Construction and Validation of a Reliable Disulfidptosis-Related LncRNAs Signature of the Subtype, Prognostic, and Immune Landscape in Colon Cancer

Disulfidptosis, a novel form of regulated cell death (RCD) associated with metabolism, represents a promising intervention target in cancer therapy. While abnormal lncRNA expression is associated with colon cancer development, the prognostic potential and biological characteristics of disulfidptosis-related lncRNAs (DRLs) remain unclear. Consequently, the research aimed to discover a novel indication of DRLs with significant prognostic implications, and to investigate their possible molecular role in the advancement of colon cancer. Here, we acquired RNA-seq data, pertinent clinical data, and genomic mutations of colon adenocarcinoma (COAD) from the TCGA database, and then DRLs were determined through Pearson correlation analysis. A total of 434 COAD patients were divided in to three subgroups through clustering analysis based on DRLs. By utilizing univariate Cox regression, the least absolute shrinkage and selection operator (LASSO) algorithm, and multivariate Cox regression analysis, we ultimately created a prognostic model consisting of four DRLs (AC007728.3, AP003555.1, ATP2B1.AS1, and NSMCE1.DT), and an external database was used to validate the prognostic features of the risk model. According to the Kaplan-Meier curve analysis, patients in the low-risk group exhibited a considerably superior survival time in comparison to those in the high-risk group. Enrichment analysis revealed a significant association between metabolic processes and the genes that were differentially expressed in the high- and low-risk groups. Additionally, significant differences in the tumor immune microenvironment landscape were observed, specifically pertaining to immune cells, function, and checkpoints. High-risk patients exhibited a low likelihood of immune evasion, as indicated by the Tumor Immune Dysfunction and Exclusion (TIDE) analysis. Patients who exhibit both a high risk and high Tumor Mutational Burden (TMB) experience the least amount of time for survival, whereas those belonging to the low-risk and low-TMB category demonstrate the most favorable prognosis. In addition, the risk groups determined by the 4-DRLs signature displayed distinct drug sensitivities. Finally, we confirmed the levels of expression for four DRLs through rt-qPCR in both tissue samples from colon cancer patients and cell lines. Taken together, the first 4-DRLs-based signature we proposed may serve for a hopeful instrument for forecasting the prognosis, immune landscape, and therapeutic responses in colon cancer patients, thereby facilitating optimal clinical decision-making.

[1]  Haibo Wang,et al.  Disulfidptosis classification of hepatocellular carcinoma reveals correlation with clinical prognosis and immune profile. , 2023, International immunopharmacology.

[2]  J. Thaler,et al.  Ongoing complete response after treatment cessation with dabrafenib, trametinib, and cetuximab as third-line treatment in a patient with advanced BRAFV600E mutated, microsatellite-stable colon cancer: A case report and literature review , 2023, Frontiers in Oncology.

[3]  Chunting Zhou,et al.  Disulfidptosis: a new target for metabolic cancer therapy , 2023, Journal of Experimental & Clinical Cancer Research.

[4]  Xi-shan Wang,et al.  Pan-cancer analysis identifies PD-L2 as a tumor promotor in the tumor microenvironment , 2023, Frontiers in Immunology.

[5]  Rui Chen,et al.  Integrated analysis identifies oxidative stress-related lncRNAs associated with progression and prognosis in colorectal cancer , 2023, BMC Bioinformatics.

[6]  D. Speiser,et al.  CD4+ T cells in cancer , 2023, Nature Cancer.

[7]  Feng Zhang,et al.  ATP2B1-AS1 exacerbates sepsis-induced cell apoptosis and inflammation by regulating miR-23a-3p/TLR4 axis. , 2023, Allergologia et immunopathologia.

[8]  Kellen L. Olszewski,et al.  Actin cytoskeleton vulnerability to disulfide stress mediates disulfidptosis , 2023, Nature Cell Biology.

[9]  F. Gao,et al.  A novel pyroptosis gene expression-based risk score for survival in gastric cancer , 2023, Frontiers in Endocrinology.

[10]  Junxin Wu,et al.  Role of a lipid metabolism-related lncRNA signature in risk stratification and immune microenvironment for colon cancer , 2022, BMC Medical Genomics.

[11]  Shannon M. Mumenthaler,et al.  Neurotransmitter signaling: a new frontier in colorectal cancer biology and treatment , 2022, Oncogene.

[12]  Junyun Luo,et al.  Integrated Analysis of a Ferroptosis-Related LncRNA Signature for Evaluating the Prognosis of Patients with Colorectal Cancer , 2022, Genes.

