Current cutting-edge omics techniques on musculoskeletal tissues and diseases

Xiaofei Li , Liang Fang , Renpeng Zhou , Lutian Yao , Sade W. Clayton , Samantha Muscat , Dakota R. Kamm , Cuicui Wang , Chuan-Ju Liu , Ling Qin , Robert J. Tower , Courtney M. Karner , Farshid Guilak , Simon Y. Tang , Alayna E. Loiselle , Gretchen A. Meyer , Jie Shen

Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 59

PDF
Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 59 DOI: 10.1038/s41413-025-00442-z
Review Article

Current cutting-edge omics techniques on musculoskeletal tissues and diseases

Author information +
History +
PDF

Abstract

Musculoskeletal disorders, including osteoarthritis, rheumatoid arthritis, osteoporosis, bone fracture, intervertebral disc degeneration, tendinopathy, and myopathy, are prevalent conditions that profoundly impact quality of life and place substantial economic burdens on healthcare systems. Traditional bulk transcriptomics, genomics, proteomics, and metabolomics have played a pivotal role in uncovering disease-associated alterations at the population level. However, these approaches are inherently limited in their ability to resolve cellular heterogeneity or to capture the spatial organization of cells within tissues, thus hindering a comprehensive understanding of the complex cellular and molecular mechanisms underlying these diseases. To address these limitations, advanced single-cell and spatial omics techniques have emerged in recent years, offering unparalleled resolution for investigating cellular diversity, tissue microenvironments, and biomolecular interactions within musculoskeletal tissues. These cutting-edge techniques enable the detailed mapping of the molecular landscapes in diseased tissues, providing transformative insights into pathophysiological processes at both the single-cell and spatial levels. This review presents a comprehensive overview of the latest omics technologies as applied to musculoskeletal research, with a particular focus on their potential to revolutionize our understanding of disease mechanisms. Additionally, we explore the power of multi-omics integration in identifying novel therapeutic targets and highlight key challenges that must be overcome to successfully translate these advancements into clinical applications.

Cite this article

Download citation ▾
Xiaofei Li, Liang Fang, Renpeng Zhou, Lutian Yao, Sade W. Clayton, Samantha Muscat, Dakota R. Kamm, Cuicui Wang, Chuan-Ju Liu, Ling Qin, Robert J. Tower, Courtney M. Karner, Farshid Guilak, Simon Y. Tang, Alayna E. Loiselle, Gretchen A. Meyer, Jie Shen. Current cutting-edge omics techniques on musculoskeletal tissues and diseases. Bone Research, 2025, 13(1): 59 DOI:10.1038/s41413-025-00442-z

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

FelsenthalN, ZelzerE. Mechanical regulation of musculoskeletal system development. Development, 2017, 144: 4271-4283

[2]

FronteraWR. Physiologic changes of the musculoskeletal system with aging: a brief review. Phys. Med. Rehabil. Clin. N. Am., 2017, 28: 705-711

[3]

Mitchell, T. The Musculoskeletal System (Master Books, 2015).

[4]

ShenJ, LiS, ChenD. TGF-beta signaling and the development of osteoarthritis. Bone Res., 2014, 2: 14002

[5]

El-TallawySN, et al.. Management of musculoskeletal pain: an update with emphasis on chronic musculoskeletal pain. Pain. Ther., 2021, 10: 181-209

[6]

MeasePJ. Inflammatory musculoskeletal disease: identification and assessment. J. Rheumatol., 2011, 38: 557-561

[7]

AbdelhamidRE, SlukaKA. ASICs mediate pain and inflammation in musculoskeletal diseases. Physiology, 2015, 30: 449-459

[8]

LiX, et al.. Mitigation of articular cartilage degeneration and subchondral bone sclerosis in osteoarthritis progression using low-intensity ultrasound stimulation. Ultrasound Med. Biol., 2019, 45: 148-159

[9]

MobasheriA. Applications of proteomics to osteoarthritis, a musculoskeletal disease characterized by aging. Front. Physiol., 2011, 2: 108

[10]

NielsonCM, JacobsJM, OrwollES. Proteomic studies of bone and skeletal health outcomes. Bone, 2019, 126: 18-26

[11]

PauperM, et al.. Proteomic profiling towards a better understanding of genetic based muscular diseases: the current picture and a look to the future. Biomolecules, 2025, 15: 130

[12]

SwankKR, et al.. Metabolomic profiling in the characterization of degenerative bone and joint diseases. Metabolites, 2020, 10: 223

[13]

KumarA, et al.. Metabolomic analysis of primary human skeletal muscle cells during myogenic progression. Sci. Rep., 2020, 10 ArticleID: 11824

[14]

WeiG, et al.. Risk of metabolic abnormalities in osteoarthritis: a new perspective to understand its pathological mechanisms. Bone Res., 2023, 11: 63

[15]

WuS, OhbaS, MatsushitaY. Single-cell RNA-sequencing reveals the skeletal cellular dynamics in bone repair and osteoporosis. Int. J. Mol. Sci., 2023, 24: 9814

[16]

WangT, et al.. Single-cell RNA sequencing in orthopedic research. Bone Res., 2023, 11: 10

[17]

WangS, et al.. The evolution of single-cell RNA sequencing technology and application: progress and perspectives. Int. J. Mol. Sci., 2023, 24: 2943

[18]

Arias-HidalgoC, et al.. Single-cell proteomics: the critical role of nanotechnology. Int. J. Mol. Sci., 2022, 23: 6707

[19]

AhmadR, BudnikB. A review of the current state of single-cell proteomics and future perspective. Anal. Bioanal. Chem., 2023, 415: 6889-6899

[20]

ZhangZ, et al.. When cancer drug resistance meets metabolomics (bulk, single-cell and/or spatial): progress, potential, and perspective. Front. Oncol., 2022, 12: 1054233

[21]

FengS, LiJ, TianJ, LuS, ZhaoY. Application of single-cell and spatial omics in musculoskeletal disorder research. Int. J. Mol. Sci., 2023, 24: 2271

[22]

RaiMF, et al.. Single cell omics for musculoskeletal research. Curr. Osteoporos. Rep., 2021, 19: 131-140

[23]

RaiMF, et al.. Three decades of advancements in osteoarthritis research: insights from transcriptomic, proteomic, and metabolomic studies. Osteoarthr. Cartil., 2024, 32: 385-397

[24]

TangF, et al.. mRNA-Seq whole-transcriptome analysis of a single cell. Nat. Methods, 2009, 6: 377-382

[25]

BuenrostroJD, et al.. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature, 2015, 523: 486-490

[26]

BendallSC, et al.. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science, 2011, 332: 687-696

[27]

HughesAJ, et al.. Single-cell western blotting. Nat. Methods, 2014, 11: 749-U794

[28]

KennedyRT, StclaireRL, WhiteJG, JorgensonJW. Chemical-analysis of single neurons by open tubular liquid-chromatography. Mikrochim. Acta, 1987, 2: 37-45

[29]

StahlPL, et al.. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science, 2016, 353: 78-82

[30]

RappsilberJ, SiniossoglouS, HurtEC, MannM. A generic strategy to analyze the spatial organization of multi-protein complexes by cross-linking and mass spectrometry. Anal. Chem., 2000, 72: 267-275

[31]

HuT, et al.. Single-cell spatial metabolomics with cell-type specific protein profiling for tissue systems biology. Nat. Commun., 2023, 14 ArticleID: 8260

[32]

RosenbergerFA, et al.. Spatial single-cell mass spectrometry defines zonation of the hepatocyte proteome. Nat. Methods, 2023, 20: 1530-1536

[33]

PlanqueM, IgelmannS, Ferreira CamposAM, FendtSM. Spatial metabolomics principles and application to cancer research. Curr. Opin. Chem. Biol., 2023, 76: 102362

[34]

PapT, Korb-PapA. Cartilage damage in osteoarthritis and rheumatoid arthritis-two unequal siblings. Nat. Rev. Rheumatol., 2015, 11: 606-615

[35]

TongL, et al.. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res., 2022, 10: 60

[36]

ChenY, et al.. A high-resolution route map reveals distinct stages of chondrocyte dedifferentiation for cartilage regeneration. Bone Res., 2022, 10: 38

[37]

JiQ, et al.. Single-cell RNA-seq analysis reveals the progression of human osteoarthritis. Ann. Rheum. Dis., 2019, 78: 100-110

[38]

YanM, et al.. Single-cell RNA sequencing reveals distinct chondrocyte states in femoral cartilage under weight-bearing load in Rheumatoid arthritis. Front. Immunol., 2023, 14 ArticleID: 1247355

[39]

SebastianA, et al.. Single-cell RNA-seq reveals transcriptomic heterogeneity and post-traumatic osteoarthritis-associated early molecular changes in mouse articular chondrocytes. Cells, 2021, 10: 1462

[40]

FanY, et al.. Unveiling inflammatory and prehypertrophic cell populations as key contributors to knee cartilage degeneration in osteoarthritis using multi-omics data integration. Ann. Rheum. Dis., 2024, 83: 926-944

[41]

SwahnH, et al.. Senescent cell population with ZEB1 transcription factor as its main regulator promotes osteoarthritis in cartilage and meniscus. Ann. Rheum. Dis., 2023, 82: 403-415

[42]

GuangZ, MinZ, Jun-TanL, Tian-XuD, XiangG. Single-cell protein activity analysis reveals a novel subpopulation of chondrocytes and the corresponding key master regulator proteins associated with anti-senescence and OA progression. Front. Immunol., 2023, 14: 1077003

[43]

KangX, ZhangK, WangY, ZhaoY, LuY. Single-cell RNA sequencing analysis of human chondrocytes reveals cell-cell communication alterations mediated by interactive signaling pathways in osteoarthritis. Front. Cell Dev. Biol., 2023, 11: 1099287

[44]

