Spatial transcriptomics reveals prognosis-associated cellular heterogeneity in the papillary thyroid carcinoma microenvironment

Kai Yan , Qing-Zhi Liu , Rong-Rong Huang , Yi-Hua Jiang , Zhen-Hua Bian , Si-Jin Li , Liang Li , Fei Shen , Koichi Tsuneyama , Qing-Ling Zhang , Zhe-Xiong Lian , Haixia Guan , Bo Xu

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1594

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (3) : e1594 DOI: 10.1002/ctm2.1594
RESEARCH ARTICLE

Spatial transcriptomics reveals prognosis-associated cellular heterogeneity in the papillary thyroid carcinoma microenvironment

Author information +
History +
PDF

Abstract

Background: Papillary thyroid carcinoma (PTC) is the most common malignant endocrine tumour, and its incidence and prevalence are increasing considerably. Cellular heterogeneity in the tumour microenvironment is important for PTC prognosis. Spatial transcriptomics is a powerful technique for cellular heterogeneity study.

Methods: In conjunction with a clinical pathologist identification method, spatial transcriptomics was employed to characterise the spatial location and RNA profiles of PTC-associated cells within the tissue sections. The spatial RNA-clinical signature genes for each cell type were extracted and applied to outlining the distribution regions of specific cells on the entire section. The cellular heterogeneity of each cell type was further revealed by ContourPlot analysis, monocle analysis, trajectory analysis, ligand–receptor analysis and Gene Ontology enrichment analysis.

Results: The spatial distribution region of tumour cells, typical and atypical follicular cells (FCs and AFCs) and immune cells were accurately and comprehensively identified in all five PTC tissue sections. AFCs were identified as a transitional state between FCs and tumour cells, exhibiting a higher resemblance to the latter. Three tumour foci were shared among all patients out of the 13 observed. Notably, tumour foci No. 2 displayed elevated expression levels of genes associated with lower relapse-free survival in PTC patients. We discovered key ligand–receptor interactions, including LAMB3–ITGA2, FN1–ITGA3 and FN1–SDC4, involved in the transition of PTC cells from FCs to AFCs and eventually to tumour cells. High expression of these patterns correlated with reduced relapse-free survival. In the tumour immune microenvironment, reduced interaction between myeloid-derived TGFB1 and TGFBR1 in tumour focus No. 2 contributed to tumourigenesis and increased heterogeneity. The spatial RNA-clinical analysis method developed here revealed prognosis-associated cellular heterogeneity in the PTC microenvironment.

Conclusions: The occurrence of tumour foci No. 2 and three enhanced ligand–receptor interactions in the AFC area/tumour foci reduced the relapse-free survival of PTC patients, potentially leading to improved prognostic strategies and targeted therapies for PTC patients.

Keywords

cellular heterogeneity / ligand‒receptor interactions / papillary thyroid carcinoma / prognosis / spatial transcriptomics / tumour microenvironment

Cite this article

Download citation ▾
Kai Yan, Qing-Zhi Liu, Rong-Rong Huang, Yi-Hua Jiang, Zhen-Hua Bian, Si-Jin Li, Liang Li, Fei Shen, Koichi Tsuneyama, Qing-Ling Zhang, Zhe-Xiong Lian, Haixia Guan, Bo Xu. Spatial transcriptomics reveals prognosis-associated cellular heterogeneity in the papillary thyroid carcinoma microenvironment. Clinical and Translational Medicine, 2024, 14(3): e1594 DOI:10.1002/ctm2.1594

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

McLeod DS, Sawka AM, Cooper DS. Controversies in primary treatment of low-risk papillary thyroid cancer. Lancet. 2013;381:1046-1057.

[2]

Schneider DF, Chen H. New developments in the diagnosis and treatment of thyroid cancer. CA Cancer J Clin. 2013;63:374-394.

[3]

Xing M, Haugen BR, Schlumberger M. Progress in molecular-based management of differentiated thyroid cancer. Lancet. 2013;381:1058-1069.

[4]

Pu W, Shi X, Yu P, et al. Single-cell transcriptomic analysis of the tumor ecosystems underlying initiation and progression of papillary thyroid carcinoma. Nat Commun. 2021;12:6058.

[5]

Hu A, Yang LY, Liang J, Lu D, Cao FF. Single-cell RNA sequencing reveals the regenerative potential of thyroid follicular epithelial cells in metastatic thyroid carcinoma. Biochem Biophys Res Commun. 2020;531:552-558.