[13]  M. Ryten,et al.  ggtranscript: an R package for the visualization and interpretation of transcript isoforms using ggplot2 , 2022, bioRxiv.

[14]  R. Samant,et al.  Ribosome Biogenesis: A Central Player in Cancer Metastasis and Therapeutic Resistance , 2022, Cancer research.

[15]  L. Antonuzzo,et al.  Temozolomide Followed by Combination With Low-Dose Ipilimumab and Nivolumab in Patients With Microsatellite-Stable, O6-Methylguanine–DNA Methyltransferase–Silenced Metastatic Colorectal Cancer: The MAYA Trial , 2022, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[16]  Chen Chen,et al.  Colon cancer diagnosis and staging classification based on machine learning and bioinformatics analysis , 2022, Comput. Biol. Medicine.

[17]  Gagandeep Singh,et al.  Machine learning assisted analysis of breast cancer gene expression profiles reveals novel potential prognostic biomarkers for triple-negative breast cancer , 2022, Computational and structural biotechnology journal.

[18]  S. Shamdani,et al.  Weighted correlation network analysis revealed novel long non-coding RNAs for colorectal cancer , 2022, Scientific Reports.

[19]  Shengli Liu,et al.  Molecular Characteristics, Clinical Significance, and Cancer Immune Interactions of Angiogenesis-Associated Genes in Gastric Cancer , 2022, Frontiers in Immunology.

[20]  Y. Zhang,et al.  Identification and Validation of Ferroptosis-Related LncRNA Signatures as a Novel Prognostic Model for Colon Cancer , 2022, Frontiers in Immunology.

[21]  C. Dang,et al.  Targeting cancer metabolism in the era of precision oncology , 2021, Nature reviews. Drug discovery.

[22]  Hossein Tabatabaeian,et al.  ESRG, LINC00518 and PWRN1 are newly-identified deregulated lncRNAs in colorectal cancer , 2021, Experimental and Molecular Pathology.

[23]  Xinxiang Li,et al.  Lnc-RP11-536 K7.3/SOX2/HIF-1α signaling axis regulates oxaliplatin resistance in patient-derived colorectal cancer organoids , 2021, Journal of experimental & clinical cancer research : CR.

[24]  Linhui Wang,et al.  Identification of a Prognosis-Related Risk Signature for Bladder Cancer to Predict Survival and Immune Landscapes , 2021, Journal of immunology research.

[25]  Lei Wang,et al.  ATP2B1-AS1 Promotes Cerebral Ischemia/Reperfusion Injury Through Regulating the miR-330-5p/TLR4-MyD88-NF-κB Signaling Pathway , 2021, Frontiers in Cell and Developmental Biology.

[26]  Robert F. Gruener,et al.  oncoPredict: an R package for predicting in vivo or cancer patient drug response and biomarkers from cell line screening data , 2021, Briefings Bioinform..

[27]  Xiaochen Bo,et al.  clusterProfiler 4.0: A universal enrichment tool for interpreting omics data , 2021, Innovation.

[28]  Yue Zhou,et al.  Insight into Crosstalk between Ferroptosis and Necroptosis: Novel Therapeutics in Ischemic Stroke , 2021, Oxidative medicine and cellular longevity.

[29]  C. Rudin,et al.  Tim-4+ cavity-resident macrophages impair anti-tumor CD8+ T cell immunity. , 2021, Cancer cell.

[30]  X. Zu,et al.  YTHDF1 Is a Potential Pan-Cancer Biomarker for Prognosis and Immunotherapy , 2021, Frontiers in Oncology.

[31]  Senlin Xu,et al.  Alternative approaches to target Myc for cancer treatment , 2021, Signal Transduction and Targeted Therapy.

[32]  Xian Shen,et al.  Long noncoding RNAs: functions and mechanisms in colon cancer , 2020, Molecular Cancer.

[33]  L. Zhuang,et al.  Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy , 2020, Protein & Cell.

[34]  L. Zitvogel,et al.  Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors , 2020, Nature Reviews Clinical Oncology.

[35]  Kaiyou Song,et al.  Silencing of ATP2B1‐AS1 contributes to protection against myocardial infarction in mouse via blocking NFKBIA‐mediated NF‐κB signalling pathway , 2020, Journal of cellular and molecular medicine.

[36]  Xipeng Zhang,et al.  A Robust 6-lncRNA Prognostic Signature for Predicting the Prognosis of Patients With Colorectal Cancer Metastasis , 2020, Frontiers in Medicine.

[37]  A. G. García Carrancá,et al.  mTORC1 as a Regulator of Mitochondrial Functions and a Therapeutic Target in Cancer , 2019, Front. Oncol..

