Advances in spatial transcriptomics and its application in the musculoskeletal system

Haoyu Wang , Peng Cheng , Juan Wang , Hongzhi Lv , Jie Han , Zhiyong Hou , Ren Xu , Wei Chen

Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 54

PDF
Bone Research ›› 2025, Vol. 13 ›› Issue (1) : 54 DOI: 10.1038/s41413-025-00429-w
Review Article

Advances in spatial transcriptomics and its application in the musculoskeletal system

Author information +
History +
PDF

Abstract

While bulk RNA sequencing and single-cell RNA sequencing have shed light on cellular heterogeneity and potential molecular mechanisms in the musculoskeletal system in both physiological and various pathological states, the spatial localization of cells and molecules and intercellular interactions within the tissue context require further elucidation. Spatial transcriptomics has revolutionized biological research by simultaneously capturing gene expression profiles and in situ spatial information of tissues, gradually finding applications in musculoskeletal research. This review provides a summary of recent advances in spatial transcriptomics and its application to the musculoskeletal system. The classification and characteristics of data acquisition techniques in spatial transcriptomics are briefly outlined, with an emphasis on widely-adopted representative technologies and the latest technological breakthroughs, accompanied by a concise workflow for incorporating spatial transcriptomics into musculoskeletal system research. The role of spatial transcriptomics in revealing physiological mechanisms of the musculoskeletal system, particularly during developmental processes, is thoroughly summarized. Furthermore, recent discoveries and achievements of this emerging omics tool in addressing inflammatory, traumatic, degenerative, and tumorous diseases of the musculoskeletal system are compiled. Finally, challenges and potential future directions for spatial transcriptomics, both as a field and in its applications in the musculoskeletal system, are discussed.

Keywords

Biological Sciences / Genetics

Cite this article

Download citation ▾
Haoyu Wang, Peng Cheng, Juan Wang, Hongzhi Lv, Jie Han, Zhiyong Hou, Ren Xu, Wei Chen. Advances in spatial transcriptomics and its application in the musculoskeletal system. Bone Research, 2025, 13(1): 54 DOI:10.1038/s41413-025-00429-w

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

MarguliesM, et al. . Genome sequencing in open microfabricated high density picoliter reactors. Nature, 2005, 437: 376-380.

[2]

WangZ, GersteinM, SnyderM. RNA-Seq: a revolutionary tool for transcriptomics. Nat. Rev. Genet., 2009, 10: 57-63.

[3]

StarkR, GrzelakM, HadfieldJ. RNA sequencing: the teenage years. Nat. Rev. Genet., 2019, 20: 631-656.

[4]

LiX, WangC-Y. From bulk, single-cell to spatial RNA sequencing. Int. J. Oral. Sci., 2021, 13: 36.

[5]

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

[6]

SalibaAE, WestermannAJ, GorskiSA, VogelJ. Single-cell RNA-seq: advances and future challenges. Nucleic Acids Res., 2014, 42: 8845-8860.

[7]

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

[8]

HwangB, LeeJH, BangD. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp. Mol. Med, 2018, 50: 1-14.

[9]

RaoA, BarkleyD, FrancaGS, YanaiI. Exploring tissue architecture using spatial transcriptomics. Nature, 2021, 596: 211-220.

[10]

VillasenorS, GrinsteinM. Spatial high-resolution analysis of gene expression levels in tendons. J. Vis. Exp., 2024.

[11]

KurenkovaAD, MedvedevaEV, NewtonPT, ChaginAS. Niches for skeletal stem cells of mesenchymal origin. Front. Cell Dev. Biol., 2020, 8: 592.

[12]

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

[13]

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

[14]

ZhangB, et al. . A human embryonic limb cell atlas resolved in space and time. Nature, 2023, 635: 668-678.

[15]

BandyopadhyayS, et al. . Mapping the cellular biogeography of human bone marrow niches using single-cell transcriptomics and proteomic imaging. Cell, 2024, 187: 3120-3140.

[16]

GreenblattMB, et al. . The unmixing problem: a guide to applying single-cell RNA sequencing to bone. J. Bone Miner. Res., 2019, 34: 1207-1219.

[17]

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.

[18]

HataK, et al. . Serinc5 regulates sequential chondrocyte differentiation by inhibiting Sox9 function in pre-hypertrophic chondrocytes. J. Cell. Physiol., 2024, 240: e31490.

[19]

HoltCE, BullockSL. Subcellular mRNA localization in animal cells and why it matters. Science, 2009, 326: 1212-1216.

[20]

LiaoJ, et al. . Uncovering an organ’s molecular architecture at single-cell resolution by spatially resolved transcriptomics. Trends Biotechnol., 2021, 39: 43-58.

[21]

MarxV. Method of the year: spatially resolved transcriptomics. Nat. Methods, 2021, 18: 9-14.

