Skip to main content
Top
Published in: BMC Cancer 1/2024

Open Access 01-12-2024 | Gastric Cancer | Research

Integrating trans-omics, cellular experiments and clinical validation to identify ILF2 as a diagnostic serum biomarker and therapeutic target in gastric cancer

Authors: Shao-Song Liu, Qin-Si Wan, Cong Lv, Jin-Ke Wang, Song Jiang, Dan Cai, Mao-Sheng Liu, Ting Wang, Kun-He Zhang

Published in: BMC Cancer | Issue 1/2024

Login to get access

Abstract

Background

Gastric cancer (GC) lacks serum biomarkers with clinical diagnostic value. Multi-omics analysis is an important approach to discovering cancer biomarkers. This study aimed to identify and validate serum biomarkers for GC diagnosis by cross-analysis of proteomics and transcriptomics datasets.

Methods

A cross-omics analysis was performed to identify overlapping differentially expressed genes (DEGs) between our previous aptamer-based GC serum proteomics dataset and the GC tissue RNA-Seq dataset in The Cancer Genome Atlas (TCGA) database, followed by lasso regression and random forest analysis to select key overlapping DEGs as candidate biomarkers for GC. The mRNA levels and diagnostic performance of these candidate biomarkers were analyzed in the original and independent GC datasets to select valuable candidate biomarkers. The valuable candidate biomarkers were subjected to bioinformatics analysis to select those closely associated with the biological behaviors of GC as potential biomarkers. The clinical diagnostic value of the potential biomarkers was validated using serum samples, and their expression levels and functions in GC cells were validated using in vitro cell experiments.

Results

Four candidate biomarkers (ILF2, PGM2L1, CHD7, and JCHAIN) were selected. Their mRNA levels differed significantly between tumor and normal tissues and showed different diagnostic performances for GC, with areas under the receiver operating characteristic curve (AUROCs) of 0.629–0.950 in the TCGA dataset and 0.736–0.840 in the Gene Expression Omnibus (GEO) dataset. In the bioinformatics analysis, only ILF2 (interleukin enhancer-binding factor 2) gene levels were associated with immune cell infiltration, some checkpoint gene expression, chemotherapy sensitivity, and immunotherapy response. Serum levels of ILF2 were higher in GC patients than in controls, with an AUROC of 0.944 for the diagnosis of GC, and it was also detected in the supernatants of GC cells. Knockdown of ILF2 by siRNA significantly reduced the proliferation and colony formation of GC cells. Overexpression of ILF2 significantly promotes the proliferation and colony formation of gastric cancer cells.

