Resolving the heterogeneous tumour microenvironment in cardiac myxoma through single-cell and spatial transcriptomics

Xuanyu Liu , Huayan Shen , Jinxing Yu , Fengming Luo , Tianjiao Li , Qi Li , Xin Yuan , Yang Sun , Zhou Zhou

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (2) : e1581

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (2) : e1581 DOI: 10.1002/ctm2.1581
RESEARCH ARTICLE

Resolving the heterogeneous tumour microenvironment in cardiac myxoma through single-cell and spatial transcriptomics

Author information +
History +
PDF

Abstract

Background: Cardiac myxoma (CM) is the most common (58%-80%) type of primary cardiac tumours. Currently, there is a need to develop medical therapies, especially for patients not physically suitable for surgeries. However, the mechanisms that shape the tumour microenvironment (TME) in CM remain largely unknown, which impedes the development of targeted therapies. Here, we aimed to dissect the TME in CM at single-cell and spatial resolution.

Methods: We performed single-cell transcriptomic sequencing and Visium CytAssist spatial transcriptomic (ST) assays on tumour samples from patients with CM. A comprehensive analysis was performed, including unsupervised clustering, RNA velocity, clonal substructure inference of tumour cells and cell-cell communication.

Results: Unsupervised clustering of 34 759 cells identified 12 clusters, which were assigned to endothelial cells (ECs), mesenchymal stroma cells (MSCs), and tumour-infiltrating immune cells. Myxoma tumour cells were found to encompass two closely related phenotypic states, namely, EC-like tumour cells (ETCs) and MSC-like tumour cells (MTCs). According to RNA velocity, our findings suggest that ETCs may be directly differentiated from MTCs. The immune microenvironment of CM was found to contain multiple factors that promote immune suppression and evasion, underscoring the potential of using immunotherapies as a treatment option. Hyperactive signals sent primarily by tumour cells were identified, such as MDK, HGF, chemerin, and GDF15 signalling. Finally, the ST assay uncovered spatial features of the subclusters, proximal cell-cell communication, and clonal evolution of myxoma tumour cells.

Conclusions: Our study presents the first comprehensive characterisation of the TME in CM at both single-cell and spatial resolution. Our study provides novel insight into the differentiation of myxoma tumour cells and advance our understanding of the TME in CM. Given the rarity of cardiac tumours, our study provides invaluable datasets and promotes the development of medical therapies for CM.

Keywords

cardiac myxoma / myxoma tumour cell / single-cell RNA sequencing / spatial transcriptomics / tumour microenvironment

Cite this article

Download citation ▾
Xuanyu Liu, Huayan Shen, Jinxing Yu, Fengming Luo, Tianjiao Li, Qi Li, Xin Yuan, Yang Sun, Zhou Zhou. Resolving the heterogeneous tumour microenvironment in cardiac myxoma through single-cell and spatial transcriptomics. Clinical and Translational Medicine, 2024, 14(2): e1581 DOI:10.1002/ctm2.1581

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Velez Torres JM, Martinez Duarte E, Diaz-Perez JA, Rosenberg AE. Cardiac myxoma: review and update of contemporary immunohistochemical markers and molecular pathology. Adv Anat Pathol. 2020;27:380-384.

[2]

Griborio-Guzman AG, Aseyev OI, Shah H, Sadreddini M. Cardiac myxomas: clinical presentation, diagnosis and management. Heart. 2022;108:827-833.

[3]

Barh D, Kumar A, Chatterjee S, Liloglou T. Molecular features, markers, drug targets, and prospective targeted therapeutics in cardiac myxoma. Curr Cancer Drug Targets. 2009;9:705-716.

[4]

Wilkes D, Charitakis K, Basson CT. Inherited disposition to cardiac myxoma development. Nat Rev Cancer. 2006;6:157-165.

[5]

Burke A, Tavora FR, Maleszewski JJ, Frazier AA. Cardiac myxoma. ATLAS TUMOR Pathol. Ser. 4 Tumors Hear. Gt. Vessel., vol. 22. Maryland: American Registry of Pathology.2015:79-107.

[6]

Di Vito A, Mignogna C, Donato G. The mysterious pathways of cardiac myxomas: a review of histogenesis, pathogenesis and pathology. Histopathology. 2015;66:321-332.

[7]

Amano J, Kono T, Wada Y, et al. Cardiac myxoma: its origin and tumor characteristics. Ann Thorac Cardiovasc Surg. 2003;9:215-221.

[8]

Scalise M, Torella M, Marino F, et al. Atrial myxomas arise from multipotent cardiac stem cells. Eur Heart J. 2020;41:4332-4345.

[9]

de Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41:374-403.

[10]

Jia Q, Chu H, Jin Z, Long H, Zhu B. High-throughput single-cell sequencing in cancer research. Signal Transduct Target Ther. 2022;7:145.