[22]

AlonS, et al. . Expansion sequencing: spatially precise in situ transcriptomics in intact biological systems. Science, 2021, 371: eaax2656.

[23]

JunkerJP, et al. . Genome-wide RNA tomography in the zebrafish embryo. Cell, 2014, 159: 662-675.

[24]

ChenJ, et al. . Spatial transcriptomic analysis of cryosectioned tissue samples with Geo-seq. Nat. Protoc., 2017, 12: 566-580.

[25]

AspM, et al. . A spatiotemporal organ-wide gene expression and cell atlas of the developing human heart. Cell, 2019, 179: 1647-1660.e1619.

[26]

Fawkner-CorbettD, et al. . Spatiotemporal analysis of human intestinal development at single-cell resolution. Cell, 2021, 184: 810-826.e823.

[27]

CalvaneseV, et al. . Mapping human haematopoietic stem cells from haemogenic endothelium to birth. Nature, 2022, 604: 534-540.

[28]

ChenA, et al. . Spatiotemporal transcriptomic atlas of mouse organogenesis using DNA nanoball-patterned arrays. Cell, 2022, 185: 1777-1792.e1721.

[29]

FarahEN, et al. . Spatially organized cellular communities form the developing human heart. Nature, 2024, 627: 854-864.

[30]

Galeano NiñoJL, et al. . Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature, 2022, 611: 810-817.

[31]

AroraR, et al. . Spatial transcriptomics reveals distinct and conserved tumor core and edge architectures that predict survival and targeted therapy response. Nat. Commun., 2023, 14: 5029.

[32]

LiuS, et al. . Spatial maps of T cell receptors and transcriptomes reveal distinct immune niches and interactions in the adaptive immune response. Immunity, 2022, 55: 1940-1952.e1945.

[33]

KrausgruberT, et al. . Single-cell and spatial transcriptomics reveal aberrant lymphoid developmental programs driving granuloma formation. Immunity, 2023, 56: 289-306.e287.

[34]

ChenW-T, et al. . Spatial transcriptomics and in situ sequencing to study Alzheimer’s disease. Cell, 2020, 182: 976-991.e919.

[35]

ZhengP, et al. . Integrated spatial transcriptome and metabolism study reveals metabolic heterogeneity in human injured brain. Cell Rep. Med., 2023, 4: 101057.

[36]

FengS, et al. . Application of single-cell and spatial omics in musculoskeletal disorder research. Int. J. Mol. Sci., 2023, 24: 2271.

[37]

AspM, BergenstråhleJ, LundebergJ. Spatially resolved transcriptomes—next generation tools for tissue exploration. BioEssays, 2020, 42: e1900221.

[38]

MosesL, PachterL. Museum of spatial transcriptomics. Nat. Methods, 2022, 19: 534-546.

[39]

WilliamsCG, et al. . An introduction to spatial transcriptomics for biomedical research. Genome Med., 2022, 14: 68.

[40]

Robles-RemachoA, Sanchez-MartinRM, Diaz-MochonJJ. Spatial transcriptomics: emerging technologies in tissue gene expression profiling. Anal. Chem., 2023, 95: 15450-15460.

[41]

YueL, et al. . A guidebook of spatial transcriptomic technologies, data resources and analysis approaches. Comput. Struct. Biotechnol. J., 2023, 21: 940-955.

[42]

GALLJG, PARDUEML. Formation and detection of RNA-DNA hybrid molecules in cytological preparations. Proc. Natl. Acad. Sci. USA, 1969, 63: 378-383.

[43]

JohnHA, BirnstielML, JonesKW. RNA-DNA hybrids at the cytological level. Nature, 1969, 223: 582-587.

[44]

FeminoAM, FayFS, FogartyK, SingerRH. Visualization of single RNA transcripts in Situ. Science, 1998, 280: 585-590.

[45]

RajA, et al. . Imaging individual mRNA molecules using multiple singly labeled probes. Nat. Methods, 2008, 5: 877-879.

[46]

WangF, et al. . RNAscope. J. Mol. Diagn., 2012, 14: 22-29.

[47]

LubeckE, et al. . Single-cell in situ RNA profiling by sequential hybridization. Nat. Methods, 2014, 11: 360-361.

[48]

EngC-HL, et al. . Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH+. Nature, 2019, 568: 235-239.

[49]

ChenKH, et al. . RNA imaging Spatially resolved, highly multiplexed RNA profiling in single cells. Science, 2015, 348: aaa6090.

[50]

BormLE, et al. . Scalable in situ single-cell profiling by electrophoretic capture of mRNA using EEL FISH. Nat. Biotechnol., 2022, 41: 222-231

[51]

HeS, et al. . High-plex imaging of RNA and proteins at subcellular resolution in fixed tissue by spatial molecular imaging. Nat. Biotechnol., 2022, 40: 1794-1806.

