AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
Home iLIVER Article
Article Link
Collect
Submit Manuscript
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Single-cell transcriptome analysis reveals a cancer-associated fibroblast marker gene signature in hepatocellular carcinoma that predicts prognosis

Hao Chia,1Dapeng Chena,1Yuliang Zhanga,1Zilin CuibYi BaibYamin Zhangb( )
Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300192, China
Department of Hepatobiliary Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China

1 These authors contributed equally to this work.

Show Author Information

Abstract

Background and aims

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death. Multi-pathway combination therapy is used to treat HCC, and immunotherapy is also a routine part of treatment. As a major component of the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) actively participate in cancer progression through complex functions. However, because CAFs dynamically change during cancer development, most of the current treatment strategies targeting CAFs fail. We created a prognostic CAF marker gene signature (CAFMGS) to investigate the utility of CAFs as a prognostic factor and therapeutic target.

Methods

Gene Expression Omnibus (GEO) single-cell RNA sequencing (Sc-RNA-seq) data were analyzed to identify CAF marker genes in HCC. The Cancer Genome Atlas (TCGA) database was used as a training cohort to construct the CAFMGS model and the International Cancer Genome Consortium (ICGC) dataset was used to validate the CAFMGS.

Results

Marker genes in the CAFMGS model were (0.0001-SPP1), (0.0084-VCX3A), (0.0015-HMGA1), (0.0082-PLOD2), and (0.0075-CACYBP). The CAFMGS_score was separated into high-risk and low-risk groups based on the median of the patients' OS. Univariate and multivariate analyses confirmed that CAFMGS_score was an independent prognostic factor in the training group. CAFMGS_score was a more accurate prognostic indicator compared with clinicopathological score and tumor mutational burden score.

Conclusion

CAFMGS offers a fresh perspective on stromal cell marker genes in HCC prognosis and expands our knowledge of CAF heterogeneity and functional diversity, perhaps paving the way for CAF-targeted immunotherapy in HCC patients.

Electronic Supplementary Material

Download File(s)
iliver-2-1-16_ESM.docx (797.5 KB)
iliver-2-1-16_ESM.xlsx (11.4 KB)

References

[1]

Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71(3):209–49.

[2]

Llovet JM, Kelley RK, Villanueva A, et al. Hepatocellular carcinoma. Nat Rev Dis Prim 2021;7(1):6.

[3]

EASL Clinical Practice Guidelines. Management of hepatocellular carcinoma. J Hepatol 2018;69(1):182–236.

[4]

Reig M, Forner A, Ávila MA, et al. Diagnosis and treatment of hepatocellular carcinoma. Update of the consensus document of the AEEH, AEC, SEOM, SERAM, SERVEI, and SETH. Med Clin 2021;156(9):463.e1–463.e30.

[5]

DeBerardinis RJ. Tumor microenvironment, metabolism, and immunotherapy. N Engl J Med 2020;382(9):869–71.

[6]

Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discov 2019;18(2):99–115.

[7]

Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer 2016; 16(9):582–98.

[8]

Shiga K, Hara M, Nagasaki T, et al. Cancer-associated fibroblasts: their characteristics and their roles in tumor growth. Cancers (Basel) 2015;7(4):2443–58.

[9]

Paauwe M, Schoonderwoerd M, Helderman R, et al. Endoglin expression on cancerassociated fibroblasts regulates invasion and stimulates colorectal cancer metastasis. Clin Cancer Res 2018;24(24):6331–44.

[10]

Torre LA, Islami F, Siegel RL, et al. Global cancer in women: burden and trends. Cancer Epidemiol Biomarkers Prev 2017;26(4):444–57.

[11]

Su S, Chen J, Yao H, et al. CD10(+)GPR77(+) cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell 2018; 172(4):841–856.e16.

[12]

Gaggioli C, Hooper S, Hidalgo-Carcedo C, et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol 2007;9(12):1392–400.

[13]

Bhome R, Goh RW, Bullock MD, et al. Exosomal microRNAs derived from colorectal cancer-associated fibroblasts: role in driving cancer progression. Aging (Albany NY) 2017;9(12):2666–94.

[14]

Shi Y, Gao W, Lytle NK, et al. Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature 2019;569(7754):131–5.