ChenY, ZhangY, GeY, RenH. Integrated single-cell and bulk RNA sequencing analysis identified pyroptosis-related signature for diagnosis and prognosis in osteoarthritis. Sci. Rep., 2023, 13 ArticleID: 17757

[45]

LiH, et al.. Combining single-cell RNA sequencing and population-based studies reveals hand osteoarthritis-associated chondrocyte subpopulations and pathways. Bone Res., 2023, 11: 58

[46]

LvZ, et al.. Single cell RNA-seq analysis identifies ferroptotic chondrocyte cluster and reveals TRPV1 as an anti-ferroptotic target in osteoarthritis. EBioMedicine, 2022, 84 ArticleID: 104258

[47]

FuW, et al.. 14-3-3 epsilon is an intracellular component of TNFR2 receptor complex and its activation protects against osteoarthritis. Ann. Rheum. Dis., 2021, 80: 1615-1627

[48]

FuW, et al.. Na(v)1.7 as a chondrocyte regulator and therapeutic target for osteoarthritis. Nature, 2024, 625: 557-565

[49]

NingY, et al.. Integrative analysis of miRNA in cartilage-derived extracellular vesicles and single-cell RNA-seq profiles in knee osteoarthritis. Arch. Biochem. Biophys., 2023, 748 ArticleID: 109785

[50]

ZhangY, et al.. Dual functions of microRNA-17 in maintaining cartilage homeostasis and protection against osteoarthritis. Nat. Commun., 2022, 13 ArticleID: 2447

[51]

LiJ, et al.. Microtubule stabilization targeting regenerative chondrocyte cluster for cartilage regeneration. Theranostics, 2023, 13: 3480-3496

[52]

FanX, et al.. Spatial analysis of the osteoarthritis microenvironment: techniques, insights, and applications. Bone Res., 2024, 12: 7

[53]

LareauCA, et al.. Mitochondrial single-cell ATAC-seq for high-throughput multi-omic detection of mitochondrial genotypes and chromatin accessibility. Nat. Protoc., 2023, 18: 1416-1440

[54]

BittnerN, et al.. Primary osteoarthritis chondrocyte map of chromatin conformation reveals novel candidate effector genes. Ann. Rheum. Dis., 2024, 83: 1048-1059

[55]

LiuY, et al.. Chromatin accessibility landscape of articular knee cartilage reveals aberrant enhancer regulation in osteoarthritis. Sci. Rep., 2018, 8 ArticleID: 15499

[56]

ZhangCH, et al.. Creb5 establishes the competence for Prg4 expression in articular cartilage. Commun. Biol., 2021, 4: 332

[57]

RichardD, et al.. Evolutionary selection and constraint on human knee chondrocyte regulation impacts osteoarthritis risk. Cell, 2020, 181: 362-381.e328

[58]

ShenJ, et al.. Inhibition of 4-aminobutyrate aminotransferase protects against injury-induced osteoarthritis in mice. JCI Insight, 2019, 4: e128568

[59]

ShenJ, et al.. DNA methyltransferase 3b regulates articular cartilage homeostasis by altering metabolism. JCI Insight, 2017, 2: e93612

[60]

LoughlinJ, ReynardLN. Osteoarthritis: epigenetics of articular cartilage in knee and hip OA. Nat. Rev. Rheumatol., 2015, 11: 6-7

[61]

RushtonMD, et al.. Characterization of the cartilage DNA methylome in knee and hip osteoarthritis. Arthritis Rheumatol., 2014, 66: 2450-2460

[62]

KehayovaYS, WilkinsonJM, RiceSJ, LoughlinJ. Mediation of the same epigenetic and transcriptional effect by independent osteoarthritis risk-conferring alleles on a shared target gene, COLGALT2. Arthritis Rheumatol., 2023, 75: 910-922

[63]

RobertsJB, et al.. Specific isoforms of the ubiquitin ligase gene WWP2 are targets of osteoarthritis genetic risk via a differentially methylated DNA sequence. Arthritis Res. Ther., 2024, 26: 78

[64]

BrumwellA, et al.. Identification of TMEM129, encoding a ubiquitin-protein ligase, as an effector gene of osteoarthritis genetic risk. Arthritis Res. Ther., 2022, 24: 189

[65]

ZhaoL, WangQ, ZhangC, HuangC. Genome-wide DNA methylation analysis of articular chondrocytes identifies TRAF1, CTGF, and CX3CL1 genes as hypomethylated in osteoarthritis. Clin. Rheumatol., 2017, 36: 2335-2342

[66]

Alvarez-GarciaO, et al.. Increased DNA methylation and reduced expression of transcription factors in human osteoarthritis cartilage. Arthritis Rheumatol., 2016, 68: 1876-1886

[67]

ZhangY, et al.. Genome-wide DNA methylation profile implicates potential cartilage regeneration at the late stage of knee osteoarthritis. Osteoarthr. Cartil., 2016, 24: 835-843

[68]

KreitmaierP, et al.. An epigenome-wide view of osteoarthritis in primary tissues. Am. J. Hum. Genet., 2022, 109: 1255-1271

[69]

IzdaV, et al.. A pilot analysis of genome-wide DNA methylation patterns in mouse cartilage reveals overlapping epigenetic signatures of aging and osteoarthritis. ACR Open Rheumatol., 2022, 4: 1004-1012

[70]

WuJ, YangF, WuX, LiuX, ZhengD. Comparison of genome-wide DNA methylation patterns between antler precartilage and cartilage. Mol. Genet. Genom., 2023, 298: 343-352

[71]

LiJ, et al.. Multi-omics molecular biomarkers and database of osteoarthritis. Database, 2022, 2022: baac052

[72]

WeiY, et al.. Progress in multi-omics studies of osteoarthritis. Biomark. Res., 2025, 13 ArticleID: 26

[73]

GarciaBA, et al.. Protein profile of osteoarthritic human articular cartilage using tandem mass spectrometry. Rapid Commun. Mass Spectrom., 2006, 20: 2999-3006

[74]

WuJ, et al.. Comparative proteomic characterization of articular cartilage tissue from normal donors and patients with osteoarthritis. Arthritis Rheum., 2007, 56: 3675-3684

[75]

GuoD, et al.. Proteomic analysis of human articular cartilage: identification of differentially expressed proteins in knee osteoarthritis. Jt. Bone Spine, 2008, 75: 439-444

[76]

HermanssonM, et al.. Proteomic analysis of articular cartilage shows increased type II collagen synthesis in osteoarthritis and expression of inhibin betaA (activin A), a regulatory molecule for chondrocytes. J. Biol. Chem., 2004, 279: 43514-43521

[77]

PeffersMJ, BeynonRJ, CleggPD. Absolute quantification of selected proteins in the human osteoarthritic secretome. Int. J. Mol. Sci., 2013, 14: 20658-20681

[78]

Ruiz-RomeroC, Lopez-ArmadaMJ, BlancoFJ. Proteomic characterization of human normal articular chondrocytes: a novel tool for the study of osteoarthritis and other rheumatic diseases. Proteomics, 2005, 5: 3048-3059

[79]

Ruiz-RomeroC, et al.. Proteomic analysis of human osteoarthritic chondrocytes reveals protein changes in stress and glycolysis. Proteomics, 2008, 8: 495-507

[80]

IkedaD, AgetaH, TsuchidaK, YamadaH. iTRAQ-based proteomics reveals novel biomarkers of osteoarthritis. Biomarkers, 2013, 18: 565-572

[81]

FuW, et al.. TNFR2/14-3-3epsilon signaling complex instructs macrophage plasticity in inflammation and autoimmunity. J. Clin. Investig., 2021, 131: e144016

[82]

LouridoL, et al.. Secretome analysis of human articular chondrocytes unravels catabolic effects of nicotine on the joint. Proteom. Clin. Appl., 2016, 10: 671-680

[83]

HsuehMF, OnnerfjordP, KrausVB. Biomarkers and proteomic analysis of osteoarthritis. Matrix Biol., 2014, 39: 56-66

[84]

SanchezC, et al.. Chondrocyte secretome: a source of novel insights and exploratory biomarkers of osteoarthritis. Osteoarthr. Cartil., 2017, 25: 1199-1209

[85]

TrachanaV, MourmouraE, PapathanasiouI, TsezouA. Understanding the role of chondrocytes in osteoarthritis: utilizing proteomics. Expert Rev. Proteom., 2019, 16: 201-213

[86]

ZhengL, ZhangZ, ShengP, MobasheriA. The role of metabolism in chondrocyte dysfunction and the progression of osteoarthritis. Ageing Res. Rev., 2021, 66: 101249

[87]

WuX, FanX, CrawfordR, XiaoY, PrasadamI. The metabolic landscape in osteoarthritis. Aging Dis., 2022, 13: 1166-1182

[88]

AdamMS, ZhuangH, RenX, ZhangY, ZhouP. The metabolic characteristics and changes of chondrocytes in vivo and in vitro in osteoarthritis. Front. Endocrinol., 2024, 15: 1393550

[89]

AraiY, et al.. Cartilage homeostasis under physioxia. Int. J. Mol. Sci., 2024, 25: 9398

[90]

StegenS, et al.. Glutamine metabolism controls chondrocyte identity and function. Dev. Cell, 2020, 53: 530-544 e538

[91]

ArraM, et al.. Glutamine metabolism modulates chondrocyte inflammatory response. eLife, 2022, 11: e80725

[92]

LeeSY, AbelED, LongF. Glucose metabolism induced by Bmp signaling is essential for murine skeletal development. Nat. Commun., 2018, 9 ArticleID: 4831

[93]

WangC, et al.. Deletion of Glut1 in early postnatal cartilage reprograms chondrocytes toward enhanced glutamine oxidation. Bone Res., 2021, 9: 38

[94]