[52]

KeR, et al. . In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods, 2013, 10: 857-860.

[53]

JanesickA, et al. . High resolution mapping of the tumor microenvironment using integrated single-cell, spatial and in situ analysis. Nat. Commun., 2023, 14: 8353.

[54]

WangX, et al. . Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science, 2018, 361: eaat5691.

[55]

LiQ, et al. . Multimodal charting of molecular and functional cell states via in situ electro-sequencing. Cell, 2023, 186: 2002-2017.e2021.

[56]

LeeJH, et al. . Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nat. Protoc., 2015, 10: 442-458.

[57]

LarssonC, GrundbergI, SöderbergO, NilssonM. In situ detection and genotyping of individual mRNA molecules. Nat. Methods, 2010, 7: 395-397.

[58]

JainS, EadonMT. Spatial transcriptomics in health and disease. Nat. Rev. Nephrol., 2024, 20: 659-671.

[59]

MegasS, et al. . Spatial transcriptomics of the respiratory system. Annu. Rev. Physiol., 2024, 87: 447-470

[60]

Emmert-BuckMR, et al. . Laser capture microdissection. Science, 1996, 274: 998-1001.

[61]

MerrittCR, et al. . Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat. Biotechnol., 2020, 38: 586-599.

[62]

Gracia VillacampaE, et al. . Genome-wide spatial expression profiling in formalin-fixed tissues. Cell Genom., 2021, 1: 100065.

[63]

LebrigandK, et al. . The spatial landscape of gene expression isoforms in tissue sections. Nucleic Acids Res., 2023, 51: e47.

[64]

MirzazadehR, et al. . Spatially resolved transcriptomic profiling of degraded and challenging fresh frozen samples. Nat. Commun., 2023, 14: 509.

[65]

McKellarDW, et al. . Spatial mapping of the total transcriptome by in situ polyadenylation. Nat. Biotechnol., 2022, 41: 513-520.

[66]

RodriquesSG, et al. . Slide-seq: a scalable technology for measuring genome-wide expression at high spatial resolution. Science, 2019, 363: 1463-1467.

[67]

StickelsRR, et al. . Highly sensitive spatial transcriptomics at near-cellular resolution with Slide-seqV2. Nat. Biotechnol., 2021, 39: 313-319.

[68]

LiuY, et al. . High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell, 2020, 183: 1665-1681.e1618.

[69]

WirthJ, et al. . Spatial transcriptomics using multiplexed deterministic barcoding in tissue. Nat. Commun., 2023, 14: 1523.

[70]

LeeY, et al. . XYZeq: Spatially resolved single-cell RNA sequencing reveals expression heterogeneity in the tumor microenvironment. Sci. Adv., 2021, 7: eabg4755.

[71]

SrivatsanSR, et al. . Embryo-scale, single-cell spatial transcriptomics. Science, 2021, 373: 111-117.

[72]

PangJB, et al. . Spatial transcriptomics and the anatomical pathologist: Molecular meets morphology. Histopathology, 2024, 84: 577-586.

[73]

PentimalliTM, KaraiskosN, RajewskyN. Challenges and opportunities in the clinical translation of high-resolution spatial transcriptomics. Annu. Rev. Pathol., 2024, 20: 405-432.

[74]

ValihrachL, ZuchaD, AbaffyP, KubistaM. A practical guide to spatial transcriptomics. Mol. Asp. Med., 2024, 97: 101276.

[75]

PiñaJO, et al. . Multimodal spatiotemporal transcriptomic resolution of embryonic palate osteogenesis. Nat. Commun., 2023, 14: 5687.

[76]

GribaudoS, et al. . Self-organizing models of human trunk organogenesis recapitulate spinal cord and spine co-morphogenesis. Nat. Biotechnol., 2023, 42: 1243-1253.

[77]

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

[78]

ZhangT, et al. . Single-cell RNA sequencing reveals cellular and molecular heterogeneity in fibrocartilaginous enthesis formation. eLife, 2023, 12: e85873.

[79]

LuiJC, et al. . Spatial regulation of gene expression during growth of articular cartilage in juvenile mice. Pediatr. Res., 2014, 77: 406-415.

[80]

MarcuKB, et al. . Gene expression profiling reveals similarities between the spatial architectures of postnatal articular and growth plate cartilage. PLoS One, 2014, 9: e103061.

[81]

BianF, et al. . The G protein-coupled receptor ADGRG6 maintains mouse growth plate homeostasis through IHH signaling. J. Bone Miner. Res., 2024, 39: 1644-1658.

[82]

TongW, et al. . Periarticular mesenchymal progenitors initiate and contribute to secondary ossification center formation during mouse long bone development. Stem Cells, 2019, 37: 677-689.