[6]

Pan J, Ye F, Yu C, et al. Papillary thyroid carcinoma landscape and its immunological link with hashimoto thyroiditis at single-cell resolution. Front Cell Dev Biol. 2021;9:758339.

[7]

Luo H, Xia X, Kim GD, et al. Characterizing dedifferentiation of thyroid cancer by integrated analysis. Sci Adv. 2021;7.

[8]

Luu MH, Fischer AH, Stockl TJ, Pisharodi L, Owens CL. Atypical follicular cells with equivocal features of papillary thyroid carcinoma is not a low-risk cytologic diagnosis. Acta Cytol. 2011;55:526-530.

[9]

VanderLaan PA, Marqusee E, Krane JF. Clinical outcome for atypia of undetermined significance in thyroid fine-needle aspirations: should repeated fna be the preferred initial approach? Am J Clin Pathol. 2011;135:770-775.

[10]

Kim M, Kim JE, Kim HJ, Chung YR, Kwak Y, Park SY. Cytologic diagnosis of noninvasive follicular thyroid neoplasm with papillary-like nuclear features and its impact on the risk of malignancy in the bethesda system for reporting thyroid cytopathology: an institutional experience. J Pathol Transl Med. 2018;52:171-178.

[11]

Ferraro V, Sgaramella LI, Di Meo G, et al. Current concepts in parathyroid carcinoma: a single centre experience. BMC Endocr Disord. 2019;19:46.

[12]

Haddad RI, Bischoff L, Ball D, et al. Thyroid carcinoma, version 2.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2022;20:925-951.

[13]

Galdiero MR, Varricchi G, Marone G. The immune network in thyroid cancer. Oncoimmunology. 2016;5:e1168556.

[14]

Angell TE, Lechner MG, Jang JK, LoPresti JS, Epstein AL. MHC class I loss is a frequent mechanism of immune escape in papillary thyroid cancer that is reversed by interferon and selumetinib treatment in vitro. Clin Cancer Res. 2014;20:6034-6044.

[15]

Hanahan D. Hallmarks of cancer: new dimensions. Cancer Discov. 2022;12:31-46.

[16]

Schlumberger MJ. Papillary and follicular thyroid carcinoma. N Engl J Med. 1998;338:297-306.

[17]

Coca-Pelaz A, Shah JP, Hernandez-Prera JC, et al. Papillary thyroid cancer-aggressive variants and impact on management: a narrative review. Adv Ther. 2020;37:3112-3128.

[18]

Yan K, Da TT, Bian ZH, et al. Multi-omics analysis identifies FoxO1 as a regulator of macrophage function through metabolic reprogramming. Cell Death Dis. 2020;11:800.

[19]

Liao J, Lu X, Shao X, Zhu L, Fan X. Uncovering an organ's molecular architecture at single-cell resolution by spatially resolved transcriptomics. Trends Biotechnol. 2021;39:43-58.

[20]

Macerola E, Poma AM, Basolo F. NanoString in the screening of genetic abnormalities associated with thyroid cancer. Semin Cancer Biol. 2022;79:132-140.

[21]

Yoo SK, Song YS, Lee EK, et al. Integrative analysis of genomic and transcriptomic characteristics associated with progression of aggressive thyroid cancer. Nat Commun. 2019;10:2764.

[22]

Peng M, Wei G, Zhang Y, et al. Single-cell transcriptomic landscape reveals the differences in cell differentiation and immune microenvironment of papillary thyroid carcinoma between genders. Cell Biosci. 2021;11:39.

[23]

Stahl PL, Salmen F, Vickovic S, et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science. 2016;353:78-82.

[24]

Maniatis S, Petrescu J, Phatnani H. Spatially resolved transcriptomics and its applications in cancer. Curr Opin Genet Dev. 2021;66:70-77.

[25]

Bassiouni R, Gibbs LD, Craig DW, Carpten JD, McEachron TA. Applicability of spatial transcriptional profiling to cancer research. Mol Cell. 2021;81:1631-1639.

[26]

Danaher P, Kim Y, Nelson B, et al. Advances in mixed cell deconvolution enable quantification of cell types in spatial transcriptomic data. Nat Commun. 2022;13:385.

[27]

Asp M, Giacomello S, Larsson L, et al. A spatiotemporal organ-wide gene expression and cell atlas of the developing human heart. Cell. 2019;179:1647-1660 e19.