[38]  Enxin Zhang,et al.  Integrated analysis of lncRNA-miRNA-mRNA ceRNA network in squamous cell carcinoma of tongue , 2019, BMC Cancer.

[39]  Z. Trajanoski,et al.  Correction to: Molecular and pharmacological modulators of the tumor immune contexture revealed by deconvolution of RNA-seq data , 2019, Genome Medicine.

[40]  J. Ross,et al.  Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[41]  G. Leone,et al.  The broken cycle: E2F dysfunction in cancer , 2019, Nature Reviews Cancer.

[42]  Yassen Assenov,et al.  Maftools: efficient and comprehensive analysis of somatic variants in cancer , 2018, Genome research.

[43]  M. Leach,et al.  Metabolic biomarkers of response to the AKT inhibitor MK-2206 in pre-clinical models of human colorectal and prostate carcinoma , 2018, British Journal of Cancer.

[44]  X. Liu,et al.  Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response , 2018, Nature Medicine.

[45]  Timothy Solberg,et al.  Machine learning algorithms for outcome prediction in (chemo)radiotherapy: An empirical comparison of classifiers , 2018, Medical physics.

[46]  D. Sansom,et al.  CTLA-4: a moving target in immunotherapy. , 2018, Blood.

[47]  Jun S. Liu,et al.  TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells. , 2017, Cancer research.

[48]  Guang-Rong Yan,et al.  A Peptide Encoded by a Putative lncRNA HOXB-AS3 Suppresses Colon Cancer Growth. , 2017, Molecular cell.

[49]  Noam Shental,et al.  Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine , 2017, Science.

[50]  A. Butte,et al.  xCell: digitally portraying the tissue cellular heterogeneity landscape , 2017, bioRxiv.

[51]  A. Ratti,et al.  The Long Non-Coding RNAs in Neurodegenerative Diseases: Novel Mechanisms of Pathogenesis. , 2016, Current Alzheimer research.

[52]  Jeffrey S. Morris,et al.  The Consensus Molecular Subtypes of Colorectal Cancer , 2015, Nature Medicine.

[53]  Ron Bose,et al.  HER2 activating mutations are targets for colorectal cancer treatment. , 2015, Cancer discovery.

[54]  R. Emerson,et al.  PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.

[55]  K Ito,et al.  Application of ggplot2 to Pharmacometric Graphics , 2013, CPT: pharmacometrics & systems pharmacology.

[56]  Justin Guinney,et al.  GSVA: gene set variation analysis for microarray and RNA-Seq data , 2013, BMC Bioinformatics.

[57]  Sridhar Ramaswamy,et al.  Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells , 2012, Nucleic Acids Res..

[58]  P. Reichardt,et al.  Human Regulatory T Cells Rapidly Suppress T Cell Receptor–Induced Ca2+, NF-κB, and NFAT Signaling in Conventional T Cells , 2011, Science Signaling.

[59]  W. El-Deiry,et al.  Off-Target Lapatinib Activity Sensitizes Colon Cancer Cells Through TRAIL Death Receptor Up-Regulation , 2011, Science Translational Medicine.

[60]  Davis J. McCarthy,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[61]  M. Cappell,et al.  Pathophysiology, clinical presentation, and management of colon cancer. , 2008, Gastroenterology clinics of North America.

[62]  Guido Kroemer,et al.  Self-eating and self-killing: crosstalk between autophagy and apoptosis , 2007, Nature Reviews Molecular Cell Biology.

[63]  Z. Trajanoski,et al.  Type, Density, and Location of Immune Cells Within Human Colorectal Tumors Predict Clinical Outcome , 2006, Science.

[64]  M. Guha,et al.  Mitochondria-to-nucleus stress signaling in mammalian cells: nature of nuclear gene targets, transcription regulation, and induced resistance to apoptosis. , 2005, Gene.

[65]  T. Hickish,et al.  Oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment for colon cancer. , 2004, The New England journal of medicine.

[66]  Ash A. Alizadeh,et al.  SUPPLEMENTARY NOTE , 1879, Botanical Gazette.

[67]  I. Endo,et al.  G2M checkpoint pathway alone is associated with drug response and survival among cell proliferation-related pathways in pancreatic cancer. , 2021, American journal of cancer research.

[68]  D. Gfeller,et al.  EPIC: A Tool to Estimate the Proportions of Different Cell Types from Bulk Gene Expression Data. , 2020, Methods in molecular biology.

[69]  Jacob D. Jaffe,et al.  Next-generation characterization of the Cancer Cell Line Encyclopedia , 2019, Nature.