[83]

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

[84]

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.

[85]

D’ErcoleC, et al. . Spatially resolved transcriptomics reveals innervation-responsive functional clusters in skeletal muscle. Cell Rep., 2022, 41: 111861.

[86]

KarlsenA, et al. . Distinct myofibre domains of the human myotendinous junction revealed by single nucleus RNA-seq. J. Cell Sci., 2023, 136: jcs260913.

[87]

SteffenD, MienaltowskiM, BaarK. Spatial gene expression in the adult rat patellar tendon. Matrix Biol., 2023.

[88]

FinckhA, et al. . Global epidemiology of rheumatoid arthritis. Nat. Rev. Rheumatol., 2022, 18: 591-602

[89]

Di MatteoA, BathonJM, EmeryP. Rheumatoid arthritis. Lancet, 2023, 402: 2019-2033.

[90]

HuangJ, et al. . Promising therapeutic targets for treatment of rheumatoid arthritis. Front. Immunol., 2021, 12: 686155.

[91]

GravalleseEM, LongoDL, FiresteinGS. Rheumatoid arthritis—common origins, divergent mechanisms. N. Engl. J. Med., 2023, 388: 529-542.

[92]

VickovicS, et al. . Three-dimensional spatial transcriptomics uncovers cell type localizations in the human rheumatoid arthritis synovium. Commun. Biol., 2022, 5: 129.

[93]

MengX, et al. . Role and therapeutic potential for targeting fibroblast growth factor 10/FGFR1 in relapsed rheumatoid arthritis. Arthritis Rheumatol., 2023, 76: 32-47.

[94]

SmithMH, et al. . Drivers of heterogeneity in synovial fibroblasts in rheumatoid arthritis. Nat. Immunol., 2023, 24: 1200-1210.

[95]

RauberS, et al. . CD200+ fibroblasts form a pro-resolving mesenchymal network in arthritis. Nat. Immunol., 2024, 25: 682-692.

[96]

ZhengL, et al. . ITGA5+ synovial fibroblasts orchestrate proinflammatory niche formation by remodelling the local immune microenvironment in rheumatoid arthritis. Ann. Rheum. Dis., 2024, 84: 232-252.

[97]

CarlbergK, et al. . Exploring inflammatory signatures in arthritic joint biopsies with Spatial Transcriptomics. Sci. Rep., 2019, 9: 18975.

[98]

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

[99]

MacDonaldL, et al. . Synovial tissue myeloid dendritic cell subsets exhibit distinct tissue-niche localization and function in health and rheumatoid arthritis. Immunity, 2024, 57: 2843-2862.

[100]

KenneyHM, et al. . Multi-omics analysis identifies IgG2b class-switching with ALCAM-CD6 co-stimulation in joint-draining lymph nodes during advanced inflammatory-erosive arthritis. Front. Immunol., 2023, 14: 1237498.

[101]

DavidsonS, et al. . Fibroblasts as immune regulators in infection, inflammation and cancer. Nat. Rev. Immunol., 2021, 21: 704-717.

[102]

FriščićJ, et al. . The complement system drives local inflammatory tissue priming by metabolic reprogramming of synovial fibroblasts. Immunity, 2021, 54: 1002-1021.e1010.

[103]

BeryAI, et al. . Role of tertiary lymphoid organs in the regulation of immune responses in the periphery. Cell. Mol. Life Sci., 2022, 79: 359.

[104]

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

[105]

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.

[106]

McInnesIB, SchettG. The pathogenesis of rheumatoid arthritis. N. Engl. J. Med., 2011, 365: 2205-2219.

[107]

MessererDAC, et al. . Immunopathophysiology of trauma-related acute kidney injury. Nat. Rev. Nephrol., 2020, 17: 91-111.

[108]

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

[109]

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.

[110]

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

[111]

LaroucheJA, et al. . Spatiotemporal mapping of immune and stem cell dysregulation after volumetric muscle loss. JCI Insight, 2023, 8: e162835.

[112]

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: 111706.

[113]

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: eade4619.

[114]

KangH, et al. . The HIF-1alpha/PLOD2 axis integrates extracellular matrix organization and cell metabolism leading to aberrant musculoskeletal repair. Bone Res, 2024, 12: 17.

[115]

MokGF, Lozano-VelascoE, MunsterbergA. microRNAs in skeletal muscle development. Semin. Cell Dev. Biol., 2017, 72: 67-76.

[116]

GiagnorioE, et al. . MyomiRs and their multifaceted regulatory roles in muscle homeostasis and amyotrophic lateral sclerosis. J. Cell Sci., 2021, 134: jcs258349.

[117]

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

[118]

WanZ, et al. . Mgp high‐expressing MSCs orchestrate the osteoimmune microenvironment of collagen/nanohydroxyapatite‐mediated bone regeneration. Adv. Sci., 2024, 11: e2308986.