[11]

Elhanani O, Ben-Uri R, Keren L. Spatial profiling technologies illuminate the tumor microenvironment. Cancer Cell. 2023;41:404-420.

[12]

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

[13]

Wolock SL, Lopez R, Klein AM. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 2019;8:281-291. e9

[14]

Miao Z, Deng K, Wang X, Zhang X. DEsingle for detecting three types of differential expression in single-cell RNA-seq data. Bioinformatics. 2018;34:3223-3224.

[15]

Litviňuková M, Talavera-López C, Maatz H, et al. Cells of the adult human heart. Nature. 2020;588:466-472.

[16]

Bindea G, Mlecnik B, Hackl H, et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics. 2009;25:1091-1093.

[17]

De Falco A, Caruso F, Su XD, Iavarone A, Ceccarelli M. A variational algorithm to detect the clonal copy number substructure of tumors from scRNA-seq data. Nat Commun. 2023;14:1074.

[18]

Morabito S, Reese F, Rahimzadeh N, Miyoshi E, Swarup V. hdWGCNA identifies co-expression networks in high-dimensional transcriptomics data. Cell Reports Methods. 2023;3:100498.

[19]

Bergen V, Lange M, Peidli S, Wolf FA, Theis FJ. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat Biotechnol. 2020;38:1408-1414.

[20]

La Manno G, Soldatov R, Zeisel A, et al. RNA velocity of single cells. Nature. 2018;560:494-498.

[21]

Pham D, Tan X, Balderson B, et al. Robust mapping of spatiotemporal trajectories and cell-cell interactions in healthy and diseased tissues. Nat Commun. 2023;14:1-25.

[22]

Jin S, Plikus MV, Nie Q, CellChat for systematic analysis of cell-cell communication from single-cell and spatially resolved transcriptomics. BioRxiv. 2023.

[23]

Jin S, Guerrero-Juarez CF, Zhang L, et al. Inference and analysis of cell-cell communication using CellChat. Nat Commun. 2021;12:1088.

[24]

Cheng J, Zhang J, Wu Z, Sun X. Inferring microenvironmental regulation of gene expression from single-cell RNA sequencing data using scMLnet with an application to COVID-19. Brief Bioinform. 2021;22:988-1005.

[25]

Heo K, Lee S. TSPAN8 as a novel emerging therapeutic target in cancer for monoclonal antibody therapy. Biomolecules. 2020;10:388.

[26]

Sidney LE, Branch MJ, Dunphy SE, Dua HS, Hopkinson A. Concise review: evidence for CD34 as a common marker for diverse progenitors. Stem Cells. 2014;32:1380-1389.

[27]

Skamrov AV, Nechaenko MA, Goryunova LE, et al. Gene expression analysis to identify mRNA markers of cardiac myxoma. J Mol Cell Cardiol. 2004;37:717-733.

[28]

Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15:486-499.

[29]

Pan Y, Yu Y, Wang X, Zhang T. Tumor-associated macrophages in tumor immunity. Front Immunol. 2020;11:583084.

[30]

Filippou PS, Karagiannis GS, Constantinidou A. Midkine (MDK) growth factor: a key player in cancer progression and a promising therapeutic target. Oncogene. 2020;39:2040-2054.

[31]

Fu J, Su X, Li Z, et al. HGF/c-MET pathway in cancer: from molecular characterization to clinical evidence. Oncogene. 2021;40:4625-4651.

[32]

Spanopoulou A, Gkretsi V. Growth differentiation factor 15 (GDF15) in cancer cell metastasis: from the cells to the patients. Clin Exp Metastasis. 2020;37:451-464.

[33]

Tyebally S, Chen D, Bhattacharyya S, et al. Cardiac tumors: state-of-the-art review. JACC CardioOncol. 2020;2:293-311.

[34]

Di Vito A, Santise G, Mignogna C, et al. Innate immunity in cardiac myxomas and its pathological and clinical correlations. Innate Immun. 2018;24:47-53.

[35]

Willis NA, Rass E, Scully R. Deciphering the code of the cancer genome: mechanisms of chromosome rearrangement genomic instability and the evolution of a cancer. Trends Cancer. 2015;1:217-230.

[36]

Dijkhuizen T, De Jong B, Meuzelaar JJ, Molenaar WM, Van Den Berg E. No cytogenetic evidence for involvement of gene(s) at 2p16 in sporadic cardiac myxomas: cytogenetic changes in ten sporadic cardiac myxomas. Cancer Genet Cytogenet. 2001;126:162-165.

[37]

Boutry J, Tissot S, Ujvari B, et al. The evolution and ecology of benign tumors. Biochim Biophys Acta—Rev Cancer. 2022;1877:188643.

[38]

Basso C, Rizzo S, Valente M, Thiene G. Cardiac masses and tumours. Heart. 2016;102:1230-1245.

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

153

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/