Conclusions

Trans-omics analysis of proteomics and transcriptomics is an efficient approach for discovering serum biomarkers, and ILF2 is a potential diagnostic biomarker and therapeutic target of gastric cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Sung H, 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.CrossRefPubMed Sung H, 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.CrossRefPubMed
4.
go back to reference Cai Q, et al. Development and validation of a prediction rule for estimating gastric cancer risk in the Chinese high-risk population: a nationwide multicentre study. Gut. 2019;68(9):1576–87.CrossRefPubMed Cai Q, et al. Development and validation of a prediction rule for estimating gastric cancer risk in the Chinese high-risk population: a nationwide multicentre study. Gut. 2019;68(9):1576–87.CrossRefPubMed
5.
go back to reference Kaise M, et al. The combination of serum trefoil factor 3 and pepsinogen testing is a valid non-endoscopic biomarker for predicting the presence of gastric cancer: a new marker for gastric cancer risk. J Gastroenterol. 2011;46(6):736–45.CrossRefPubMed Kaise M, et al. The combination of serum trefoil factor 3 and pepsinogen testing is a valid non-endoscopic biomarker for predicting the presence of gastric cancer: a new marker for gastric cancer risk. J Gastroenterol. 2011;46(6):736–45.CrossRefPubMed
6.
go back to reference Ikeda A, et al. Serum metabolomics as a novel diagnostic approach for gastrointestinal cancer. Biomed Chromatogr. 2012;26(5):548–58.CrossRefPubMed Ikeda A, et al. Serum metabolomics as a novel diagnostic approach for gastrointestinal cancer. Biomed Chromatogr. 2012;26(5):548–58.CrossRefPubMed
7.
go back to reference Thanh HP et al. Emerging role of circulating tumor cells in gastric cancer. Cancers (Basel). 2020;12(3). Thanh HP et al. Emerging role of circulating tumor cells in gastric cancer. Cancers (Basel). 2020;12(3).
9.
go back to reference Yuan L, et al. Long non-coding RNAs towards precision medicine in gastric cancer: early diagnosis, treatment, and drug resistance. Mol Cancer. 2020;19(1):96.CrossRefPubMedPubMedCentral Yuan L, et al. Long non-coding RNAs towards precision medicine in gastric cancer: early diagnosis, treatment, and drug resistance. Mol Cancer. 2020;19(1):96.CrossRefPubMedPubMedCentral
11.
go back to reference Wang K, Huang C, Nice EC. Proteomics, genomics and transcriptomics: their emerging roles in the discovery and validation of colorectal cancer biomarkers. Expert Rev Proteom. 2014;11(2):179–205.CrossRef Wang K, Huang C, Nice EC. Proteomics, genomics and transcriptomics: their emerging roles in the discovery and validation of colorectal cancer biomarkers. Expert Rev Proteom. 2014;11(2):179–205.CrossRef
12.
go back to reference Yang W, et al. Integrating proteomics and transcriptomics for the identification of potential targets in early colorectal cancer. Int J Oncol. 2019;55(2):439–50.PubMedPubMedCentral Yang W, et al. Integrating proteomics and transcriptomics for the identification of potential targets in early colorectal cancer. Int J Oncol. 2019;55(2):439–50.PubMedPubMedCentral
13.
go back to reference Bartha Á et al. Proteotranscriptomic discrimination of tumor and normal tissues in renal cell carcinoma. Int J Mol Sci. 2023;24(5). Bartha Á et al. Proteotranscriptomic discrimination of tumor and normal tissues in renal cell carcinoma. Int J Mol Sci. 2023;24(5).
14.
go back to reference Lapitz A, et al. Liquid biopsy-based protein biomarkers for risk prediction, early diagnosis, and prognostication of cholangiocarcinoma. J Hepatol. 2023;79(1):93–108.CrossRefPubMedPubMedCentral Lapitz A, et al. Liquid biopsy-based protein biomarkers for risk prediction, early diagnosis, and prognostication of cholangiocarcinoma. J Hepatol. 2023;79(1):93–108.CrossRefPubMedPubMedCentral
15.
go back to reference Hirsch FR, et al. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389(10066):299–311.CrossRefPubMed Hirsch FR, et al. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389(10066):299–311.CrossRefPubMed
16.
go back to reference Blum A, Wang P, Zenklusen JC. SnapShot: TCGA-analyzed tumors. Cell. 2018;173(2):530. Blum A, Wang P, Zenklusen JC. SnapShot: TCGA-analyzed tumors. Cell. 2018;173(2):530.
17.
go back to reference Ucaryilmaz MC, Ozcan G. Comprehensive bioinformatic analysis reveals a cancer-associated fibroblast gene signature as a poor prognostic factor and potential therapeutic target in gastric cancer. BMC Cancer. 2022;22(1):692.CrossRef Ucaryilmaz MC, Ozcan G. Comprehensive bioinformatic analysis reveals a cancer-associated fibroblast gene signature as a poor prognostic factor and potential therapeutic target in gastric cancer. BMC Cancer. 2022;22(1):692.CrossRef
18.
go back to reference Wang Q, et al. Nomogram established on account of Lasso-Cox regression for predicting recurrence in patients with early-stage hepatocellular carcinoma. Front Immunol. 2022;13:1019638.CrossRefPubMedPubMedCentral Wang Q, et al. Nomogram established on account of Lasso-Cox regression for predicting recurrence in patients with early-stage hepatocellular carcinoma. Front Immunol. 2022;13:1019638.CrossRefPubMedPubMedCentral
19.
21.
go back to reference Zeng D, et al. Gene expression profiles for a prognostic immunoscore in gastric cancer. Br J Surg. 2018;105(10):1338–48.CrossRefPubMed Zeng D, et al. Gene expression profiles for a prognostic immunoscore in gastric cancer. Br J Surg. 2018;105(10):1338–48.CrossRefPubMed