[15]

Bruzzese F, Hägglöf C, Leone A, et al. Local and systemic protumorigenic effects of cancer-associated fibroblast-derived GDF15. Cancer Res 2014;74(13):3408–17.

[16]

Monteran L, Erez N. The dark side of fibroblasts: cancer-associated fibroblasts as mediators of immunosuppression in the tumor microenvironment. Front Immunol 2019;10:1835.

[17]

Kaps L, Schuppan D. Targeting cancer associated fibroblasts in liver fibrosis and liver cancer using nanocarriers. Cells 2020;9(9).

[18]

Heinrich S, Craig AJ, Ma L, et al. Understanding tumour cell heterogeneity and its implication for immunotherapy in liver cancer using single-cell analysis. J Hepatol 2021;74(3):700–15.

[19]

Sebastian A, Hum NR, Martin KA, et al. Single-cell transcriptomic analysis of tumorderived fibroblasts and normal tissue-resident fibroblasts reveals fibroblast heterogeneity in breast cancer. Cancers (Basel) 2020;12(5).

[20]

Sha D, Jin Z, Budczies J, et al. Tumor mutational burden as a predictive biomarker in solid tumors. Cancer Discov 2020;10(12):1808–25.

[21]

Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017;377(25):2500–1.

[22]

Li C, Chen J, Li Y, et al. 6-Phosphogluconolactonase promotes hepatocellular carcinogenesis by activating pentose phosphate pathway. Front Cell Dev Biol 2021; 9:753196.

[23]

Aran D, Looney AP, Liu L, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol 2019;20(2):163–72.

[24]

Becht E, McInnes L, Healy J, et al. Dimensionality reduction for visualizing singlecell data using UMAP. Nat Biotechnol 2018.

[25]

Walter W, Sánchez-Cabo F, Ricote M. GOplot: an R package for visually combining expression data with functional analysis. Bioinformatics 2015;31(17):2912–4.

[26]

Aibar S, González-Blas CB, Moerman T, et al. SCENIC: single-cell regulatory network inference and clustering. Nat Methods 2017;14(11):1083–6.

[27]

Trapnell C, Cacchiarelli D, Grimsby J, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol 2014;32(4):381–6.

[28]

Moons KG, Altman DG, Reitsma JB, et al. Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis (TRIPOD): explanation and elaboration. Ann Intern Med 2015;162(1):W1–73.

[29]

Li M, Wang J, Wang C, et al. Microenvironment remodeled by tumor and stromal cells elevates fibroblast-derived COL1A1 and facilitates ovarian cancer metastasis. Exp Cell Res 2020;394(1):112153.

[30]

Hutchenreuther J, Vincent K, Norley C, et al. Activation of cancer-associated fibroblasts is required for tumor neovascularization in a murine model of melanoma. Matrix Biol 2018;74:52–61.

[31]

Dasgupta S, Ghosh T, Dhar J, et al. RGS5-TGFβ-Smad2/3 axis switches pro- to antiapoptotic signaling in tumor-residing pericytes, assisting tumor growth. Cell Death Differ 2021;28(11):3052–76.

[32]

Tsata V, Möllmert S, Schweitzer C, et al. A switch in pdgfrb(+) cell-derived ECM composition prevents inhibitory scarring and promotes axon regeneration in the zebrafish spinal cord. Dev Cell 2021;56(4):509–524.e9.

[33]

Little K, Llorián-Salvador M, Tang M, et al. Macrophage to myofibroblast transition contributes to subretinal fibrosis secondary to neovascular age-related macular degeneration. J Neuroinflammation 2020;17(1):355.

[34]

Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100(1):57–70.

[35]

Asghar U, Witkiewicz AK, Turner NC, et al. The history and future of targeting cyclin-dependent kinases in cancer therapy. Nat Rev Drug Discov 2015;14(2):130–46.

[36]

Oshi M, Takahashi H, Tokumaru Y, et al. G2M cell cycle pathway score as a prognostic biomarker of metastasis in estrogen receptor (ER)-Positive breast cancer. Int J Mol Sci 2020;21(8).

[37]

Yuen VW, Wong CC. Hypoxia-inducible factors and innate immunity in liver cancer. J Clin Invest 2020;130(10):5052–62.