[119]

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

[120]

RiosJJ, et al. . Spatial transcriptomics implicates impaired BMP signaling in NF1 fracture pseudarthrosis in murine and patient tissues. JCI Insight, 2024, 9: e176802

[121]

MathavanN, et al. . Spatial transcriptomics in bone mechanomics: Exploring the mechanoregulation of fracture healing in the era of spatial omics. Sci. Adv., 2025, 11: eadp8496.

[122]

PenaOA, MartinP. Cellular and molecular mechanisms of skin wound healing. Nat. Rev. Mol. Cell Biol., 2024, 25: 599-616.

[123]

FosterDS, et al. . Integrated spatial multiomics reveals fibroblast fate during tissue repair. Proc. Natl. Acad. Sci. USA, 2021, 118: e2110025118.

[124]

YangY, et al. . Tracing immune cells around biomaterials with spatial anchors during large-scale wound regeneration. Nat. Commun., 2023, 14: 5995.

[125]

ChenP, et al. . Single‐cell and spatial transcriptomics decodes Wharton’s jelly‐derived mesenchymal stem cells heterogeneity and a subpopulation with wound repair signatures. Adv. Sci., 2022, 10: e2204786.

[126]

XiaX, et al. . Mesenchymal stem cells promote healing of nonsteroidal anti-inflammatory drug-related peptic ulcer through paracrine actions in pigs. Sci. Transl. Med., 2019, 11: eaat7455.

[127]

LevyO, et al. . Shattering barriers toward clinically meaningful MSC therapies. Sci. Adv., 2020, 6: eaba6884.

[128]

ZhouT, et al. . Challenges and advances in clinical applications of mesenchymal stromal cells. J. Hematol. Oncol., 2021, 14: 24.

[129]

Elosua-BayesM, et al. . SPOTlight: seeded NMF regression to deconvolute spatial transcriptomics spots with single-cell transcriptomes. Nucleic Acids Res., 2021, 49: e50.

[130]

Glyn-JonesS, et al. . Osteoarthritis. Lancet, 2015, 386: 376-387.

[131]

HongJW, NohJH, KimDJ. The prevalence of and demographic factors associated with radiographic knee osteoarthritis in Korean adults aged ≥ 50 years: the 2010-2013 Korea National Health and Nutrition Examination Survey. PLoS One, 2020, 15: e0230613.

[132]

HunterDJ, Bierma-ZeinstraS. Osteoarthritis. Lancet, 2019, 393: 1745-1759.

[133]

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

[134]

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.

[135]

LoeserRF. Age-related changes in the musculoskeletal system and the development of osteoarthritis. Clin. Geriatr. Med., 2010, 26: 371-386.

[136]

ThompsonS, et al. . Twenty-year outcomes of a longitudinal prospective evaluation of isolated endoscopic anterior cruciate ligament reconstruction with patellar tendon autografts. Am. J. Sports Med., 2015, 43: 2164-2174.

[137]

YangR, et al. . A single-cell atlas depicting the cellular and molecular features in human anterior cruciate ligamental degeneration: A single cell combined spatial transcriptomics study. eLife, 2023, 12: e85700.

[138]

PerezK, et al. . Single nuclei profiling identifies cell specific markers of skeletal muscle aging, frailty, and senescence. Aging, 2022, 14: 9393-9422

[139]

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

[140]

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

[141]

MaffulliN, LongoUG, KadakiaA, SpieziaF. Achilles tendinopathy. Foot Ankle Surg., 2020, 26: 240-249.

[142]

MillarNL, et al. . Tendinopathy. Nat. Rev. Dis. Prim., 2021, 7: 1.

[143]

WalshLA, QuailDF. Decoding the tumor microenvironment with spatial technologies. Nat. Immunol., 2023, 24: 1982-1993.

[144]

LiuT, et al. . Cancer-associated fibroblasts: an emerging target of anti-cancer immunotherapy. J. Hematol. Oncol., 2019, 12: 86.

[145]

SahaiE, et al. . A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer, 2020, 20: 174-186.

[146]

GillCM, FowkesM, ShrivastavaRK. Emerging therapeutic targets in chordomas: a review of the literature in the genomic era. Neurosurgery, 2020, 86: E118-E123.

[147]

Al ShihabiA, et al. . Personalized chordoma organoids for drug discovery studies. Sci. Adv., 2022, 8: eabl3674.

[148]

ZhangT-L, et al. . Integrating single-cell and spatial transcriptomics reveals endoplasmic reticulum stress-related CAF subpopulations associated with chordoma progression. Neuro Oncol., 2024, 26: 295-308.

[149]

RiggiN, LongoDL, SuvàML, StamenkovicI. Ewing’s Sarcoma. N. Engl. J. Med., 2021, 384: 154-164.

