Skip to main content

Advertisement

Log in

Molecular Characterization of Cuproptosis-related lncRNAs: Defining Molecular Subtypes and a Prognostic Signature of Ovarian Cancer

  • Research
  • Published:
Biological Trace Element Research Aims and scope Submit manuscript

Abstract

Cuproptosis, a newly discovered form of programmed cell death, relies on mitochondrial respiration, the chain of which has been found to be altered in ovarian cancer (OC). The current work probed into the effects of Cuproptosis on the prognosis, immune microenvironment and therapeutic response of OC based on Cuproptosis-related lncRNAs. Data on OC gene expression and clinical characteristics were collected from TCGA, ICGC and GEO databases, and mRNA and lncRNA were distinguished. Cuproptosis-related lncRNAs were screened for consensus clustering analysis. Differentially expressed lncRNAs (DElncRNAs) were identified between clusters, and least absolute shrinkage and selection operator (LASSO) and Cox regression analysis were performed to establish a prognostic signature. Its potential value in OC was evaluated by Gene Set Enrichment Analysis (GSEA), tumor cell mutation and immune microenvironment analysis, and response to immunotherapy and antineoplastic drugs. According to the classification scheme of Cuproptosis-related lncRNAs, OC was divided into four molecular subtypes, which were different in survival time, immune characteristics and somatic mutation. The prognostic signature between subtypes included 10 lncRNAs, which were significantly correlated with the prognosis, immune microenvironment related indexes, the expression of immune checkpoint molecules and the sensitivity of antineoplastic drug Paclitaxel and Gefitinib of OC. We examined the expression of ten LncRNAs in OC cell lines and found that LINC00189, ZFHX4-AS1, RPS6KA2-IT1 and C9orf106 were expressed elevated in OC cell lines, and LINC00861, LINC00582, DEPDC1-AS1, LINC01556, LEMD1-AS1, TYMSOS expression was decreased in OC cell lines. The results of CCK8 showed that the cell viability of OC cells decreased after inhibition of C9orf106, whereas the cell viability of OC cells increased after inhibition of LEMD1-AS1. This work revealed new Cuproptosis-related lncRNA molecular subtypes exhibiting tumor microenvironment (TME) heterogeneity for OC and proposed a prognostic signature that may have benefits in understanding the prognosis, pathological features and immune microenvironment of OC patients.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

Data Availability

The datasets generated and/or analyzed during the current study are available in the [GSE102073] repository, [https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE102073].

Abbreviations

OC:

Ovarian cancer

DElncRNAs:

Differentially expressed Long non-coding RNAs

LASSO:

Least absolute shrinkage and selection operator

GSEA:

Gene Set Enrichment Analysis

RNA-seq:

RNA sequencing

TCGA:

The Cancer Genome Atlas

GEO:

Gene Expression Omnibus

FPKM:

Fragments per kilobase of exon model per million mapped fragments

SNV:

Single nucleotide variation

CNV:

Copy number variation

ICGC:

International Cancer Genome Consortium

TPM:

Transcripts per kilobase

ROC:

Receiver operating characteristic

AUC:

Area under the curve

TME:

Tumor microenvironment

Fges:

Functional gene expression signatures

MATH:

Mutant-allele tumor heterogeneity

TMB:

Tumor mutational burden

HRD:

Homologous recombination DNA repair deficiency

GSEA:

Gene Set Enrichment Analysis

TIDE:

Tumor Immune Dysfunction and Exclusion

ICI:

Immune checkpoint inhibitor

GDSC:

Genome of Drug Sensitivity in Cancer

IC50:

Half maximal inhibitory concentration

MDSCs:

Myeloid-derived suppressor cells

CAFs:

Cancer-associated fibroblasts

TAMs:

Tumor-associated macrophages

References

  1. Kurnit KC, Fleming GF, Lengyel E (2021) Updates and new options in advanced epithelial ovarian cancer treatment. Obstet Gynecol 137(1):108–121

    Article  CAS  PubMed  Google Scholar 

  2. Ovejero-Sanchez M, Gonzalez-Sarmiento R, Herrero AB (2023) DNA damage response alterations in ovarian cancer: from molecular mechanisms to therapeutic opportunities. Cancers (Basel) 15(2):448

  3. Battistini C, Cavallaro U (2023) Patient-derived in vitro models of ovarian cancer: powerful tools to explore the biology of the disease and develop personalized treatments. Cancers (Basel) 15(2):368