[38]

Feng J, Li J, Wu L, et al. Emerging roles and the regulation of aerobic glycolysis in hepatocellular carcinoma. J Exp Clin Cancer Res 2020;39(1):126.

[39]

Briscoe J, Thérond PP. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol 2013;14(7):416–29.

[40]

Gu Y, Wang Y, He L, et al. Circular RNA circIPO11 drives self-renewal of liver cancer initiating cells via Hedgehog signaling. Mol Cancer 2021;20(1):132.

[41]

Cubillos-Ruiz JR, Silberman PC, Rutkowski MR, et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 2015; 161(7):1527–38.

[42]

Feng YX, Jin DX, Sokol ES, et al. Cancer-specific PERK signaling drives invasion and metastasis through CREB3L1. Nat Commun 2017;8(1):1079.

[43]

Tran LL, Dang T, Thomas R, et al. ELF3 mediates IL-1α induced differentiation of mesenchymal stem cells to inflammatory iCAFs. Stem Cell 2021;39(12): 1766–77.

[44]

Yassin N, AbouZid SF, El-Kalaawy AM, et al. Silybum marianum total extract, silymarin and silibinin abate hepatocarcinogenesis and hepatocellular carcinoma growth via modulation of the HGF/c-Met, Wnt/β-catenin, and PI3K/Akt/mTOR signaling pathways. Biomed Pharmacother 2022;145:112409.

[45]

Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371(23):2189–99.

[46]

Levine AJ, Momand J, Finlay CA. The p53 tumour suppressor gene. Nature 1991; 351(6326):453–6.

[47]

Xu F, Jin T, Zhu Y, et al. Immune checkpoint therapy in liver cancer. J Exp Clin Cancer Res 2018;37(1):110.

[48]

Liu Z, Lin Y, Zhang J, et al. Molecular targeted and immune checkpoint therapy for advanced hepatocellular carcinoma. J Exp Clin Cancer Res 2019;38(1):447.

[49]

Mishra S, Yadav T, Rani V. Exploring miRNA based approaches in cancer diagnostics and therapeutics. Crit Rev Oncol Hematol 2016;98:12–23.

[50]

Billan S, Kaidar-Person O, Gil Z. Treatment after progression in the era of immunotherapy. Lancet Oncol 2020;21(10):e463–76.

[51]

Zhang Y, Du W, Chen Z, et al. Upregulation of PD-L1 by SPP1 mediates macrophage polarization and facilitates immune escape in lung adenocarcinoma. Exp Cell Res 2017;359(2):449–57.

[52]

Deng G, Zeng F, Su J, et al. BET inhibitor suppresses melanoma progression via the noncanonical NF-κB/SPP1 pathway. Theranostics 2020;10(25):11428–43.

[53]

Taguchi A, Taylor AD, Rodriguez J, et al. A search for novel cancer/testis antigens in lung cancer identifies VCX/Y genes, expanding the repertoire of potential immunotherapeutic targets. Cancer Res 2014;74(17):4694–705.

[54]

Wei F, Zhang T, Deng SC, et al. PD-L1 promotes colorectal cancer stem cell expansion by activating HMGA1-dependent signaling pathways. Cancer Lett 2019; 450:1–13.

[55]

Du H, Pang M, Hou X, et al. PLOD2 in cancer research. Biomed Pharmacother 2017; 90:670–6.

[56]

Topolska-Woś AM, Chazin WJ, Filipek A. CacyBP/SIP–Structure and variety of functions. Biochim Biophys Acta 2016;1860(1 Pt A):79–85.

iLIVER
Pages 16-25
Cite this article:
Chi H, Chen D, Zhang Y, et al. Single-cell transcriptome analysis reveals a cancer-associated fibroblast marker gene signature in hepatocellular carcinoma that predicts prognosis. iLIVER, 2023, 2(1): 16-25. https://doi.org/10.1016/j.iliver.2022.12.002

436

Views

1

Crossref

Altmetrics

Received: 06 September 2022
Revised: 12 December 2022
Accepted: 14 December 2022
Published: 13 January 2023
© 2023 The Author(s). Published by Elsevier Ltd on behalf of Tsinghua University Press.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return