22.
23.
go back to reference Zeng D, et al. Tumor microenvironment characterization in gastric cancer identifies prognostic and immunotherapeutically relevant gene signatures. Cancer Immunol Res. 2019;7(5):737–50.CrossRefPubMed Zeng D, et al. Tumor microenvironment characterization in gastric cancer identifies prognostic and immunotherapeutically relevant gene signatures. Cancer Immunol Res. 2019;7(5):737–50.CrossRefPubMed
27.
go back to reference Yang W, et al. Genomics of drug sensitivity in cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41(Database issue):D955–61.PubMed Yang W, et al. Genomics of drug sensitivity in cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41(Database issue):D955–61.PubMed
28.
go back to reference Kao PN, et al. Cloning and expression of cyclosporin A- and FK506-sensitive nuclear factor of activated T-cells: NF45 and NF90. J Biol Chem. 1994;269(32):20691–9.CrossRefPubMed Kao PN, et al. Cloning and expression of cyclosporin A- and FK506-sensitive nuclear factor of activated T-cells: NF45 and NF90. J Biol Chem. 1994;269(32):20691–9.CrossRefPubMed
29.
go back to reference Zhang Y, et al. lncRNA AK085865 promotes macrophage M2 polarization in CVB3-induced VM by regulating ILF2-ILF3 complex-mediated miRNA-192 biogenesis. Mol Ther Nucleic Acids. 2020;21:441–51.CrossRefPubMedPubMedCentral Zhang Y, et al. lncRNA AK085865 promotes macrophage M2 polarization in CVB3-induced VM by regulating ILF2-ILF3 complex-mediated miRNA-192 biogenesis. Mol Ther Nucleic Acids. 2020;21:441–51.CrossRefPubMedPubMedCentral
30.
go back to reference Marchesini M, et al. ILF2 is a regulator of RNA splicing and DNA damage response in 1q21-amplified multiple myeloma. Cancer Cell. 2017;32(1):88–e1006.CrossRefPubMedPubMedCentral Marchesini M, et al. ILF2 is a regulator of RNA splicing and DNA damage response in 1q21-amplified multiple myeloma. Cancer Cell. 2017;32(1):88–e1006.CrossRefPubMedPubMedCentral
31.
go back to reference Zhang X, et al. Interleukin enhancer-binding factor 2 promotes cell proliferation and DNA damage response in metastatic melanoma. Clin Transl Med. 2021;11(10):e608.CrossRefPubMedPubMedCentral Zhang X, et al. Interleukin enhancer-binding factor 2 promotes cell proliferation and DNA damage response in metastatic melanoma. Clin Transl Med. 2021;11(10):e608.CrossRefPubMedPubMedCentral
32.
go back to reference Du H et al. ILF2 directly binds and stabilizes CREB to stimulate malignant phenotypes of liver cancer cells. Anal Cell Pathol (Amst). 2019;2019:1575031. Du H et al. ILF2 directly binds and stabilizes CREB to stimulate malignant phenotypes of liver cancer cells. Anal Cell Pathol (Amst). 2019;2019:1575031.
33.
go back to reference Cheng S et al. Expression and critical role of interleukin enhancer binding factor 2 in Hepatocellular Carcinoma. Int J Mol Sci. 2016;17(8). Cheng S et al. Expression and critical role of interleukin enhancer binding factor 2 in Hepatocellular Carcinoma. Int J Mol Sci. 2016;17(8).
34.
go back to reference Zhao M, et al. ILF2 cooperates with E2F1 to maintain mitochondrial homeostasis and promote small cell lung cancer progression. Cancer Biol Med. 2019;16(4):771–83.CrossRefPubMedPubMedCentral Zhao M, et al. ILF2 cooperates with E2F1 to maintain mitochondrial homeostasis and promote small cell lung cancer progression. Cancer Biol Med. 2019;16(4):771–83.CrossRefPubMedPubMedCentral
35.
go back to reference Noblejas-López M et al. Genomic mapping of splicing-related genes identify amplifications in LSM1, CLNS1A, and ILF2 in luminal breast cancer. Cancers (Basel). 2021;13(16). Noblejas-López M et al. Genomic mapping of splicing-related genes identify amplifications in LSM1, CLNS1A, and ILF2 in luminal breast cancer. Cancers (Basel). 2021;13(16).
36.
go back to reference Yin ZH et al. Expression and clinical significance of ILF2 in gastric cancer. Dis Markers. 2017;2017:4387081. Yin ZH et al. Expression and clinical significance of ILF2 in gastric cancer. Dis Markers. 2017;2017:4387081.
37.
go back to reference Arai H, et al. Expression of DNA damage response proteins in gastric cancer: comprehensive protein profiling and histological analysis. Int J Oncol. 2018;52(3):978–88.PubMed Arai H, et al. Expression of DNA damage response proteins in gastric cancer: comprehensive protein profiling and histological analysis. Int J Oncol. 2018;52(3):978–88.PubMed
38.
go back to reference Li D, et al. LncRNA ELF3-AS1 inhibits gastric cancer by forming a negative feedback loop with SNAI2 and regulates ELF3 mRNA stability via interacting with ILF2/ILF3 complex. J Exp Clin Cancer Res. 2022;41(1):332.CrossRefPubMedPubMedCentral Li D, et al. LncRNA ELF3-AS1 inhibits gastric cancer by forming a negative feedback loop with SNAI2 and regulates ELF3 mRNA stability via interacting with ILF2/ILF3 complex. J Exp Clin Cancer Res. 2022;41(1):332.CrossRefPubMedPubMedCentral
Metadata
Title
Integrating trans-omics, cellular experiments and clinical validation to identify ILF2 as a diagnostic serum biomarker and therapeutic target in gastric cancer
Authors
Shao-Song Liu
Qin-Si Wan
Cong Lv
Jin-Ke Wang
Song Jiang
Dan Cai
Mao-Sheng Liu
Ting Wang
Kun-He Zhang
Publication date
01-12-2024
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2024
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-024-12175-z

Other articles of this Issue 1/2024

BMC Cancer 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine