col11a1-KO mice with induced DDH. Col11a1-KO mice demonstrated aggravated joint degeneration and severe OA phenotype. To explore the underlying mechanism of Col11a1 in cartilage and DDH development, we generated scRNA-seq profiles for DDH and Col11a1-KO cartilage, demonstrating disrupted chondrocyte homeostasis and cellular senescence caused by Col11a1-HIF1α-mediated glycolysis-OXPHOS shift in chondrocytes. Genetically and biologically inspired, we further fabricated an intra-articular injection therapy to preventing cartilage degeneration by generating a Col11a1-over-expressed (OE) SMSC mini-organoids. Col11a1-OE organoids demonstrated superior chondrogenesis and ameliorated cartilage degeneration in DDH mice via regulating cellular senescence by up-regulated Col11a1/HIF1α-mediated glycolysis in chondrocytes.

Conclusion: We reported association between COL11A1 loci and DDH with GWAS and exome sequencing. Further studies demonstrated involvement of COL11A1 in DDH patients and Col11a1-KO mice. ScRNA-seq for DDH and Col11a1-KO cartilage demonstrated disrupted chondrocyte homeostasis and cellular senescence caused by Col11a1-HIF1α-mediated glycolysis-OXPHOS shift in chondrocytes. Genetically and biologically inspired, an intra-articular injection therapy was fabricated to prevent cartilage degeneration with Col11a1-OE SMSC organoids. Col11a1-OE organoids ameliorated cartilage degeneration in DDH mice via regulating cellular senescence by up-regulated Col11a1/HIF1α-mediated glycolysis in chondrocytes.

Keywords

cellular senescence / genetics / glycolysis / hip dysplasia / organoids / osteoarthritis / single cell-sequencing

Cite this article

Download citation ▾
Ye Sun, Yongqing You, Qiang Wu, Rui Hu, Kerong Dai. Genetically inspired organoids prevent joint degeneration and alleviate chondrocyte senescence via Col11a1-HIF1α-mediated glycolysis-OXPHOS metabolism shift. Clinical and Translational Medicine, 2024, 14(2): e1574 DOI:10.1002/ctm2.1574

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Yang S, Zusman N, Lieberman E, Goldstein RY. Developmental dysplasia of the hip. Pediatrics. 2019;143(1):e20181147.

[2]

Sewell MD, Rosendahl K, Eastwood DM. Developmental dysplasia of the hip. BMJ. 2009;339:b4454.

[3]

Woodacre T, Ball T, Cox P. Epidemiology of developmental dysplasia of the hip within the UK: refining the risk factors. J Child Orthop. 2016;10(6):633-642.

[4]

Woodacre T, Dhadwal A, Ball T, Edwards C, Cox PJA. The costs of late detection of developmental dysplasia of the hip. J Child Orthop. 2014;8(4):325-332.

[5]

Ortiz-Neira CL, Paolucci EO, Donnon T. A meta-analysis of common risk factors associated with the diagnosis of developmental dysplasia of the hip in newborns. Eur J Radiol. 2012;81(3):e344-e351.

[6]

De Hundt M, Vlemmix F, Bais JMJ, et al. Risk factors for developmental dysplasia of the hip: a meta-analysis. Eur J Obstet Gynecol Reprod Biol. 2012;165(1):8-17.

[7]

Panagiotopoulou N, Bitar K, Hart WJ. The association between mode of delivery and developmental dysplasia of the hip in breech infants: a systematic review of 9 cohort studies. Acta Orthop Belg. 2012;78(6):697-702.

[8]

Li L, Wang Xi, Zhao Q, et al. CX3CR1 polymorphisms associated with an increased risk of developmental dysplasia of the hip in human. J Orthop Res. 2017;35(2):377-380.

[9]

Nejadhosseinian M, Haerian H, Shirkoohi R, Karami J, Mortazavi SMJ. Evaluation of CX3CR1 gene DNA methylation in developmental dysplasia of the hip (DDH). J Orthop Surg Res. 2022;17(1):436.

[10]

Baghdadi T, Nejadhosseinian M, Shirkoohi R, et al. DNA hypermethylation of GDF5 in developmental dysplasia of the hip (DDH). Mol Genet Genomic Med. 2019;7(9):e887.

[11]

Hong L, Wang K, Yan W, et al. High performance immunochromatographic assay for simultaneous quantitative detection of multiplex cardiac markers based on magnetic nanobeads. Theranostics. 2018;8(22):6121-6131.

[12]

Shi D, Dai J, Ikegawa S, Jiang Q. Genetic study on developmental dysplasia of the hip. Eur J Clin Invest. 2012;42(10):1121-1125.

[13]

Ma W, Zha Z, Chen K, et al. Genetic association study of common variants in TGFB1 and IL-6 with developmental dysplasia of the hip in Han Chinese population. Sci Rep. 2017;7(1):10287.

[14]

Sun M, Luo EY, Adams SM, et al. Collagen XI regulates the acquisition of collagen fibril structure, organization and functional properties in tendon. Matrix Biol. 2020;94:77-94.

[15]

Hafez A, Squires R, Pedracini A, Joshi A, Seegmiller R, Oxford J. Col11a1 regulates bone microarchitecture during embryonic development. J Dev Biol. 2015;3(4):158-176.

[16]

Hufnagel SB, Weaver KN, Hufnagel RB, Bader PI, Schorry EK, Hopkin RJ. A novel dominant COL11A1 mutation resulting in a severe skeletal dysplasia. Am J Med Genet A. 2014;164A(10):2607-2612.

[17]

Guo L, Elcioglu NH, Wang Z, et al. Novel and recurrent COL11A1 and COL2A1 mutations in the Marshall-Stickler syndrome spectrum. Hum Genome Var. 2017;4:17040.

[18]

Styrkarsdottir U, Lund SH, Thorleifsson G, 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. 2018;50(12):1681-1687.

[19]

Jiang H, Yang Q, Jiang J, Zhan X, Xiao Z. Association between COL11A1 (rs1337185) and ADAMTS5 (rs162509) gene polymorphisms and lumbar spine pathologies in Chinese Han population: an observational study. BMJ Open. 2017;7(5):e015644.

[20]

Todhunter RJ, Garrison SJ, Jordan J, et al. Gene expression in hip soft tissues in incipient canine hip dysplasia and osteoarthritis. J Orthop Res. 2019;37(2):313-324.

[21]

Sugimoto S, Kobayashi E, Fujii M, et al. An organoid-based organ-repurposing approach to treat short bowel syndrome. Nature. 2021;592(7852):99-104.

[22]

Sampaziotis F, Muraro D, Tysoe OC, et al. Cholangiocyte organoids can repair bile ducts after transplantation in the human liver. Science. 2021;371(6531):839-846.

[23]

Sun Ye, You Y, Jiang W, Zhai Z, Dai K. 3D-bioprinting a genetically inspired cartilage scaffold with GDF5-conjugated BMSC-laden hydrogel and polymer for cartilage repair. Theranostics. 2019;9(23):6949-6961.

[24]

Sun Ye, Wu Q, Dai K, You Y, Jiang W. Generating 3D-cultured organoids for pre-clinical modeling and treatment of degenerative joint disease. Signal Transduct Target Ther. 2021;6(1):380.

[25]

Sun Ye, Wang C, Hao Z, et al. A common variant of ubiquinol-cytochrome c reductase complex is associated with DDH. PLoS One. 2015;10(4):e0120212.

[26]

Hatzikotoulas K, Roposch A, Wainwright A, et al. Genome-wide association study of developmental dysplasia of the hip identifies an association with GDF5. Commun Biol. 2018;1:56.

[27]

Lek M, Karczewski KJ, Minikel EV, et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536(7616):285-291.

[28]

Booth KT, Askew JW, Talebizadeh Z, et al. Splice-altering variant in COL11A1 as a cause of nonsyndromic hearing loss DFNA37. Genet Med. 2018;21(4):948-954.

[29]

Matsuzaki T, Alvarez-Garcia O, Mokuda S, et al. FoxO transcription factors modulate autophagy and proteoglycan 4 in cartilage homeostasis and osteoarthritis. Sci Transl Med. 2018;10(428):eaan0746.

[30]

Kreuser U, Buchert J, Haase A, Richter W, Diederichs S. Initial WNT/beta-catenin activation enhanced mesoderm commitment, extracellular matrix expression, cell aggregation and cartilage tissue yield from induced pluripotent stem cells. Front Cell Dev Biol. 2020;8:581331.