TabbaaSM, et al.. Elevated lipid metabolites in stored clinical OCA media correlate with chondrocyte death. Am. J. Sports Med., 2024, 52: 2119-2128

[95]

TangR, et al.. Gene therapy for fat-1 prevents obesity-induced metabolic dysfunction, cellular senescence, and osteoarthritis. Proc. Natl. Acad. Sci. USA, 2024, 121 ArticleID: e2402954121

[96]

DefoisA, et al.. Osteoarthritic chondrocytes undergo a glycolysis-related metabolic switch upon exposure to IL-1b or TNF. Cell Commun. Signal., 2023, 21 ArticleID: 137

[97]

WuX, et al.. Dysregulated energy metabolism impairs chondrocyte function in osteoarthritis. Osteoarthr. Cartil., 2023, 31: 613-626

[98]

TanC, LiL, HanJ, XuK, LiuX. A new strategy for osteoarthritis therapy: inhibition of glycolysis. Front. Pharmacol., 2022, 13: 1057229

[99]

PiP, et al.. The role of targeting glucose metabolism in chondrocytes in the pathogenesis and therapeutic mechanisms of osteoarthritis: a narrative review. Front. Endocrinol., 2024, 15, ArticleID: 1319827

[100]

JaggardMKJ, et al.. A systematic review of the small molecule studies of osteoarthritis using nuclear magnetic resonance and mass spectroscopy. Osteoarthr. Cartil., 2019, 27: 560-570

[101]

WelhavenHD, et al.. Metabolomic profiles and pathways in osteoarthritic human cartilage: a comparative analysis with healthy cartilage. Metabolites, 2024, 14: 183

[102]

Brahmachary, P., Erdogan, E., Myers, E. & June, R. K. Metabolomic profiling and characterization of a novel 3D culture system for studying chondrocyte mechanotransduction. bioRxiv (2024).

[103]

BatushanskyA, LopesEBP, ZhuS, HumphriesKM, GriffinTM. GC-MS method for metabolic profiling of mouse femoral head articular cartilage reveals distinct effects of tissue culture and development. Osteoarthr. Cartil., 2019, 27: 1361-1371

[104]

ZhaiG, RandellEW, RahmanP. Metabolomics of osteoarthritis: emerging novel markers and their potential clinical utility. Rheumatology, 2018, 57: 2087-2095

[105]

BorelM, et al.. Longitudinal profiling of articular cartilage degradation in osteoarthritis by high-resolution magic angle spinning 1H NMR spectroscopy: experimental study in the meniscectomized guinea pig model. J. Proteome Res., 2009, 8: 2594-2600

[106]

McCutchenCN, ZignegoDL, JuneRK. Metabolic responses induced by compression of chondrocytes in variable-stiffness microenvironments. J. Biomech., 2017, 64: 49-58

[107]

Bergstrom, A. R. et al. Metabolic profiles of encapsulated chondrocytes exposed to short-term simulated microgravity. bioRxiv (2024).

[108]

SouthanJ, McHughE, WalkerH, IsmailHM. Metabolic signature of articular cartilage following mechanical injury: an integrated transcriptomics and metabolomics analysis. Front. Mol. Biosci., 2020, 7: 592905

[109]

HuX, et al.. Identification of cellular heterogeneity and immunogenicity of chondrocytes via single-cell RNA sequencing technique in human osteoarthritis. Front. Pharmacol., 2022, 13 ArticleID: 1004766

[110]

XuW, et al.. Engineered biomechanical microenvironment of articular chondrocytes based on heterogeneous GelMA hydrogel composites and dynamic mechanical compression. Biomater. Adv., 2023, 153 ArticleID: 213567

[111]

ZhangC, Le DevedecSE, AliA, HankemeierT. Single-cell metabolomics by mass spectrometry: ready for primetime?. Curr. Opin. Biotechnol., 2023, 82: 102963

[112]

WeiD, XuM, WangZ, TongJ. The development of single-cell metabolism and its role in studying cancer emergent properties. Front. Oncol., 2021, 11: 814085

[113]

Cillero-PastorB, EijkelG, KissA, BlancoFJ, HeerenRM. Time-of-flight secondary ion mass spectrometry-based molecular distribution distinguishing healthy and osteoarthritic human cartilage. Anal. Chem., 2012, 84: 8909-8916

[114]

Eveque-MourrouxMR, et al.. Heterogeneity of lipid and protein cartilage profiles associated with human osteoarthritis with or without type 2 diabetes mellitus. J. Proteome Res., 2021, 20: 2973-2982

[115]

WuCL, et al.. Single cell transcriptomic analysis of human pluripotent stem cell chondrogenesis. Nat. Commun., 2021, 12 ArticleID: 362

[116]

BaldwinM, et al.. A roadmap for delivering a human musculoskeletal cell atlas. Nat. Rev. Rheumatol., 2023, 19: 738-752

[117]

DonlinLT, et al.. Methods for high-dimensional analysis of cells dissociated from cryopreserved synovial tissue. Arthritis Res. Ther., 2018, 20: 139

[118]

StephensonW, et al.. Single-cell RNA-seq of rheumatoid arthritis synovial tissue using low-cost microfluidic instrumentation. Nat. Commun., 2018, 9 ArticleID: 791

[119]

SimondsMM, SullivanKE, BresciaAC. Single-cell analysis reveals heterogeneity of juvenile idiopathic arthritis fibroblast-like synoviocytes with implications for disease subtype. Arthritis Res. Ther., 2022, 24: 225

[120]

WeiK, et al.. Notch signalling drives synovial fibroblast identity and arthritis pathology. Nature, 2020, 582: 259-264

[121]

BaiZ, et al.. Synovial fibroblast gene expression is associated with sensory nerve growth and pain in rheumatoid arthritis. Sci. Transl. Med., 2024, 16: eadk3506

[122]

ChenN, FanB, HeZ, YuX, WangJ. Identification of HBEGF+ fibroblasts in the remission of rheumatoid arthritis by integrating single-cell RNA sequencing datasets and bulk RNA sequencing datasets. Arthritis Res. Ther., 2022, 24: 215

[123]

WijesingheSN, et al.. Obesity defined molecular endotypes in the synovium of patients with osteoarthritis provides a rationale for therapeutic targeting of fibroblast subsets. Clin. Transl. Med., 2023, 13 ArticleID: e1232

[124]

TsuchiyaM, et al.. CD39+CD55- Fb subset exhibits myofibroblast-like phenotype and is associated with pain in osteoarthritis of the knee. Biomedicines, 2023, 11: 3047

[125]

ZhangF, et al.. Defining inflammatory cell states in rheumatoid arthritis joint synovial tissues by integrating single-cell transcriptomics and mass cytometry. Nat. Immunol., 2019, 20: 928-942

[126]

LiuF, et al.. Identification of FOXO1 as a geroprotector in human synovium through single-nucleus transcriptomic profiling. Protein Cell, 2024, 15: 441-459

[127]

CroftAP, et al.. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature, 2019, 570: 246-251

[128]

CulemannS, et al.. Locally renewing resident synovial macrophages provide a protective barrier for the joint. Nature, 2019, 572: 670-675

[129]

Harasymowicz, N. et al. Injury and obesity differentially and synergistically induce dysregulation of synovial immune cells in osteoarthritis. Ann. Rheum. Dis. S0003-4967, 00813-1 (2024).

[130]

WuCL, et al.. Conditional macrophage depletion increases inflammation and does not inhibit the development of osteoarthritis in obese macrophage Fas-induced apoptosis-transgenic mice. Arthritis Rheumatol., 2017, 69: 1772-1783

[131]

LiX, et al.. A single-cell RNA-sequencing analysis of distinct subsets of synovial macrophages in rheumatoid arthritis. DNA Cell Biol., 2023, 42: 212-222

[132]

HardtU, et al.. Integrated single cell and spatial transcriptomics reveal autoreactive differentiated B cells in joints of early rheumatoid arthritis. Sci. Rep., 2022, 12 ArticleID: 11876

[133]

DurhamLE, et al.. Substantive similarities between synovial fluid and synovial tissue T cells in inflammatory arthritis via single-cell RNA and T cell receptor sequencing. Arthritis Rheumatol., 2024, 76: 1594-1601

[134]

LiJ, et al.. Synovium and infrapatellar fat pad share common mesenchymal progenitors and undergo coordinated changes in osteoarthritis. J. Bone Min. Res., 2024, 39: 161-176

[135]

TangS, et al.. Single-cell atlas of human infrapatellar fat pad and synovium implicates APOE signaling in osteoarthritis pathology. Sci. Transl. Med., 2024, 16 ArticleID: eadf4590

[136]

CiechomskaM, RoszkowskiL, MaslinskiW. DNA methylation as a future therapeutic and diagnostic target in rheumatoid arthritis. Cells, 2019, 8: 953

[137]

TaylorPC, MooreA, VasilescuR, AlvirJ, TaralloM. A structured literature review of the burden of illness and unmet needs in patients with rheumatoid arthritis: a current perspective. Rheumatol. Int., 2016, 36: 685-695

[138]

HamS, et al.. Epigenetic analysis in rheumatoid arthritis synoviocytes. Exp. Mol. Med., 2019, 51: 1-13

[139]

AiR, et al.. Comprehensive epigenetic landscape of rheumatoid arthritis fibroblast-like synoviocytes. Nat. Commun., 2018, 9 ArticleID: 1921

[140]

Li YimAYF, et al.. Novel insights into rheumatoid arthritis through characterization of concordant changes in DNA methylation and gene expression in synovial biopsies of patients with differing numbers of swollen joints. Front. Immunol., 2021, 12 ArticleID: 651475

[141]

JiangF, et al.. A landscape of gene expression regulation for synovium in arthritis. Nat. Commun., 2024, 15 ArticleID: 1409

[142]

