Abstract
Background
Muscle-invasive bladder cancer (MIBC) is a prevalent and aggressive
malignancy. Ferroptosis and cuproptosis are recently discovered forms
of programmed cell death (PCD) that have attracted much attention.
However, their interactions and impacts on MIBC overall survival (OS)
and treatment outcomes remain unclear.
Methods
Data from the TCGA-BLCA project (as the training set), cBioPortal
database, and GEO datasets ([32]GSE13507 and [33]GSE32894, as the test
sets) were utilized to identify hub ferroptosis/cuproptosis-related
genes (FRGs and CRGs) and develop a prognostic signature. Differential
expression analysis (DEA) was conducted, followed by univariate and
multivariate Cox’s regression analyses and multiple machine learning
(ML) techniques to select genetic features. The performance of the
ferroptosis/cuproptosis-related signature was evaluated using
Kaplan–Meier (K–M) survival analysis and receiver-operating
characteristics (ROC) curves. Mutational and tumour immune
microenvironment landscapes were also explored. Real-time quantitative
reverse transcription polymerase chain reaction (RT-qPCR) experiments
confirmed the expression patterns of the hub genes, and functional
assays assessed the effects of SCD knockdown on cell viability,
proliferation, and migration.
Results
DEA revealed dysregulated FRGs and CRGs in the TCGA MIBC cohort. SCD,
DDR2, and MT1A were identified as hub genes. A prognostic signature
based on the sum of the weighted expression of these genes demonstrated
strong predictive efficacy in the training and test sets. Nomogram
incorporating this signature accurately predicted 1-, 3-, and 5-year
survival probabilities in the TCGA cohort and [34]GSE13507 dataset.
Copy number variation (CNV) and tumour immune microenvironment analysis
revealed that high risk score level groups were associated with
immunosuppression and lower tumour purity. The associations of risk
scores with immunotherapy and chemical drugs were also explored,
indicating their potential for guiding treatment for MIBC patients. The
dysregulated expression patterns of three hub genes were validated by
RT-qPCR experiments.
Conclusions
Targeting hub FRGs and CRGs could be a promising therapeutic approach
for MIBC. Our prognostic model offers a new framework for MIBC
subtyping and can inform personalized therapeutic strategies.
Supplementary Information
The online version contains supplementary material available at
10.1186/s12885-024-12741-5.
Keywords: Muscle-invasive bladder cancer, Ferroptosis, Cuproptosis,
TCGA, Nomogram, Prognostic model
Introduction
Bladder cancer stands as the most common malignancy within the urinary
system [[35]1]. In the U.S., it ranks as the sixth most prevalent
cancer, with 83,190 new cases annually, and is the ninth leading cause
of cancer-related deaths [[36]2]. Men are notably more affected by this
disease than women [[37]1, [38]2]. Muscle-invasive bladder cancer
(MIBC) is an aggressive form of urothelial carcinoma (UC) characterized
by tumour invasion into the detrusor muscle [[39]3]. It exhibits
significant heterogeneity and prognostic variability, posing
considerable challenges to current therapeutic approaches [[40]4].
Despite the application of multiple treatment modalities including
surgical resection, chemotherapy, and radiotherapy, the 5-year survival
rate for MIBC remains at only 40–60% [[41]5]. Therefore, there is an
urgent need to develop a validated prognostic model and discover novel
therapeutic targets.
Recent advancements in programmed cell death (PCD), particularly
ferroptosis and cuproptosis, have greatly enhanced our understanding of
cancer progression [[42]6–[43]8]. Ferroptosis is an iron-dependent cell
death marked by lipid peroxidation [[44]9, [45]10], while cuproptosis
involves copper-induced protein aggregation and cell death [[46]11,
[47]12]. The crosstalk between these two cell death pathways suggests
that targeting both types of cell death could enhance the effectiveness
of cancer treatments [[48]13]. A study on hepatocellular carcinoma
identified a signature combining ferroptosis-related or
cuproptosis-related genes (FRGs, CRGs) (G6PD, NRAS, RRM2, SQSTM1,
SRXN1, TXNRD1, and ZFP69B) for prognosis and therapy prediction
[[49]14]. In breast cancer, another study established a prognostic
model based on five CRGs and FRGs (ANKRD52, HOXC10, KNOP1, SGPP1, and
TRIM45). These genes are significantly associated with immune
infiltration and can predict patient outcomes [[50]15]. However, the
combined effect of ferroptosis and cuproptosis on MIBC survival and
treatment outcomes remains unclear.
Given the heterogeneity of MIBC and the urgent need for effective
treatment options, immunotherapy, including immune checkpoint
inhibitors (ICIs), has gathered widespread attention as a novel
anti-tumour strategy. The expression of programmed death-ligand 1
(PD-L1) in UC patients is a significant predictive biomarker for
improved overall survival (OS) with immune checkpoint inhibitors (ICIs)
compared to chemotherapy, particularly in PD-L1–positive patients
[[51]16, [52]17].
Multi-omics approaches, encompassing genomics, transcriptomics,
proteomics, and metabolomics, have been proven to be pivotal in
advancing cancer research [[53]18]. By analysing transcriptomics and
genomics data, researchers identified a ferroptosis/cuproptosis-related
signature, which was used to develop a prognostic model for patients
with MIBC. Integrating multi-omics analysis techniques, this study
investigates the roles of ferroptosis and cuproptosis in MIBC
progression. Furthermore, it evaluates the signature’s predictive value
for immunotherapy efficacy and drug activity, thereby improving disease
management.
Methods
The workflow of this study is shown in Additional file 1: Figure S1.