[150]

WrennED, et al. . Cancer-associated fibroblast-like tumor cells remodel the Ewing sarcoma tumor microenvironment. Clin. Cancer Res., 2023, 29: 5140-5154.

[151]

LeiX, et al. . Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett., 2020, 470: 126-133.

[152]

LiR, et al. . PSME2 offers value as a biomarker of M1 macrophage infiltration in pan-cancer and inhibits osteosarcoma malignant phenotypes. Int. J. Biol. Sci., 2024, 20: 1452-1470.

[153]

PingpingB, et al. . Incidence and mortality of sarcomas in Shanghai, China, during 2002–2014. Front. Oncol., 2019, 9: 662.

[154]

SiegelRL, MillerKD, JemalA. Cancer statistics, 2019. CA Cancer J. Clin., 2019, 69: 7-34.

[155]

WeilbaecherKN, GuiseTA, McCauleyLK. Cancer to bone: a fatal attraction. Nat. Rev. Cancer, 2011, 11: 411-425.

[156]

IhleCL, et al. . Distinct tumor microenvironments of lytic and blastic bone metastases in prostate cancer patients. J. Immunother. Cancer, 2019, 7: 293.

[157]

HuangR, et al. . Construction of bone metastasis-specific regulation network based on prognostic stemness-related signatures in breast invasive carcinoma. Front. Oncol., 2021, 10: 613333.

[158]

PaganiS, et al. . RNA extraction from cartilage: issues, methods, tips. Int. J. Mol. Sci., 2023, 24: 2120.

[159]

SailerV, et al. . Bone biopsy protocol for advanced prostate cancer in the era of precision medicine. Cancer, 2018, 124: 1008-1015.

[160]

SinghVM, et al. . Analysis of the effect of various decalcification agents on the quantity and quality of nucleic acid (DNA and RNA) recovered from bone biopsies. Ann. Diagn. Pathol., 2013, 17: 322-326.

[161]

ZhengG, et al. . Clinical mutational profiling of bone metastases of lung and colon carcinoma and malignant melanoma using next-generation sequencing. Cancer Cytopathol., 2016, 124: 744-753.

[162]

VincentTL, McClurgO, TroebergL. The extracellular matrix of articular cartilage controls the bioavailability of pericellular matrix-bound growth factors to drive tissue homeostasis and repair. Int. J. Mol. Sci., 2022, 23: 6003.

[163]

RuettgerA, NeumannS, WiederandersB, HuberR. Comparison of different methods for preparation and characterization of total RNA from cartilage samples to uncover osteoarthritis in vivo. BMC Res. Notes, 2010, 3: 7.

[164]

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

[165]

GaoXH, et al. . Comparison of fresh frozen tissue with formalin-fixed paraffin-embedded tissue for mutation analysis using a multi-gene panel in patients with colorectal cancer. Front. Oncol., 2020, 10: 310.

[166]

LinX, et al. . A comparative analysis of RNA sequencing methods with ribosome RNA depletion for degraded and low-input total RNA from formalin-fixed and paraffin-embedded samples. BMC Genomics, 2019, 20: 831.

[167]

ZhongJ, et al. . Multi-species atlas resolves an axolotl limb development and regeneration paradox. Nat. Commun., 2023, 14: 6346.

[168]

GomarizA, et al. . Quantitative spatial analysis of haematopoiesis-regulating stromal cells in the bone marrow microenvironment by 3D microscopy. Nat. Commun., 2018, 9: 2532.

[169]

BoulaisPE, et al. . The majority of CD45(-) Ter119(-) CD31(-) bone marrow cell fraction is of hematopoietic origin and contains erythroid and lymphoid progenitors. Immunity, 2018, 49: 627-639.

[170]

LiH, et al. . A comprehensive benchmarking with practical guidelines for cellular deconvolution of spatial transcriptomics. Nat. Commun., 2023, 14: 1548.

[171]

GulatiGS, et al. . Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat. Rev. Mol. Cell Biol., 2024, 26: 11-31.

[172]

ChenJY, et al. . Hoxb5 marks long-term haematopoietic stem cells and reveals a homogenous perivascular niche. Nature, 2016, 530: 223-227.

[173]

ZiegenhainC, et al. . Comparative analysis of single-cell RNA sequencing methods. Mol. Cell, 2017, 65: 631-643.

[174]

WegmannR, et al. . CellSIUS provides sensitive and specific detection of rare cell populations from complex single-cell RNA-seq data. Genome Biol., 2019, 20: 142.

[175]

SongD, et al. . scPNMF: sparse gene encoding of single cells to facilitate gene selection for targeted gene profiling. Bioinformatics, 2021, 37: i358-i366.