  4. van Zyl B, Tang D, Bowden NA (2018) Biomarkers of platinum resistance in ovarian cancer: what can we use to improve treatment. Endocr Relat Cancer 25(5):R303–R318

    Article  PubMed  Google Scholar 

  5. Marrelli D, Ansaloni L, Federici O, Asero S, Carbone L, Marano L et al (2022) Cytoreductive surgery (CRS) and HIPEC for advanced ovarian cancer with peritoneal metastases: italian PSM oncoteam evidence and study purposes. Cancers (Basel) 14(23):6010

  6. Shukla P, Singh KK (2021) The mitochondrial landscape of ovarian cancer: emerging insights. Carcinogenesis 42(5):663–671

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. De Rasmo D, Cormio A, Cormio G, Signorile A (2023) Ovarian cancer: a landscape of mitochondria with emphasis on mitochondrial dynamics. Int J Mol Sci; 24(2).

  8. Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdusamad M et al (2022) Copper induces cell death by targeting lipoylated TCA cycle proteins. Science 375(6586):1254–1261

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  9. Huang X, Zhou S, Toth J, Hajdu A (2022) Cuproptosis-related gene index: a predictor for pancreatic cancer prognosis, immunotherapy efficacy, and chemosensitivity. Front Immunol 13:978865

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chen B, Zhou X, Yang L, Zhou H, Meng M, Zhang L et al (2022) A Cuproptosis Activation Scoring model predicts neoplasm-immunity interactions and personalized treatments in glioma. Comput Biol Med 148:105924

    Article  CAS  PubMed  Google Scholar 

  11. Tong X, Tang R, Xiao M, Xu J, Wang W, Zhang B et al (2022) Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol 15(1):174

    Article  PubMed  PubMed Central  Google Scholar 

  12. Yoshihara K, Shahmoradgoli M, Martinez E, Vegesna R, Kim H, Torres-Garcia W et al (2013) Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun 4:2612

    Article  PubMed  ADS  Google Scholar 

  13. Bagaev A, Kotlov N, Nomie K, Svekolkin V, Gafurov A, Isaeva O et al (2021) Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell 39(6):845-65e7

    Article  CAS  PubMed  Google Scholar 

  14. Xu L, Deng C, Pang B, Zhang X, Liu W, Liao G et al (2018) TIP: a web server for resolving tumor immunophenotype profiling. Cancer Res 78(23):6575–6580

    Article  CAS  PubMed  Google Scholar 

  15. Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y et al (2015) Robust enumeration of cell subsets from tissue expression profiles. Nat Methods 12(5):453–457

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mroz EA, Rocco JW (2013) MATH, a novel measure of intratumor genetic heterogeneity, is high in poor-outcome classes of head and neck squamous cell carcinoma. Oral Oncol 49(3):211–215

    Article  CAS  PubMed  Google Scholar 

  17. Stover EH, Fuh K, Konstantinopoulos PA, Matulonis UA, Liu JF (2020) Clinical assays for assessment of homologous recombination DNA repair deficiency. Gynecol Oncol 159(3):887–898

    Article  CAS  PubMed  Google Scholar 

  18. Jiang P, Gu S, Pan D, Fu J, Sahu A, Hu X et al (2018) Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat Med 24(10):1550–1558

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Auslander N, Zhang G, Lee JS, Frederick DT, Miao B, Moll T et al (2018) Robust prediction of response to immune checkpoint blockade therapy in metastatic melanoma. Nat Med 24(10):1545–1549

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Varanasi SK, Kaech SM, Bui JD (2022) SnapShot: cancer immunoediting. Cell 185(21):4038-e1

    Article  CAS  PubMed  Google Scholar 

  21. van den Bulk J, Verdegaal EM, de Miranda NF (2018) Cancer immunotherapy: broadening the scope of targetable tumours. Open Biol 8(6):180037

  22. Tang D, Chen X, Kroemer G (2022) Cuproptosis: a copper-triggered modality of mitochondrial cell death. Cell Res 32(5):417–8

    Article  PubMed  PubMed Central  Google Scholar 

  23. Liu H, Tang T (2022) Pan-cancer genetic analysis of cuproptosis and copper metabolism-related gene set. Front Oncol 12:952290

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. De Leo A, Santini D, Ceccarelli C, Santandrea G, Palicelli A, Acquaviva G et al (2021) What is new on ovarian carcinoma: integrated morphologic and molecular analysis following the new 2020 World Health Organization classification of female genital tumors. Diagnostics (Basel) 11(4):697