[28]

Mantri M, Scuderi GJ, Abedini-Nassab R, et al. Spatiotemporal single-cell RNA sequencing of developing chicken hearts identifies interplay between cellular differentiation and morphogenesis. Nat Commun. 2021;12:1771.

[29]

Fawkner-Corbett D, Antanaviciute A, Parikh K, et al. Spatiotemporal analysis of human intestinal development at single-cell resolution. Cell. 2021;184:810-826 e23.

[30]

Ortiz C, Navarro JF, Jurek A, Martin A, Lundeberg J, Meletis K. Molecular atlas of the adult mouse brain. Sci Adv. 2020;6:eabb3446.

[31]

Joglekar A, Prjibelski A, Mahfouz A, et al. A spatially resolved brain region- and cell type-specific isoform atlas of the postnatal mouse brain. Nat Commun. 2021;12:463.

[32]

Maynard KR, Collado-Torres L, Weber LM, et al. Transcriptome-scale spatial gene expression in the human dorsolateral prefrontal cortex. Nat Neurosci. 2021;24:425-436.

[33]

Crosse EI, Gordon-Keylock S, Rybtsov S, et al. Multi-layered spatial transcriptomics identify secretory factors promoting human hematopoietic stem cell development. Cell Stem Cell. 2020;27:822-839 e8.

[34]

Haas S, Baccin C, Al-Sabah J, Velten L, Lars S, Trumpp A. Combined single-cell and spatial transcriptomics to deconvolute the hematopoietic stem cell niche. Blood. 2018;132:876.

[35]

Gao S, Shi Q, Zhang Y, et al. Identification of HSC/MPP expansion units in fetal liver by single-cell spatiotemporal transcriptomics. Cell Res. 2022;32:38-53.

[36]

Ji AL, Rubin AJ, Thrane K, et al. Multimodal analysis of composition and spatial architecture in human squamous cell carcinoma. Cell. 2020;182:497-514 e22.

[37]

Moncada R, Barkley D, Wagner F, et al. Integrating microarray-based spatial transcriptomics and single-cell RNA-seq reveals tissue architecture in pancreatic ductal adenocarcinomas. Nat Biotechnol. 2020;38:333-342.

[38]

Nagasawa S, Kuze Y, Maeda I, et al. Genomic profiling reveals heterogeneous populations of ductal carcinoma in situ of the breast. Commun Biol. 2021;4:438.

[39]

Andersson A, Larsson L, Stenbeck L, et al. Spatial deconvolution of HER2-positive breast cancer delineates tumor-associated cell type interactions. Nat Commun. 2021;12:6012.

[40]

Wu R, Guo W, Qiu X, et al. Comprehensive analysis of spatial architecture in primary liver cancer. Sci Adv. 2021;7:eabg3750.

[41]

French JD, Bible K, Spitzweg C, Haugen BR, Ryder M. Leveraging the immune system to treat advanced thyroid cancers. Lancet Diabet Endocrinol. 2017;5:469-481.

[42]

Cabanillas ME, McFadden DG, Durante C. Thyroid cancer. Lancet. 2016;388:2783-2795.

[43]

Ahn S, Kim TH, Kim SW, et al. Comprehensive screening for PD-L1 expression in thyroid cancer. Endocr Relat Cancer. 2017;24:97-106.

[44]

Longo SK, Guo MG, Ji AL, Khavari PA. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat Rev Genet. 2021;22:627-644.

[45]

LiVolsi VA. Papillary thyroid carcinoma: an update. Mod Pathol. 2011;24(2):S1-S9. Suppl.

[46]

Kleino I, Frolovaite P, Suomi T, Elo LL. Computational solutions for spatial transcriptomics. Comput Struct Biotechnol J. 2022;20:4870-4884.

[47]

Hong S, Xie Y, Cheng Z, et al. Distinct molecular subtypes of papillary thyroid carcinoma and gene signature with diagnostic capability. Oncogene. 2022;41:5121-5132.

[48]

Ravi VM, Will P, Kueckelhaus J, et al. Spatially resolved multi-omics deciphers bidirectional tumor-host interdependence in glioblastoma. Cancer Cell. 2022;40:639-655.

[49]

Ren Y, Huang Z, Zhou L, et al. Spatial transcriptomics reveals niche-specific enrichment and vulnerabilities of radial glial stem-like cells in malignant gliomas. Nat Commun. 2023;14:1028.