TsuchiyaH, OtaM, FujioK. Multiomics landscape of synovial fibroblasts in rheumatoid arthritis. Inflamm. Regen., 2021, 41: 7

[143]

GongX, et al.. An overview of multi-omics technologies in rheumatoid arthritis: applications in biomarker and pathway discovery. Front. Immunol., 2024, 15 ArticleID: 1381272

[144]

FalconerJ, et al.. Review: synovial cell metabolism and chronic inflammation in rheumatoid arthritis. Arthritis Rheumatol., 2018, 70: 984-999

[145]

ScanzelloCR, GoldringSR. The role of synovitis in osteoarthritis pathogenesis. Bone, 2012, 51: 249-257

[146]

HermanssonM, et al.. MS analysis of rheumatoid arthritic synovial tissue identifies specific citrullination sites on fibrinogen. Proteom. Clin. Appl., 2010, 4: 511-518

[147]

RochaB, et al.. Identification of a distinct lipidomic profile in the osteoarthritic synovial membrane by mass spectrometry imaging. Osteoarthr. Cartil., 2021, 29: 750-761

[148]

SuQH, et al.. Course-based intra-articular injection of medical chitosan mitigates excessive deposition of triacylglycerides in the synovial tissue of the knee osteoarthritis. J. Chin. Med. Assoc., 2024, 87: 870-877

[149]

GeM, et al.. Multi-omics analysis of synovial tissue and fluid reveals differentially expressed proteins and metabolites in osteoarthritis. J. Transl. Med., 2025, 23 ArticleID: 285

[150]

BaryawnoN, et al.. A cellular taxonomy of the bone marrow stroma in homeostasis and leukemia. Cell, 2019, 177: 1915-1932.e1916

[151]

TikhonovaAN, et al.. The bone marrow microenvironment at single-cell resolution. Nature, 2019, 569: 222-228

[152]

ZhongL, et al.. Single cell transcriptomics identifies a unique adipose lineage cell population that regulates bone marrow environment. eLife, 2020, 9 ArticleID: e54695

[153]

AyturkUM, et al.. Single-cell RNA sequencing of calvarial and long-bone endocortical cells. J. Bone Miner. Res., 2020, 35: 1981-1991

[154]

BaccinC, et al.. Combined single-cell and spatial transcriptomics reveal the molecular, cellular and spatial bone marrow niche organization. Nat. Cell Biol., 2020, 22: 38-48

[155]

MatsushitaY, et al.. A Wnt-mediated transformation of the bone marrow stromal cell identity orchestrates skeletal regeneration. Nat. Commun., 2020, 11 ArticleID: 332

[156]

ShiY, et al.. Gli1+ progenitors mediate bone anabolic function of teriparatide via Hh and Igf signaling. Cell Rep., 2021, 36: 109542

[157]

MatsushitaY, ChuAKY, OnoW, WelchJD, OnoN. Intercellular interactions of an adipogenic CXCL12-expressing stromal cell subset in murine bone marrow. J. Bone Miner. Res., 2021, 36: 1145-1158

[158]

ZhongL, YaoL, SealeP, QinL. Marrow adipogenic lineage precursor: a new cellular component of marrow adipose tissue. Best. Pr. Res. Clin. Endocrinol. Metab., 2021, 35: 101518

[159]

ZhongL, et al.. Transient expansion and myofibroblast conversion of adipogenic lineage precursors mediate bone marrow repair after radiation. JCI Insight, 2022, 7: e150323

[160]

LeimkuhlerNB, et al.. Heterogeneous bone-marrow stromal progenitors drive myelofibrosis via a druggable alarmin axis. Cell Stem Cell, 2021, 28: 637-652.e638

[161]

HallTD, et al.. Murine fetal bone marrow does not support functional hematopoietic stem and progenitor cells until birth. Nat. Commun., 2022, 13 ArticleID: 5403

[162]

LiuY, et al.. A specialized bone marrow microenvironment for fetal haematopoiesis. Nat. Commun., 2022, 13 ArticleID: 1327

[163]

de JongMME, et al.. The multiple myeloma microenvironment is defined by an inflammatory stromal cell landscape. Nat. Immunol., 2021, 22: 769-780

[164]

LiH, et al.. Identification of phenotypically, functionally, and anatomically distinct stromal niche populations in human bone marrow based on single-cell RNA sequencing. eLife, 2023, 12 ArticleID: e81656

[165]

WangZ, et al.. Single-cell transcriptome atlas of human mesenchymal stem cells exploring cellular heterogeneity. Clin. Transl. Med., 2021, 11 ArticleID: e650

[166]

WangZ, et al.. Single-cell RNA sequencing deconvolutes the in vivo heterogeneity of human bone marrow-derived mesenchymal stem cells. Int. J. Biol. Sci., 2021, 17: 4192-4206

[167]

BandyopadhyayS, et al.. Mapping the cellular biogeography of human bone marrow niches using single-cell transcriptomics and proteomic imaging. Cell, 2024, 2: 00408-00402

[168]

JardineL, et al.. Blood and immune development in human fetal bone marrow and Down syndrome. Nature, 2021, 598: 327-331

[169]

ZhangP, et al.. Characterization of mesenchymal stem cells in human fetal bone marrow by single-cell transcriptomic and functional analysis. Signal Transduct. Target. Ther., 2023, 8: 126

[170]

XiaoX, et al.. Spatial transcriptomic interrogation of the murine bone marrow signaling landscape. Bone Res., 2023, 11: 59

[171]

Bigham-SadeghA, OryanA. Basic concepts regarding fracture healing and the current options and future directions in managing bone fractures. Int. Wound J., 2015, 12: 238-247

[172]

Sheen, J. R., Mabrouk, A. & Garla, V. V. Fracture healing overview. in StatPearls (StatPearls Publishing, 2024).

[173]

WildemannB, et al.. Non-union bone fractures. Nat. Rev. Dis. Prim., 2021, 7: 57

[174]

SalichosL, ThayavallyR, KloenP, HadjiargyrouM. Human nonunion tissues display differential gene expression in comparison to physiological fracture callus. Bone, 2024, 183: 117091

[175]

WangC, Abu-AmerY, O’KeefeRJ, ShenJ. Loss of Dnmt3b in chondrocytes leads to delayed endochondral ossification and fracture repair. J. Bone Miner. Res., 2018, 33: 283-297

[176]

AmbrosiTH, et al.. Aged skeletal stem cells generate an inflammatory degenerative niche. Nature, 2021, 597: 256-262

[177]

LinX, et al.. Aged callus skeletal stem/progenitor cells contain an inflammatory osteogenic population with increased IRF and NF-kappaB pathways and reduced osteogenic potential. Front. Mol. Biosci., 2022, 9 ArticleID: 806528

[178]

LiuJ, et al.. Molecular signatures distinguish senescent cells from inflammatory cells in aged mouse callus stromal cells. Front. Endocrinol., 2023, 14, ArticleID: 1090049

[179]

ZouNY, et al.. Age-related secretion of grancalcin by macrophages induces skeletal stem/progenitor cell senescence during fracture healing. Bone Res., 2024, 12: 6

[180]

ZhangH, et al.. Single-cell RNA sequencing reveals B cells are important regulators in fracture healing. Front. Endocrinol., 2021, 12, ArticleID: 666140

[181]

XiaoD, et al.. DNA methylation-mediated Rbpjk suppression protects against fracture nonunion caused by systemic inflammation. J. Clin. Investig., 2023, 134: e168558

[182]

AvinKG, et al.. Single-cell RNAseq provides insight into altered immune cell populations in human fracture nonunions. J. Orthop. Res., 2023, 41: 1060-1069

[183]

SerowokyMA, et al.. A murine model of large-scale bone regeneration reveals a selective requirement for Sonic Hedgehog. NPJ Regen. Med., 2022, 7: 30

[184]

YaoL, et al.. Activin A marks a novel progenitor cell population during fracture healing and reveals a therapeutic strategy. eLife, 2023, 12: e89822

[185]

XuT, WangC, ShenJ, TongP, O’KeefeR. Ablation of Dnmt3b in chondrocytes suppresses cell maturation during embryonic development. J. Cell Biochem., 2018, 119: 5852-5863

[186]

WangC, et al.. Targeting angiogenesis for fracture nonunion treatment in inflammatory disease. Bone Res., 2021, 9: 29

[187]

Mancinelli, L., Schoedel, K. E., Weiss, K. R. & Intini, G. Methods to enable spatial transcriptomics of bone tissues. J. Vis. Exp. 3 (2024).