  25. Palmirotta R, Silvestris E, D’Oronzo S, Cardascia A, Silvestris F (2017) Ovarian cancer: novel molecular aspects for clinical assessment. Crit Rev Oncol Hematol 117:12–29

    Article  PubMed  Google Scholar 

  26. Yu F, Quan F, Xu J, Zhang Y, Xie Y, Zhang J et al (2019) Breast cancer prognosis signature: linking risk stratification to disease subtypes. Brief Bioinform 20(6):2130–40

    Article  CAS  PubMed  Google Scholar 

  27. Radu MR, Pradatu A, Duica F, Micu R, Cretoiu SM, Suciu N et al (2021) Ovarian cancer: biomarkers and targeted therapy. Biomedicines 9(6):693

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Reina-Campos M, Scharping NE, Goldrath AW (2021) CD8(+) T cell metabolism in infection and cancer. Nat Rev Immunol 21(11):718–38

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hu X, Bian C, Zhao X, Yi T (2022) Efficacy evaluation of multi-immunotherapy in ovarian cancer: from bench to bed. Front Immunol 13:1034903

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Marth C, Wieser V, Tsibulak I, Zeimet AG (2019) Immunotherapy in ovarian cancer: fake news or the real deal? Int J Gynecol Cancer 29(1):201–11

    Article  PubMed  Google Scholar 

  32. Zhang Y, Zhang X, Zhu H, Liu Y, Cao J, Li D et al (2020) Identification of potential prognostic long non-coding RNA biomarkers for predicting recurrence in patients with cervical cancer. Cancer Manag Res 12:719–30

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Gao Q, Shi Y, Sun Y, Zhou S, Liu Z, Sun X et al (2023) Identification and verification of aging-related lncRNAs for prognosis prediction and immune microenvironment in patients with head and neck squamous carcinoma. Oncol Res 31(1):35–61

    Article  PubMed  PubMed Central  Google Scholar 

  34. Wang X, Wang Y, Sun F, Xu Y, Zhang Z, Yang C et al (2022) Novel LncRNA ZFHX4-AS1 as a potential prognostic biomarker that affects the immune microenvironment in ovarian cancer. Front Oncol 12:945518

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cheng Y, Wang X, Qi P, Liu C, Wang S, Wan Q et al (2021) Tumor microenvironmental competitive endogenous RNA network and immune cells act as robust prognostic predictor of acute myeloid leukemia. Front Oncol 11:584884

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Guo R, Qin Y (2020) LEMD1-AS1 suppresses ovarian cancer progression through regulating miR-183-5p/TP53 axis. Onco Targets Ther 13:7387–98

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhang Z, Wang J, Zhang X, Ran B, Wen J, Zhang H (2023) TYMSOS-miR-101-3p-NETO2 axis promotes osteosarcoma progression. Mol Cell Probes 67:101887

    Article  CAS  PubMed  Google Scholar 

  38. Gu Y, Wan C, Zhou G, Zhu J, Shi Z, Zhuang Z (2021) TYMSOS drives the proliferation, migration, and invasion of gastric cancer cells by regulating ZNF703 via sponging miR-4739. Cell Biol Int 45(8):1710–9

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This research was supported by Guangxi Medical high-level and sub backbone talents training “139” program (No. G202003005), the Key Research and Development Program of Guangxi (No. GuikeAB18126056) and Liuzhou Science and Technology Bureau Project (No. 2018BJ10301, No. 2019BJ10604), Guangxi Science and Technology Plan Project (Guangxi Clinical Research Center for Obstetrics and Gynecology, GuiKe AD22035223) and Guangxi Self-Financing Research Program of Guangxi Region Health Commission (No. Z20210518, No. Z-B20221595).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to this present work: [NL] and [KY] designed the study; [DLH] and [SL] acquired the data; [DYZ] drafted the manuscript; [JJL] and [LF] revised the manuscript. All authors read and approved the manuscript.

Corresponding authors

Correspondence to Jingjing Li or Li Fan.

Ethics declarations

Conflict of Interest

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (XLSX 18 KB)

Supplementary file2 (PDF 578 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, N., Yu, K., Huang, D. et al. Molecular Characterization of Cuproptosis-related lncRNAs: Defining Molecular Subtypes and a Prognostic Signature of Ovarian Cancer. Biol Trace Elem Res 202, 1428–1445 (2024). https://doi.org/10.1007/s12011-023-03780-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12011-023-03780-3

Keywords

Navigation