[176]

ChengC, ChenW, JinH, ChenX. A review of single-cell RNA-seq annotation, integration, and cell–cell communication. Cells, 2023, 12: 1970.

[177]

WangX, et al. . MarsGT: Multi-omics analysis for rare population inference using single-cell graph transformer. Nat. Commun., 2024.

[178]

HaoY, et al. . Integrated analysis of multimodal single-cell data. Cell, 2021, 184: 3573-3587.

[179]

WeiR, et al. . Spatial charting of single-cell transcriptomes in tissues. Nat. Biotechnol., 2022, 40: 1190-1199.

[180]

KleshchevnikovV, et al. . Cell2location maps fine-grained cell types in spatial transcriptomics. Nat. Biotechnol., 2022, 40: 661-671.

[181]

MaY, ZhouX. Spatially informed cell type deconvolution for spatial transcriptomics. Nat. Biotechnol., 2022, 40: 1349-1359.

[182]

MillerBF, et al. . Reference-free cell type deconvolution of multi-cellular pixel-resolution spatially resolved transcriptomics data. Nat. Commun., 2022, 13: 2339.

[183]

TilburgJ, et al. . Spatial transcriptomics of murine bone marrow megakaryocytes at single-cell resolution. Res Pr. Thromb. Haemost., 2023, 7: 100158.

[184]

ChoC-S, et al. . Microscopic examination of spatial transcriptome using Seq-Scope. Cell, 2021, 184: 3559-3572.e3522.

[185]

VickovicS, et al. . High-definition spatial transcriptomics for in situ tissue profiling. Nat. Methods, 2019, 16: 987-990.

[186]

ToosiS, BehravanN, BehravanJ. Nonunion fractures, mesenchymal stem cells and bone tissue engineering. J. Biomed. Mater. Res. A, 2018, 106: 2552-2562.

[187]

CompstonJE, McClungMR, LeslieWD. Osteoporosis. Lancet, 2019, 393: 364-376.

[188]

FranciscoV, et al. . A new immunometabolic perspective of intervertebral disc degeneration. Nat. Rev. Rheumatol., 2022, 18: 47-60.

[189]

AicaleR, TarantinoD, MaffulliN. Overuse injuries in sport: a comprehensive overview. J. Orthop. Surg. Res., 2018, 13: 309.

[190]

SchedeHH, et al. . Spatial tissue profiling by imaging-free molecular tomography. Nat. Biotechnol., 2021, 39: 968-977.

[191]

ZengH, et al. . Integrative in situ mapping of single-cell transcriptional states and tissue histopathology in a mouse model of Alzheimer’s disease. Nat. Neurosci., 2023, 26: 430-446

[192]

SchottM, et al. . Open-ST: High-resolution spatial transcriptomics in 3D. Cell, 2024, 187: 3953-3972.e26.

[193]

ZeiraR, LandM, StrzalkowskiA, RaphaelBJ. Alignment and integration of spatial transcriptomics data. Nat. Methods, 2022, 19: 567-575.

[194]

JonesA, TownesFW, LiD, EngelhardtBE. Alignment of spatial genomics data using deep Gaussian processes. Nat. Methods, 2023, 20: 1379-1387.

[195]

VandereykenK, SifrimA, ThienpontB, VoetT. Methods and applications for single-cell and spatial multi-omics. Nat. Rev. Genet., 2023, 24: 494-515.

[196]

BaysoyA, BaiZ, SatijaR, FanR. The technological landscape and applications of single-cell multi-omics. Nat. Rev. Mol. Cell Biol., 2023, 24: 695-713.

[197]

LiuY, et al. . High-plex protein and whole transcriptome co-mapping at cellular resolution with spatial CITE-seq. Nat. Biotechnol., 2023, 41: 1405-1409.

[198]

VickovicS, et al. . SM-Omics is an automated platform for high-throughput spatial multi-omics. Nat. Commun., 2022, 13: 795.

[199]

Ben-ChetritN, et al. . Integration of whole transcriptome spatial profiling with protein markers. Nat. Biotechnol., 2023, 41: 788-793.

[200]

ZhangD, et al. . Spatial epigenome-transcriptome co-profiling of mammalian tissues. Nature, 2023, 616: 113-122.

[201]

MarconatoL, et al. . SpatialData: an open and universal data framework for spatial omics. Nat. Methods, 2024, 22: 58-62.

[202]

EisensteinM. Seven technologies to watch in 2022. Nature, 2022, 601: 658-661.

[203]

MadissoonE, et al. . A spatially resolved atlas of the human lung characterizes a gland-associated immune niche. Nat. Genet., 2022, 55: 66-77.

[204]

SakamotoY, SereewattanawootS, SuzukiA. A new era of long-read sequencing for cancer genomics. J. Hum. Genet., 2020, 65: 3-10.