[188]

JiangW, CaruanaDL, BackJ, LeeFY. Unique spatial transcriptomic profiling of the murine femoral fracture callus: a preliminary report. Cells, 2024, 13: 522

[189]

RiosJJ, et al.. Spatial transcriptomics implicates impaired BMP signaling in NF1 fracture pseudarthrosis in murine and patient tissues. JCI Insight, 2024, 9: 522

[190]

ShenL, HuG, KarnerCM. Bioenergetic metabolism in osteoblast differentiation. Curr. Osteoporos. Rep., 2022, 20: 53-64

[191]

StegenS, CarmelietG. Metabolic regulation of skeletal cell fate and function. Nat. Rev. Endocrinol., 2024, 20: 399-413

[192]

van GastelN, CarmelietG. Metabolic regulation of skeletal cell fate and function in physiology and disease. Nat. Metab., 2021, 3: 11-20

[193]

YuY, et al.. Glutamine metabolism regulates proliferation and lineage allocation in skeletal stem cells. Cell Metab., 2019, 29: 966-978.e964

[194]

StegenS, et al.. Glutamine metabolism in osteoprogenitors is required for bone mass accrual and PTH-induced bone anabolism in male mice. J. Bone Miner. Res., 2021, 36: 604-616

[195]

StegenS, et al.. HIF-1alpha promotes glutamine-mediated redox homeostasis and glycogen-dependent bioenergetics to support postimplantation bone cell survival. Cell Metab., 2016, 23: 265-279

[196]

HuG, et al.. Glutathione limits RUNX2 oxidation and degradation to regulate bone formation. JCI Insight, 2023, 8: e166888

[197]

SharmaD, YuY, ShenL, ZhangGF, KarnerCM. SLC1A5 provides glutamine and asparagine necessary for bone development in mice. eLife, 2021, 10: e71595

[198]

ShenL, et al.. SLC38A2 provides proline to fulfill unique synthetic demands arising during osteoblast differentiation and bone formation. eLife, 2022, 11: e76963

[199]

ShenL, YuY, KarnerCM. SLC38A2 provides proline and alanine to regulate postnatal bone mass accrual in mice. Front. Physiol., 2022, 13: 992679

[200]

ElefteriouF, et al.. ATF4 mediation of NF1 functions in osteoblast reveals a nutritional basis for congenital skeletal dysplasiae. Cell Metab., 2006, 4: 441-451

[201]

RachedMT, et al.. FoxO1 is a positive regulator of bone formation by favoring protein synthesis and resistance to oxidative stress in osteoblasts. Cell Metab., 2010, 11: 147-160

[202]

BianXue-Feng, LJ-F, SunJia-Ming, ZhangHui. 1HNMR metabolomics of MC3T3-E mouse osteoblast proliferation and alkaline phosphatase content by deer antler peptide amine. Chin. J. Anal. Chem., 2021, 49: 103-109

[203]

AraujoR, et al.. NMR metabolomics to study the metabolic response of human osteoblasts to non-poled and poled poly (L-lactic) acid. Magn. Reson. Chem., 2019, 57: 919-933

[204]

BispoDSC, et al.. NMR metabolomics assessment of osteogenic differentiation of adipose-tissue-derived mesenchymal stem cells. J. Proteome Res., 2022, 21: 654-670

[205]

MisraBB, JayapalanS, RichardsAK, HeldermanRCM, Rendina-RuedyE. Untargeted metabolomics in primary murine bone marrow stromal cells reveals distinct profile throughout osteoblast differentiation. Metabolomics, 2021, 17 ArticleID: 86

[206]

HeldermanRC, et al.. Loss of function of lysosomal acid lipase (LAL) profoundly impacts osteoblastogenesis and increases fracture risk in humans. Bone, 2021, 148 ArticleID: 115946

[207]

TencerovaM, et al.. Metabolic programming determines the lineage-differentiation fate of murine bone marrow stromal progenitor cells. Bone Res., 2019, 7: 35

[208]

WelhavenHD, et al.. The cortical bone metabolome of C57BL/6J mice is sexually dimorphic. JBMR, 2022, 6 e10654

[209]

NamM, et al.. Metabolic alterations in the bone tissues of aged osteoporotic mice. Sci. Rep., 2018, 8 ArticleID: 8127

[210]

SongF, et al.. Osteoblast-intrinsic defect in glucose metabolism impairs bone formation in type II diabetic male mice. eLife, 2023, 12: e85714

[211]

ZhangX, et al.. Metabolomics insights into osteoporosis through association with bone mineral density. J. Bone Miner. Res., 2021, 36: 729-738

[212]

MeiZ, et al.. Association between the metabolome and bone mineral density in a Chinese population. EBioMedicine, 2020, 62 ArticleID: 103111

[213]

HuY, ZhangX, ShanY. LC-MS-based plasma metabolomics reveals metabolic variations in ovariectomy-induced osteoporosis in female Wistar rats. RSC Adv., 2018, 8: 24932-24941

[214]

GunturAR, et al.. Osteoblast-like MC3T3-E1 cells prefer glycolysis for ATP production but adipocyte-like 3T3-L1 cells prefer oxidative phosphorylation. J. Bone Min. Res., 2018, 33: 1052-1065

[215]

EsenE, et al.. WNT-LRP5 signaling induces Warburg effect through mTORC2 activation during osteoblast differentiation. Cell Metab., 2013, 17: 745-755

[216]

van GastelN, et al.. Lipid availability determines fate of skeletal progenitor cells via SOX9. Nature, 2020, 579: 111-117

[217]

SuY, et al.. Circulating amino acids are associated with bone mineral density decline and ten-year major osteoporotic fracture risk in older community-dwelling adults. Bone, 2019, 129 ArticleID: 115082

[218]

LiangW, et al.. An integrated multi-omics analysis reveals osteokines involved in global regulation. Cell Metab., 2024, 36: 1144-1163 e1147

[219]

ViolanteS, BerisaM, ThomasTH, CrossJR. Stable isotope tracers for metabolic pathway analysis. Methods Mol. Biol., 2019, 1978: 269-283

[220]

JangC, ChenL, RabinowitzJD. Metabolomics and isotope tracing. Cell, 2018, 173: 822-837

[221]

KarnerCM, EsenE, OkunadeAL, PattersonBW, LongF. Increased glutamine catabolism mediates bone anabolism in response to WNT signaling. J. Clin. Investig., 2015, 125: 551-562

[222]

LeeWC, JiX, NissimI, LongF. Malic enzyme couples mitochondria with aerobic glycolysis in osteoblasts. Cell Rep., 2020, 32: 108108

[223]

ZhangY, et al.. BAP1 links metabolic regulation of ferroptosis to tumour suppression. Nat. Cell Biol., 2018, 20: 1181-1192

[224]

DeVilbissAW, et al.. Metabolomic profiling of rare cell populations isolated by flow cytometry from tissues. eLife, 2021, 10: e61980

[225]

SemenzaGL. Hypoxia-inducible factors in physiology and medicine. Cell, 2012, 148: 399-408

[226]

WalshMC, et al.. Osteoimmunology: interplay between the immune system and bone metabolism. Annu. Rev. Immunol., 2006, 24: 33-63

[227]

BadurMG, ZhangH, MetalloCM. Enzymatic passaging of human embryonic stem cells alters central carbon metabolism and glycan abundance. Biotechnol. J., 2015, 10: 1600-1611

[228]

LaroucheJA, WallaceEC, SpenceBD, BurasE, AguilarCA. Spatiotemporal mapping of immune and stem cell dysregulation after volumetric muscle loss. JCI Insight, 2023, 8: e162835

[229]

TowerRJ, et al.. Spatial transcriptomics reveals a role for sensory nerves in preserving cranial suture patency through modulation of BMP/TGF-beta signaling. Proc. Natl. Acad. Sci. USA, 2021, 118: e2103087118

[230]

TowerRJ, et al.. Spatial transcriptomics reveals metabolic changes underly age-dependent declines in digit regeneration. eLife, 2022, 11: e71542

[231]

CheriefM, et al.. TrkA-mediated sensory innervation of injured mouse tendon supports tendon sheath progenitor cell expansion and tendon repair. Sci. Transl. Med., 2023, 15 ArticleID: eade4619

[232]

FuW, YangR, LiJ. Single-cell and spatial transcriptomics reveal changes in cell heterogeneity during progression of human tendinopathy. BMC Biol., 2023, 21 ArticleID: 132

[233]

Zhang, B. et al. A human embryonic limb cell atlas resolved in space and time. Nature 635, 668–678 (2023).

[234]

Mathavan, N. et al. Spatial transcriptomics in mechanomics: new horizons in exploring the mechanoregulation of bone regeneration. bioRxiv (2024).

[235]

Bian, F. et al. Spatial transcriptomics reveals the requirement of ADGRG6 in maintaining chondrocyte homeostasis in mouse growth plates. bioRxiv (2023).

[236]

WehrleE, GuntherD, MathavanN, SinghA, MullerR. Protocol for preparing formalin-fixed paraffin-embedded musculoskeletal tissue samples from mice for spatial transcriptomics. STAR Protoc., 2024, 5: 102986

[237]

BuckA, PradeVM, KunzkeT, ErbenRG, WalchA. Spatial metabolomics reveals upregulation of several pyrophosphate-producing pathways in cortical bone of Hyp mice. JCI Insight, 2022, 7: e162138

[238]

LuoL, et al.. Spatial metabolomics reveals skeletal myofiber subtypes. Sci. Adv., 2023, 9 ArticleID: eadd0455

[239]

ChanWC, SzeKL, SamartzisD, LeungVY, ChanD. Structure and biology of the intervertebral disk in health and disease. Orthop. Clin. North Am., 2011, 42: 447-464

[240]

Walk, R. et al. The progression of neurovascular features and chemokine signatures of the intervertebral disc with degeneration. bioRxiv (2024).