[50]

Lotfollahi M, Naghipourfar M, Luecken MD, et al. Mapping single-cell data to reference atlases by transfer learning. Nat Biotechnol. 2022;40:121-130.

[51]

Stuart T, Butler A, Hoffman P, et al. Comprehensive integration of single-cell data. Cell. 2019;177:1888-1902 e21.

[52]

Tang Z, Kang B, Li C, Chen T, Zhang Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res. 2019;47:W556-W560.

[53]

Ligorio M, Sil S, Malagon-Lopez J, et al. Stromal microenvironment shapes the intratumoral architecture of pancreatic cancer. Cell. 2019;178:160-175 e27.

[54]

Armingol E, Officer A, Harismendy O, Lewis NE. Deciphering cell-cell interactions and communication from gene expression. Nat Rev Genet. 2021;22:71-88.

[55]

Morton LM, Karyadi DM, Stewart C, et al. Radiation-related genomic profile of papillary thyroid carcinoma after the Chernobyl accident. Science. 2021:372.

[56]

Hovhannisyan G, Harutyunyan T, Aroutiounian R, Liehr T. DNA copy number variations as markers of mutagenic impact. Int J Mol Sci. 2019:20.

[57]

Tanaka H, Watanabe T. Mechanisms underlying recurrent genomic amplification in human cancers. Trends Cancer. 2020;6:462-477.

[58]

Ma Y, Zhou X. Spatially informed cell-type deconvolution for spatial transcriptomics. Nat Biotechnol. 2022;40:1349-1359.

[59]

Verrecchia F, Chu ML, Mauviel A. Identification of novel TGF-beta /Smad gene targets in dermal fibroblasts using a combined cDNA microarray/promoter transactivation approach. J Biol Chem. 2001;276:17058-17062.

[60]

Chen Y, Zhu S, Liu T, et al. Epithelial cells activate fibroblasts to promote esophageal cancer development. Cancer Cell. 2023;41:903-918 e8.

[61]

Haffner MC, Zwart W, Roudier MP, et al. Genomic and phenotypic heterogeneity in prostate cancer. Nat Rev Urol. 2021;18:79-92.

[62]

Hiroshima Y, Kasajima R, Kimura Y, et al. Novel targets identified by integrated cancer-stromal interactome analysis of pancreatic adenocarcinoma. Cancer Lett. 2020;469:217-227.

[63]

Sautes-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19:307-325.

[64]

Ding GY, Ma JQ, Yun JP, et al. Distribution and density of tertiary lymphoid structures predict clinical outcome in intrahepatic cholangiocarcinoma. J Hepatol. 2022;76:608-618.

[65]

Feng W, Huang W, Chen J, et al. CXCL12-mediated HOXB5 overexpression facilitates colorectal cancer metastasis through transactivating CXCR4 and ITGB3. Theranostics. 2021;11:2612-2633.

[66]

Aoki T, Chong LC, Takata K, et al. Single-cell profiling reveals the importance of CXCL13/CXCR5 axis biology in lymphocyte-rich classic Hodgkin lymphoma. Proc Natl Acad Sci USA. 2021;118.

[67]

Cabrita R, Lauss M, Sanna A, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561-565.

[68]

Wang J, Lin D, Peng H, Huang Y, Huang J, Gu J. Cancer-derived immunoglobulin G promotes tumor cell growth and proliferation through inducing production of reactive oxygen species. Cell Death Dis. 2013;4:e945.

[69]

Wang Z, Geng Z, Shao W, et al. Cancer-derived sialylated IgG promotes tumor immune escape by binding to Siglecs on effector T cells. Cell Mol Immunol. 2020;17:1148-1162.

[70]

Zhao J, Peng H, Gao J, et al. Current insights into the expression and functions of tumor-derived immunoglobulins. Cell Death Discov. 2021;7:148.

[71]

Zhao Y, Liu Y, Chen Z, Korteweg C, Gu J. Immunoglobulin g (IgG) expression in human umbilical cord endothelial cells. J Histochem Cytochem. 2011;59:474-488.

[72]

Shao W, Zhang C, Liu E, et al. Identification of liver epithelial cell-derived Ig expression in mu chain-deficient mice. Sci Rep. 2016;6:23669.

[73]

Shao W, Hu F, Ma J, et al. Epithelial cells are a source of natural IgM that contribute to innate immune responses. Int J Biochem Cell Biol. 2016;73:19-29.

RIGHTS & PERMISSIONS

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

AI Summary AI Mindmap
PDF

175

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/