[31]

Kennedy D, Mnich K, Oommen D, et al. HSPB1 facilitates ERK-mediated phosphorylation and degradation of BIM to attenuate endoplasmic reticulum stress-induced apoptosis. Cell Death Dis. 2017;8(8):e3026.

[32]

Wei T, Kulkarni NH, Zeng QQ, et al. Analysis of early changes in the articular cartilage transcriptisome in the rat meniscal tear model of osteoarthritis: pathway comparisons with the rat anterior cruciate transection model and with human osteoarthritic cartilage. Osteoarthritis Cartilage. 2010;18(7):992-1000.

[33]

Ji Q, Zheng Y, Zhang G, et al. Single-cell RNA-seq analysis reveals the progression of human osteoarthritis. Ann Rheum Dis. 2019;78(1):100-110.

[34]

Chen Y, Yu Y, Wen Ya, et al. A high-resolution route map reveals distinct stages of chondrocyte dedifferentiation for cartilage regeneration. Bone Res. 2022;10(1):38.

[35]

Mobasheri A, Rayman MP, Gualillo O, Sellam J, Van Der Kraan P, Fearon U. The role of metabolism in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2017;13(5):302-311.

[36]

Arra M, Swarnkar G, Ke Ke, et al. LDHA-mediated ROS generation in chondrocytes is a potential therapeutic target for osteoarthritis. Nat Commun. 2020;11(1):3427.

[37]

Matsuoka K, Bakiri L, Bilban M, et al. Metabolic rewiring controlled by c-Fos governs cartilage integrity in osteoarthritis. Ann Rheum Dis. 2023;82(9):1227-1239.

[38]

Wu X, Fan X, Crawford R, Xiao Y, Prasadam I. The metabolic landscape in osteoarthritis. Aging Dis. 2022;13(4):1166-1182.

[39]

Nishida T, Kubota S, Aoyama E, Takigawa M. Impaired glycolytic metabolism causes chondrocyte hypertrophy-like changes via promotion of phospho-Smad1/5/8 translocation into nucleus. Osteoarthritis Cartilage. 2013;21(5):700-709.

[40]

Mladenova M, Todorov T, Grozdanova L, Mitev V, Todorova A. Novel mutation in the COL11A1 gene causing Marshall-Stickler syndrome in three generations of a Bulgarian family. Balkan J Med Genet. 2021;24(1):95-98.

[41]

Brizola E, Gnoli M, Tremosini M, et al. Variable clinical expression of Stickler Syndrome: a case report of a novel COL11A1 mutation. Mol Genet Genomic Med. 2020;8(9):e1353.

[42]

Micale L, Morlino S, Schirizzi A, et al. Exon-trapping assay improves clinical interpretation of COL11A1 and COL11A2 intronic variants in Stickler syndrome type 2 and otospondylomegaepiphyseal dysplasia. Genes (Basel). 2020;11(12):1513.

[43]

Tompson SW, Bacino CA, Safina NP, et al. Fibrochondrogenesis results from mutations in the COL11A1 type XI collagen gene. Am J Hum Genet. 2010;87(5):708-712.

[44]

Wang J, Zhang C, Wu SG, et al. Additional evidence supports association of common variants in COL11A1 with increased risk of hip osteoarthritis susceptibility. Genet Test Mol Biomarkers. 2017;21(2):86-91.

[45]

Raine EVA, Dodd AW, Reynard LN, Loughlin J. Allelic expression analysis of the osteoarthritis susceptibility gene COL11A1 in human joint tissues. BMC Musculoskelet Disord. 2013;14:85.

[46]

Taylor SEB, Li YH, Wong WH, Bhutani N. Genome-wide mapping of DNA hydroxymethylation in osteoarthritic chondrocytes. Arthritis Rheumatol. 2015;67(8):2129-2140.

[47]

Holyoak DT, Otero M, Armar NS, et al. Collagen XI mutation lowers susceptibility to load-induced cartilage damage in mice. J Orthop Res. 2018;36(2):711-720.

[48]

Riuzzi F, Sorci G, Donato R. The amphoterin (HMGB1)/receptor for advanced glycation end products (RAGE) pair modulates myoblast proliferation, apoptosis, adhesiveness, migration, and invasiveness. Functional inactivation of RAGE in L6 myoblasts results in tumor formation in vivo. J Biol Chem. 2006;281(12):8242-8253.

RIGHTS & PERMISSIONS

2024 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

227

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/