[241]

LintzM, WalkRE, TangSY, BonassarLJ. The degenerative impact of hyperglycemia on the structure and mechanics of developing murine intervertebral discs. JOR Spine, 2022, 5: e1191

[242]

Diaz-HernandezME, et al.. Derivation of notochordal cells from human embryonic stem cells reveals unique regulatory networks by single cell-transcriptomics. J. Cell Physiol., 2020, 235: 5241-5255

[243]

LiZ, et al.. Single-cell RNA sequencing reveals the difference in human normal and degenerative nucleus pulposus tissue profiles and cellular interactions. Front. Cell Dev. Biol., 2022, 10: 910626

[244]

FernandesLM, et al.. Single-cell RNA-seq identifies unique transcriptional landscapes of human nucleus pulposus and annulus fibrosus cells. Sci. Rep., 2020, 10 ArticleID: 15263

[245]

GanY, et al.. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res., 2021, 9: 37

[246]

HanS, et al.. Single-cell RNA sequencing of the nucleus pulposus reveals chondrocyte differentiation and regulation in intervertebral disc degeneration. Front. Cell Dev. Biol., 2022, 10 ArticleID: 824771

[247]

KuchynskyK, et al.. Transcriptional profiling of human cartilage endplate cells identifies novel genes and cell clusters underlying degenerated and non-degenerated phenotypes. Arthritis Res. Ther., 2024, 26: 12

[248]

WangJ, et al.. Novel biomarkers of intervertebral disc cells and evidence of stem cells in the intervertebral disc. Osteoarthr. Cartil., 2021, 29: 389-401

[249]

PanebiancoCJ, DaveA, CharytonowiczD, SebraR, IatridisJC. Single-cell RNA-sequencing atlas of bovine caudal intervertebral discs: discovery of heterogeneous cell populations with distinct roles in homeostasis. FASEB J., 2021, 35: e21919

[250]

CalióM, GantenbeinB, EgliM, PovedaL, IlleF. The cellular composition of bovine coccygeal intervertebral discs: a comprehensive single-cell RNAseq analysis. Int. J. Mol. Sci., 2021, 22: 4917

[251]

DiseaseGBD, InjuryI, PrevalenceC. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet, 2018, 392: 1789-1858

[252]

CherifH, et al.. Single-cell RNA-seq analysis of cells from degenerating and non-degenerating intervertebral discs from the same individual reveals new biomarkers for intervertebral disc degeneration. Int. J. Mol. Sci., 2022, 23: 3993

[253]

WangD, et al.. Single-cell transcriptomics reveals heterogeneity and intercellular crosstalk in human intervertebral disc degeneration. iScience, 2023, 26 ArticleID: 106692

[254]

WalkRE, MoonHJ, TangSY, GuptaMC. Contrast-enhanced microCT evaluation of degeneration following partial and full width injuries to the mouse lumbar intervertebral disc. Sci. Rep., 2022, 12 ArticleID: 15555

[255]

Clayton, S. W., Walk, R. E., Mpofu, L., Easson, G. W. D. & Tang, S. Y. Analysis of infiltrating immune cells following intervertebral disc injury reveals recruitment of gamma-delta (gammadelta) T cells in female mice. bioRxiv (2024).

[256]

LingZ, et al.. Single-cell RNA-seq analysis reveals macrophage involved in the progression of human intervertebral disc degeneration. Front. Cell Dev. Biol., 2021, 9 ArticleID: 833420

[257]

RohanifarM, et al.. Single cell RNA-sequence analyses reveal uniquely expressed genes and heterogeneous immune cell involvement in the rat model of intervertebral disc degeneration. Appl. Sci., 2022, 12: 8244

[258]

SwahnH, et al.. Shared and compartment-specific processes in nucleus pulposus and annulus fibrosus during intervertebral disc degeneration. Adv. Sci., 2024, 11, ArticleID: e2309032

[259]

JiangW, et al.. Intervertebral disc human nucleus pulposus cells associated with back pain trigger neurite outgrowth in vitro and pain behaviors in rats. Sci. Transl. Med., 2023, 15 ArticleID: eadg7020

[260]

ShaoT, et al.. The role of immunocyte infiltration regulatory network based on hdWGCNA and single-cell bioinformatics analysis in intervertebral disc degeneration. Inflammation, 2024, 47: 1987-1999

[261]

WangP, LiZ, YeD. Single-cell RNA-seq analysis reveals the Wnt/Ca(2+) signaling pathway with inflammation, apoptosis in nucleus pulposus degeneration. BMC Musculoskelet. Disord., 2024, 25 ArticleID: 321

[262]

ZhangY, et al.. Single-cell RNA-seq analysis identifies unique chondrocyte subsets and reveals involvement of ferroptosis in human intervertebral disc degeneration. Osteoarthr. Cartil., 2021, 29: 1324-1334

[263]

KamataniT, et al.. Human iPS cell-derived cartilaginous tissue spatially and functionally replaces nucleus pulposus. Biomaterials, 2022, 284 ArticleID: 121491

[264]

ZhouT, et al.. Spatiotemporal characterization of human early intervertebral disc formation at single-cell resolution. Adv. Sci., 2023, 10, ArticleID: e2206296

[265]

JiangW, et al.. Single-cell atlas unveils cellular heterogeneity and novel markers in human neonatal and adult intervertebral discs. iScience, 2022, 25 ArticleID: 104504

[266]

WangL, et al.. Revealing the immune infiltration landscape and identifying diagnostic biomarkers for lumbar disc herniation. Front. Immunol., 2021, 12 ArticleID: 666355

[267]

LiW, et al.. Deciphering the sequential changes of monocytes/macrophages in the progression of IDD with longitudinal approach using single-cell transcriptome. Front. Immunol., 2023, 14 ArticleID: 1090637

[268]

SunH, WangH, ZhangW, MaoH, LiB. Single-cell RNA sequencing reveals resident progenitor and vascularization-associated cell subpopulations in rat annulus fibrosus. J. Orthop. Transl., 2023, 38: 256-267

[269]

ZhangS, et al.. Inhibiting heat shock protein 90 attenuates nucleus pulposus fibrosis and pathologic angiogenesis induced by macrophages via down-regulating cell migration-inducing protein. Am. J. Pathol., 2023, 193: 960-976

[270]

ZhangZ, HuoJ, JiX, WeiL, ZhangJ. GREM1, LRPPRC and SLC39A4 as potential biomarkers of intervertebral disc degeneration: a bioinformatics analysis based on multiple microarray and single-cell sequencing data. BMC Musculoskelet. Disord., 2023, 24 ArticleID: 729

[271]

ChenY, et al.. Characterization of the nucleus pulposus progenitor cells via spatial transcriptomics. Adv. Sci., 2024, 11, ArticleID: e2303752

[272]

WilliamsCG, LeeHJ, AsatsumaT, Vento-TormoR, HaqueA. An introduction to spatial transcriptomics for biomedical research. Genome Med., 2022, 14 ArticleID: 68

[273]

ChenG, NingB, ShiT. Single-cell RNA-seq technologies and related computational data analysis. Front. Genet., 2019, 10: 317

[274]

HaglundL, OuelletJ, RoughleyP. Variation in chondroadherin abundance and fragmentation in the human scoliotic disc. Spine, 2009, 34: 1513-1518

[275]

McCannMR, et al.. Proteomic signature of the murine intervertebral disc. PLoS ONE, 2015, 10: e0117807

[276]

KudelkoM, et al.. PRIMUS: comprehensive proteomics of mouse intervertebral discs that inform novel biology and relevance to human disease modelling. Matrix Biol., 2021, 12, ArticleID: 100082

[277]

RajasekaranS, et al.. Proteomic signatures of healthy intervertebral discs from organ donors: a comparison with previous studies on discs from scoliosis, animals, and trauma. Neurospine, 2020, 17: 426-442

[278]

RajasekaranS, et al.. Can scoliotic discs be controls for molecular studies in intervertebral disc research? Insights from proteomics. Glob. Spine J., 2022, 12: 598-609

[279]

YeD, et al.. Comparative and quantitative proteomic analysis of normal and degenerated human annulus fibrosus cells. Clin. Exp. Pharmacol. Physiol., 2015, 42: 530-536

[280]

YeeA, et al.. Fibrotic-like changes in degenerate human intervertebral discs revealed by quantitative proteomic analysis. Osteoarthr. Cartil., 2016, 24: 503-513

[281]

EassonGWD, et al.. Modulation of TRPV4 protects against degeneration induced by sustained loading and promotes matrix synthesis in the intervertebral disc. FASEB J., 2023, 37 ArticleID: e22714

[282]

EassonGWD, SavadipourA, GonzalezC, GuilakF, TangSY. TRPV4 differentially controls inflammatory cytokine networks during static and dynamic compression of the intervertebral disc. JOR Spine, 2023, 6: e1282

[283]

WangY, et al.. Deficiency of MIF accentuates overloaded compression-induced nucleus pulposus cell oxidative damage via depressing mitophagy. Oxid. Med. Cell. Longev., 2021, 2021 ArticleID: 6192498

[284]

ZhangY, et al.. Early onset of disc degeneration in SM/J mice is associated with changes in ion transport systems and fibrotic events. Matrix Biol., 2018, 70: 123-139

[285]

PattappaG, et al.. CCL5/RANTES is a key chemoattractant released by degenerative intervertebral discs in organ culture. Eur. Cell Mater., 2014, 27: 124-136 discussion 136

[286]

ErwinWM, et al.. The biological basis of degenerative disc disease: proteomic and biomechanical analysis of the canine intervertebral disc. Arthritis Res. Ther., 2015, 17: 240

[287]

BachFC, et al.. Soluble and pelletable factors in porcine, canine and human notochordal cell-conditioned medium: implications for IVD regeneration. Eur. Cell Mater., 2016, 32: 163-180

[288]

RajasekaranS, et al.. Proteomic signature of nucleus pulposus in fetal intervertebral disc. Asian Spine J., 2020, 14: 409-420

[289]

RajasekaranS, et al.. Novel biomarkers of health and degeneration in human intervertebral discs: in-depth proteomic analysis of collagen framework of fetal, healthy, scoliotic, degenerate, and herniated discs. Asian Spine J., 2023, 17: 17-29

[290]

CaldeiraJ, et al.. Matrisome profiling during intervertebral disc development and ageing. Sci. Rep., 2017, 7 ArticleID: 11629

[291]

MolinosM, et al.. Alterations of bovine nucleus pulposus cells with aging. Aging Cell, 2023, 22 ArticleID: e13873

[292]

LundbergE, BornerGHH. Spatial proteomics: a powerful discovery tool for cell biology. Nat. Rev. Mol. Cell Biol., 2019, 20: 285-302

[293]

TamV, et al.. DIPPER, a spatiotemporal proteomics atlas of human intervertebral discs for exploring ageing and degeneration dynamics. eLife, 2020, 9: e64940

[294]

EckersleyA, et al.. Peptide location fingerprinting reveals tissue region-specific differences in protein structures in an ageing human organ. Int. J. Mol. Sci., 2021, 22: 10408

[295]