[205]

CharnwichaiP, et al. . Rapid decalcification of articular cartilage and subchondral bone using an ultrasonic cleaner with EDTA. Acta Histochem., 2023, 125: 152009.

[206]

BosiaC, et al. . RNAs competing for microRNAs mutually influence their fluctuations in a highly non-linear microRNA-dependent manner in single cells. Genome Biol., 2017, 18: 37.

[207]

CaiL, FriedmanN, XieXS. Stochastic protein expression in individual cells at the single molecule level. Nature, 2006, 440: 358-362.

[208]

ChuLX, et al. . Spatiotemporal multi-omics: exploring molecular landscapes in aging and regenerative medicine. Mil. Med. Res., 2024, 11: 31

[209]

ChenW, et al. . Live-seq enables temporal transcriptomic recording of single cells. Nature, 2022, 608: 733-740.

[210]

PadeLR, et al. . Biological mass spectrometry enables spatiotemporal ‘omics: From tissues to cells to organelles. Mass Spectrom. Rev., 2024, 43: 106-138.

[211]

ZahediR, et al. . Deep learning in spatially resolved transcriptfomics: a comprehensive technical view. Brief. Bioinform., 2024, 25: bbae082.

[212]

HuJ, et al. . SpaGCN: integrating gene expression, spatial location and histology to identify spatial domains and spatially variable genes by graph convolutional network. Nat. Methods, 2021, 18: 1342-1351.

[213]

PhamD, et al. . Robust mapping of spatiotemporal trajectories and cell-cell interactions in healthy and diseased tissues. Nat. Commun., 2023, 14: 7739.

[214]

DongK, ZhangS. Deciphering spatial domains from spatially resolved transcriptomics with an adaptive graph attention auto-encoder. Nat. Commun., 2022, 13: 1739.

[215]

ShengquanC, et al. . stPlus: a reference-based method for the accurate enhancement of spatial transcriptomics. Bioinformatics, 2021, 37: i299-i307.

[216]

BiancalaniT, et al. . Deep learning and alignment of spatially resolved single-cell transcriptomes with Tangram. Nat. Methods, 2021, 18: 1352-1362.

[217]

YuanY, Bar-JosephZ. GCNG: graph convolutional networks for inferring gene interaction from spatial transcriptomics data. Genome Biol., 2020, 21: 300.

[218]

SongQ, SuJ. DSTG: deconvoluting spatial transcriptomics data through graph-based artificial intelligence. Brief. Bioinform., 2021, 22: bbaa414.

[219]

PetukhovV, et al. . Cell segmentation in imaging-based spatial transcriptomics. Nat. Biotechnol., 2022, 40: 345-354.

[220]

LittmanR, et al. . Joint cell segmentation and cell type annotation for spatial transcriptomics. Mol. Syst. Biol., 2021, 17: e10108.

[221]

AmodioM, et al. . Exploring single-cell data with deep multitasking neural networks. Nat. Methods, 2019, 16: 1139-1145.

[222]

LiX, et al. . Deep learning enables accurate clustering with batch effect removal in single-cell RNA-seq analysis. Nat. Commun., 2020, 11: 2338.

[223]

LakkisJ, et al. . A joint deep learning model enables simultaneous batch effect correction, denoising, and clustering in single-cell transcriptomics. Genome Res, 2021, 31: 1753-1766.

[224]

XuH, et al. . Unsupervised spatially embedded deep representation of spatial transcriptomics. Genome Med, 2024, 16: 12.

[225]

ShahS, et al. . Single-molecule RNA detection at depth by hybridization chain reaction and tissue hydrogel embedding and clearing. Development, 2016, 143: 2862-2867.

[226]

XiaC, et al. . Spatial transcriptome profiling by MERFISH reveals subcellular RNA compartmentalization and cell cycle-dependent gene expression. Proc. Natl. Acad. Sci. USA, 2019, 116: 19490-19499.

[227]

LeeJH. Quantitative approaches for investigating the spatial context of gene expression. WIREs Syst. Biol. Med., 2016, 9: e1369.

[228]

Ferri-BorgognoS, et al. . Molecular, metabolic, and subcellular mapping of the tumor immune microenvironment via 3D targeted and non-targeted multiplex multi-omics analyses. Cancers, 2024, 16: 846.

[229]

YinY, et al. . Unraveling spatial complexity of the tumor microenvironment: a whole transcriptomic perspective with Visium HD. Cancer Res., 2024, 84: abstr. 3645.

[230]

FanZ, ChenR, ChenX. SpatialDB: a database for spatially resolved transcriptomes. Nucleic Acids Res., 2020, 48: D233-D237

Funding

The National Natural Science Youth Foundation of China (Grant No. 82102584).

RIGHTS & PERMISSIONS

The Author(s)

AI Summary AI Mindmap
PDF

502

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/