EckersleyA, et al.. Peptide location fingerprinting identifies species- and tissue-conserved structural remodelling of proteins as a consequence of ageing and disease. Matrix Biol., 2022, 114: 108-137

[296]

DaiX, ShenL. Advances and trends in omics technology development. Front. Med., 2022, 9: 911861

[297]

VerasMA, et al.. Protocol for parallel proteomic and metabolomic analysis of mouse intervertebral disc tissues. JOR Spine, 2020, 3 ArticleID: e1099

[298]

ChenQ, et al.. Quiescence preconditioned nucleus pulposus stem cells alleviate intervertebral disc degeneration by enhancing cell survival via adaptive metabolism pattern in rats. Front. Bioeng. Biotechnol., 2023, 11 ArticleID: 1073238

[299]

WuX, et al.. Glycine-serine-threonine metabolic axis delays intervertebral disc degeneration through antioxidant effects: an imaging and metabonomics study. Oxid. Med. Cell. Longev., 2021, 2021 ArticleID: 5579736

[300]

GuoZ, et al.. The role of IL-6 and TMEM100 in lumbar discogenic pain and the mechanism of the glycine-serine-threonine metabolic axis: a metabolomic and molecular biology study. J. Pain. Res., 2023, 16: 437-461

[301]

WymannMP, SchneiterR. Lipid signalling in disease. Nat. Rev. Mol. Cell Biol., 2008, 9: 162-176

[302]

FranciscoV, et al.. Metabolomic signature and molecular profile of normal and degenerated human intervertebral disc cells. Spine J., 2023, 23: 1549-1562

[303]

Granville SmithI, et al.. Evidence for infection in intervertebral disc degeneration: a systematic review. Eur. Spine J., 2022, 31: 414-430

[304]

RajasekaranS, et al.. Bacteria in human lumbar discs - subclinical infection or contamination? Metabolomic evidence for colonization, multiplication, and cell-cell cross-talk of bacteria. Spine J., 2023, 23: 163-177

[305]

BoonenK, et al.. Beyond genes: re-identifiability of proteomic data and its implications for personalized medicine. Genes, 2019, 10: 682

[306]

WilliamsonPM, et al.. Tendinopathy and tendon material response to load: What we can learn from small animal studies. Acta Biomater., 2021, 134: 43-56

[307]

RoffinoS, et al.. Negative impact of disuse and unloading on tendon enthesis structure and function. Life Sci. Space Res., 2021, 29: 46-52

[308]

RielH, LindstromCF, RathleffMS, JensenMB, OlesenJL. Prevalence and incidence rate of lower-extremity tendinopathies in a Danish general practice: a registry-based study. BMC Musculoskelet. Disord., 2019, 20 ArticleID: 239

[309]

LinsellL, et al.. Prevalence and incidence of adults consulting for shoulder conditions in UK primary care; patterns of diagnosis and referral. Rheumatology, 2006, 45: 215-221

[310]

NicholsAEC, BestKT, LoiselleAE. The cellular basis of fibrotic tendon healing: challenges and opportunities. Transl. Res., 2019, 209: 156-168

[311]

KendalAR, et al.. Multi-omic single cell analysis resolves novel stromal cell populations in healthy and diseased human tendon. Sci. Rep., 2020, 10 ArticleID: 13939

[312]

StillC, et al.. Single-cell transcriptomic profiling reveals distinct mechanical responses between normal and diseased tendon progenitor cells. Cell Rep. Med., 2021, 2 ArticleID: 100343

[313]

AkbarM, et al.. Single cell and spatial transcriptomics in human tendon disease indicate dysregulated immune homeostasis. Ann. Rheum. Dis., 2021, 80: 1494-1497

[314]

AckermanJE, et al.. Defining the spatial-molecular map of fibrotic tendon healing and the drivers of Scleraxis-lineage cell fate and function. Cell Rep., 2022, 41 ArticleID: 111706

[315]

Nichols, A. E. C., Wagner, N. W., Ketonis, C. & Loiselle, A. E. Epitenon-derived cells comprise a distinct progenitor population that contributes to both tendon fibrosis and regeneration following acute injury. bioRxiv (2023).

[316]

RajanA, et al.. Single-cell analysis reveals distinct fibroblast plasticity during tenocyte regeneration in zebrafish. Sci. Adv., 2023, 9: eadi5771

[317]

MimpenJY, et al.. Single nucleus and spatial transcriptomic profiling of healthy human hamstring tendon. FASEB J., 2024, 38 ArticleID: e23629

[318]

SteffenD, MienaltowskiM, BaarK. Spatial gene expression in the adult rat patellar tendon. Matrix Biol., 2023, 19-20: 100138

[319]

DisserNP, et al.. Widespread diversity in the transcriptomes of functionally divergent limb tendons. J. Physiol., 2020, 598: 1537-1550

[320]

DisserNP, et al.. Achilles tendons display region-specific transcriptomic signatures associated with distinct mechanical properties. Am. J. Sports Med., 2022, 50: 3866-3874

[321]

BestKT, LoiselleAE. Scleraxis lineage cells contribute to organized bridging tissue during tendon healing and identify a subpopulation of resident tendon cells. FASEB J., 2019, 33: 8578-8587

[322]

KorcariA, PrzybelskiSJ, GingeryA, LoiselleAE. Impact of aging on tendon homeostasis, tendinopathy development, and impaired healing. Connect Tissue Res., 2023, 64: 1-13

[323]

SvenssonRB, HeinemeierKM, CouppeC, KjaerM, MagnussonSP. Effect of aging and exercise on the tendon. J. Appl Physiol., 2016, 121: 1237-1246

[324]

PeffersMJ, et al.. Transcriptome analysis of ageing in uninjured human Achilles tendon. Arthritis Res. Ther., 2015, 17: 33

[325]

ZamboulisDE, et al.. The interfascicular matrix of energy storing tendons houses heterogenous cell populations disproportionately affected by aging. Aging Dis., 2024, 15: 295-310

[326]

PatelD, et al.. Structure-function specialisation of the interfascicular matrix in the human Achilles tendon. Acta Biomater., 2021, 131: 381-390

[327]

SoslowskyLJ, et al.. Neer Award 1999: overuse activity injures the supraspinatus tendon in an animal model: a histologic and biomechanical study. J. Shoulder Elb. Surg., 2000, 9: 79-84

[328]

Malmgaard-ClausenNM, KjaerM, DakinSG. Pathological tendon histology in early and chronic human patellar tendinopathy. Transl. Sports Med., 2022, 2022: 2799665

[329]

GuoJ, et al.. Integrative single-cell RNA and ATAC sequencing reveals that the FOXO1-PRDX2-TNF axis regulates tendinopathy. Front. Immunol., 2023, 14 ArticleID: 1092778

[330]

BestKT, et al.. Scleraxis-lineage cell depletion improves tendon healing and disrupts adult tendon homeostasis. eLife, 2021, 10: e62203

[331]

Howell, K. et al. Novel model of tendon regeneration reveals distinct cell mechanisms underlying regenerative and fibrotic tendon healing. Sci. Rep. 7, 45238 (2017).

[332]

SakabeT, et al.. Transcription factor scleraxis vitally contributes to progenitor lineage direction in wound healing of adult tendon in mice. J. Biol. Chem., 2018, 293: 5766-5780

[333]

WagnerDE, KleinAM. Lineage tracing meets single-cell omics: opportunities and challenges. Nat. Rev. Genet., 2020, 21: 410-427

[334]

ZhangX, et al.. Multi-omics analysis of human tendon adhesion reveals that ACKR1-regulated macrophage migration is involved in regeneration. Bone Res., 2024, 12: 27

[335]

MukundK, SubramaniamS. Skeletal muscle: a review of molecular structure and function, in health and disease. Wiley Interdiscip. Rev. Syst. Biol. Med., 2020, 12: e1462

[336]

MauroA. Satellite cell of skeletal muscle fibers. J. Biophys. Biochem. Cytol., 1961, 9: 493-495

[337]

BentzingerCF, WangYX, DumontNA, RudnickiMA. Cellular dynamics in the muscle satellite cell niche. EMBO Rep., 2013, 14: 1062-1072

[338]

AsakuraA. Stem cells in adult skeletal muscle. Trends Cardiovasc. Med., 2003, 13: 123-128

[339]

McFarlandAJ, RayPR, BhaiS, LevineBD, PriceTJ. RNA sequencing on muscle biopsy from a 5-week bed rest study reveals the effect of exercise and potential interactions with dorsal root ganglion neurons. Physiol. Rep., 2022, 10: e15176

[340]

ZhangX, et al.. Characterization of cellular senescence in aging skeletal muscle. Nat. Aging, 2022, 2: 601-615

[341]

WangL, ZhouY, WangY, ShanT. Integrative cross-species analysis reveals conserved and unique signatures in fatty skeletal muscles. Sci. Data, 2024, 11 ArticleID: 290

[342]

TumasianRA, et al.. Skeletal muscle transcriptome in healthy aging. Nat. Commun., 2021, 12 ArticleID: 2014

[343]

SoutherlandKW, et al.. Skeletal muscle regeneration failure in ischemic-damaged limbs is associated with pro-inflammatory macrophages and premature differentiation of satellite cells. Genome Med., 2023, 15 ArticleID: 95

[344]

PillonNJ, et al.. Transcriptomic profiling of skeletal muscle adaptations to exercise and inactivity. Nat. Commun., 2020, 11 ArticleID: 470

[345]

Nieves-RodriguezS, et al.. Transcriptomic analysis of paired healthy human skeletal muscles to identify modulators of disease severity in DMD. Front. Genet., 2023, 14 ArticleID: 1216066

[346]

CaiC, YueY, YueB. Single-cell RNA sequencing in skeletal muscle developmental biology. Biomed. Pharmacother., 2023, 162: 114631

[347]

De MicheliAJ, SpectorJA, ElementoO, CosgroveBD. A reference single-cell transcriptomic atlas of human skeletal muscle tissue reveals bifurcated muscle stem cell populations. Skelet. Muscle, 2020, 10 ArticleID: 19

[348]

GiordaniL, et al.. High-dimensional single-cell cartography reveals novel skeletal muscle-resident cell populations. Mol. Cell, 2019, 74: 609-621.e606

[349]

ChoDS, DolesJD. Single cell transcriptome analysis of muscle satellite cells reveals widespread transcriptional heterogeneity. Gene, 2017, 636: 54-63

[350]

Dell’OrsoS, et al.. Single cell analysis of adult mouse skeletal muscle stem cells in homeostatic and regenerative conditions. Development, 2019, 146: dev174177

[351]

BarruetE, et al.. Functionally heterogeneous human satellite cells identified by single cell RNA sequencing. eLife, 2020, 9: e51576

[352]

XuD, et al.. Single-cell RNA-sequencing provides insight into skeletal muscle evolution during the selection of muscle characteristics. Adv. Sci., 2023, 10, ArticleID: e2305080

[353]

De MicheliAJ, et al.. Single-cell analysis of the muscle stem cell hierarchy identifies heterotypic communication signals involved in skeletal muscle regeneration. Cell Rep., 2020, 30: 3583-3595.e3585

[354]

QinT, et al.. Single-cell RNA-seq reveals novel mitochondria-related musculoskeletal cell populations during adult axolotl limb regeneration process. Cell Death Differ., 2021, 28: 1110-1125

[355]

UapinyoyingP, et al.. Single-cell transcriptomic analysis of the identity and function of fibro/adipogenic progenitors in healthy and dystrophic muscle. iScience, 2023, 26 ArticleID: 107479

[356]

FitzgeraldG, et al.. MME+ fibro-adipogenic progenitors are the dominant adipogenic population during fatty infiltration in human skeletal muscle. Commun. Biol., 2023, 6: 111

[357]

NegroniE, et al.. Muscle fibro-adipogenic progenitors from a single-cell perspective: focus on their “virtual” secretome. Front. Cell Dev. Biol., 2022, 10 ArticleID: 952041

[358]

OprescuSN, YueF, QiuJ, BritoLF, KuangS. Temporal dynamics and heterogeneity of cell populations during skeletal muscle regeneration. iScience, 2020, 23: 100993

[359]

MaL, et al.. Single-cell RNA-seq reveals novel interaction between muscle satellite cells and fibro-adipogenic progenitors mediated with FGF7 signalling. J. Cachexia Sarcopenia Muscle, 2024, 15: 1388-1403

[360]

McKellarDW, et al.. Large-scale integration of single-cell transcriptomic data captures transitional progenitor states in mouse skeletal muscle regeneration. Commun. Biol., 2021, 4: 1280

[361]

KedlianVR, et al.. Human skeletal muscle aging atlas. Nat. Aging, 2024, 4: 727-744

[362]

VoisinS, et al.. Meta-analysis of genome-wide DNA methylation and integrative omics of age in human skeletal muscle. J. Cachexia Sarcopenia Muscle, 2021, 12: 1064-1078

[363]

Dos SantosM, et al.. Opposing gene regulatory programs governing myofiber development and maturation revealed at single nucleus resolution. Nat. Commun., 2023, 14 ArticleID: 4333

[364]

KimmelJC, HwangAB, ScaramozzaA, MarshallWF, BrackAS. Aging induces aberrant state transition kinetics in murine muscle stem cells. Development, 2020, 147: dev183855

[365]

KurlandJV, et al.. Aging disrupts gene expression timing during muscle regeneration. Stem Cell Rep., 2023, 18: 1325-1339

[366]

KrasniewskiLK, et al.. Single-cell analysis of skeletal muscle macrophages reveals age-associated functional subpopulations. eLife, 2022, 11: e77974

[367]

CameronA, et al.. Identification of underexplored mesenchymal and vascular-related cell populations in human skeletal muscle. Am. J. Physiol. Cell Physiol., 2022, 323: C1586-C1600

[368]

ProiettiD, et al.. Activation of skeletal muscle-resident glial cells upon nerve injury. JCI Insight, 2021, 6: e143469

[369]

BarthelsonRA, LambertGM, VanierC, LynchRM, GalbraithDW. Comparison of the contributions of the nuclear and cytoplasmic compartments to global gene expression in human cells. BMC Genom., 2007, 8, ArticleID: 340

[370]

PetranyMJ, et al.. Single-nucleus RNA-seq identifies transcriptional heterogeneity in multinucleated skeletal myofibers. Nat. Commun., 2020, 11 ArticleID: 6374

[371]

KimM, et al.. Single-nucleus transcriptomics reveals functional compartmentalization in syncytial skeletal muscle cells. Nat. Commun., 2020, 11 ArticleID: 6375

[372]

Dos SantosM, et al.. Single-nucleus RNA-seq and FISH identify coordinated transcriptional activity in mammalian myofibers. Nat. Commun., 2020, 11 ArticleID: 5102

[373]

ChauJ, et al.. Relationship of DUX4 and target gene expression in FSHD myocytes. Hum. Mutat., 2021, 42: 421-433

[374]

SolovyevaEM, et al.. New insights into molecular changes in skeletal muscle aging and disease: differential alternative splicing and senescence. Mech. Ageing Dev., 2021, 197 ArticleID: 111510

[375]

KannAP, KraussRS. Multiplexed RNAscope and immunofluorescence on whole-mount skeletal myofibers and their associated stem cells. Development, 2019, 146: dev179259

[376]

YoungLV, et al.. Muscle injury induces a transient senescence-like state that is required for myofiber growth during muscle regeneration. FASEB J., 2022, 36 ArticleID: e22587

[377]

StecMJ, et al.. A cellular and molecular spatial atlas of dystrophic muscle. Proc. Natl. Acad. Sci. USA, 2023, 120 ArticleID: e2221249120

[378]

CoulisG, et al.. Single-cell and spatial transcriptomics identify a macrophage population associated with skeletal muscle fibrosis. Sci. Adv., 2023, 9 ArticleID: eadd9984

[379]

RuossS, et al.. Spatial transcriptomics tools allow for regional exploration of heterogeneous muscle pathology in the pre-clinical rabbit model of rotator cuff tear. J. Orthop. Surg. Res., 2022, 17: 440

[380]

WilliamsK, YokomoriK, MortazaviA. Heterogeneous skeletal muscle cell and nucleus populations identified by single-cell and single-nucleus resolution transcriptome assays. Front. Genet., 2022, 13: 835099

[381]

CristCG, MontarrasD, BuckinghamM. Muscle satellite cells are primed for myogenesis but maintain quiescence with sequestration of Myf5 mRNA targeted by microRNA-31 in mRNP granules. Cell Stem Cell, 2012, 11: 118-126

[382]

HostrupM, et al.. High-intensity interval training remodels the proteome and acetylome of human skeletal muscle. eLife, 2022, 11: e69802

[383]

MuchaO, et al.. Proteome profiling of the dystrophic. Biomolecules, 2023, 13: 1648

[384]

SzczerbinskiL, et al.. Metabolomic profile of skeletal muscle and its change under a mixed-mode exercise intervention in progressively dysglycemic subjects. Front. Endocrinol., 2021, 12, ArticleID: 778442

[385]

DeshmukhAS, et al.. Deep muscle-proteomic analysis of freeze-dried human muscle biopsies reveals fiber type-specific adaptations to exercise training. Nat. Commun., 2021, 12 ArticleID: 304

[386]

HuntLC, et al.. Integrated genomic and proteomic analyses identify stimulus-dependent molecular changes associated with distinct modes of skeletal muscle atrophy. Cell Rep., 2021, 37 ArticleID: 109971

[387]

Ubaida-MohienC, et al.. Discovery proteomics in aging human skeletal muscle finds change in spliceosome, immunity, proteostasis and mitochondria. eLife, 2019, 8: e49874

[388]

KallabisS, et al.. High-throughput proteomics fiber typing (ProFiT) for comprehensive characterization of single skeletal muscle fibers. Skelet. Muscle, 2020, 10 ArticleID: 7

[389]

MelbyJA, et al.. High sensitivity top-down proteomics captures single muscle cell heterogeneity in large proteoforms. Proc. Natl. Acad. Sci. USA, 2023, 120 ArticleID: e2222081120

[390]

MurgiaM, et al.. Protein profile of fiber types in human skeletal muscle: a single-fiber proteomics study. Skelet. Muscle, 2021, 11 ArticleID: 24

[391]

MurgiaM, et al.. Single muscle fiber proteomics reveals unexpected mitochondrial specialization. EMBO Rep., 2015, 16: 387-395

[392]

BorokM, et al.. Progressive and coordinated mobilization of the skeletal muscle niche throughout tissue repair revealed by single-cell proteomic analysis. Cells, 2021, 10: 744

[393]

MaerkensA, et al.. New insights into the protein aggregation pathology in myotilinopathy by combined proteomic and immunolocalization analyses. Acta Neuropathol. Commun., 2016, 4: 8

[394]

OlieCS, et al.. The metabolic landscape in chronic rotator cuff tear reveals tissue-region-specific signatures. J. Cachexia Sarcopenia Muscle, 2022, 13: 532-543

[395]

DowlingP, SwandullaD, OhlendieckK. Mass spectrometry-based proteomic technology and its application to study skeletal muscle cell biology. Cells, 2023, 12: 2560

Funding

U.S. Department of Health & Human Services | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)(AR083900)

P30 center grants (AR074992 and AR073752)

T32 grant HD007434 to DRK

DoD grants HT94252310534 to RJT

DoD grant HT94252310519 to CMK

RIGHTS & PERMISSIONS

The Author(s)

AI Summary AI Mindmap
PDF

188

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/