Patient cohorts
Transcriptional gene expression matrix (RNA-sequencing [RNA-seq] data)
and clinical information of patients in The Cancer Genome Atlas
(TCGA)-BLCA project were obtained from the Genomic Data Commons (GDC)
Data Portal ([54]https://portal.gdc.cancer.gov/). The segmental CNV
data of the TCGA-BLCA cohort were simultaneously downloaded from the
cBioPortal for Cancer Genomics software platform
([55]https://www.cbioportal.org/) [[56]19]. Two probe intensity
datasets, [57]GSE13507 [[58]20] and [59]GSE32894 [[60]21], were
accessed through the Gene Expression Omnibus (GEO) repository
([61]https://www.ncbi.nlm.nih.gov/geo/). Detailed data sources, sample
inclusion–exclusion criteria, and data processing methods are provided
in Additional file 2. The final TCGA cohort (training cohort 1)
included a total of 384 tumour samples (segmental CNV profiles: 380
tumour samples) and 17 histologically normal adjacent tumour (NAT)
tissues. [62]GSE13507 dataset comprised 165 primary bladder cancer
patients, and [63]GSE32894 dataset consisted of 224 UC cases.
Cell culture
Human bladder cancer cell lines T24 and UM-UC-3, along with the normal
human uroepithelial cell line SV-HUC-1, were obtained from the American
Type Culture Collection (ATCC, Manassas, VA, USA). These cell lines
were cultured in media supplemented with 10% fetal bovine serum (FBS)
and 1% penicillin/streptomycin, under a 5% CO[2] atmosphere at 37 °C.
The specific media used were Roswell Park Memorial Institute medium
(RPMI)-1640 for T24 cells, Minimum Essential Medium (MEM) for UM-UC-3
cells, and F12K for SV-HUC-1 cells. Each cell line was tested using
three samples, and all experiments were carried out in triplicate.
RT-qPCR experiment
Total RNA from T24, UM-UC-3, and SV-HUC-1 cells was extracted using the
FastPure Cell/Tissue Total RNA Isolation Kit V2. Reverse transcription
was performed with the HiScript® III 1st Strand cDNA Synthesis Kit
(+ gDNA wiper). The real-time quantitative reverse transcription
polymerase chain reaction (RT-qPCR) assays utilized the Taq Pro
Universal SYBR qPCR Master Mix, purchased from Vazyme (Nanjing, China).
GAPDH served as an internal control. Primer sequences for SCD, DDR2,
MT1A, and GAPDH are listed in Table [64]1.
Table 1.
List of primer sequences for SCD, DDR2, MT1A, and GAPDH (housekeeping
gene)
Gene Sequence Length (bp)
SCD Forward CCCGACGTGGCTTTTTCTTC 20
Reverse GCCAGGTTTGTAGTACCTCCTC 22
DDR2 Forward GTTGGGGAAACGCAGTGGAT 20
Reverse GGTCTCCCTTGATGGAGGTTTC 22
MT1A Forward CTCTTGCTGTTGCTGATGGG 20
Reverse TCGTGAGACCTTCGCTCTTGT 21
GAPDH Forward GACAGTCAGCCGCATCTTCT 20
(housekeeping gene) Reverse GCGCCCAATACGACCAAATC 20
[65]Open in a new tab
Cell Transfection
Three siRNAs targeting SCD and a negative control siRNA (si-NC) were
procured from GenePharma (China). Cell transfection was executed using
jetPRiME (Polyplus, NY, USA) following the manufacturer’s instructions.
Sequences for the siRNAs targeting SCD are provided in Table [66]2.
Table 2.
List of primer sequences for three siRNAs targeting SCD and a negative
control siRNA (si-NC)
Gene Sequence Length (bp)
si-#1 Sense 5’-CUUCGUUUGAAGCAAGAAUTT-3’ 21
Antisense 5’-AUUCUUGCUUCAAACGAAGTT-3’ 21
si-#2 Sense 5’-CAAGUCCUCUACCGAAUGATT-3’ 21
Antisense 5’-UCAUUCGGUAGAGGACUUGT-3’ 20
si-#3 Sense 5’-GAUGAGCAGUCCAAAGCAUTT-3’ 21
Antisense 5’-AUGCUUUGGACUGCUCAUCT-3’ 20
si-NC Sense 5’-UUCUUCGAACGUGUCACGUTT-3’ 21
Antisense 5’-ACGUGACACGUUCGGAGAATT-3’ 21
[67]Open in a new tab
NC Negative control
CCK-8 Assay
Cells were seeded in 96-well plates at a density of 1000 cells per
well. CCK-8 solution (10 μL) at a 1:10 dilution with Dulbecco’s
modified eagle medium (DMEM) (100 μL) was added to each well for
measuring cell viability and proliferation rate. Cell proliferation was
assessed at 0, 24, 48, and 72 h post-seeding using a microplate reader
to measure optical density at 450 nm.
Transwell assay
Cell invasion assays were conducted using 24-well Transwell chambers
(Corning, NY, USA). Approximately 40,000 cells, suspended in serum-free
medium, were placed into the upper chamber, while the lower chamber was
filled with medium containing 20% FBS as a chemoattractant. Migrated
cells were fixed with polyformaldehyde, stained with crystal violet,
and photographed under a fluorescent microscope.
Wound healing assay
Cells were seeded in 12-well plates at a density of 3 × 10^5 cells per
well and grown to confluence. A scratch was made in the monolayer using
a tip, and cells were then washed with PBS to remove detached cells.
Cells were cultured in complete medium. The representative of the
scratch, from the same field, was photographed at 0 and 24 h
post-scratch.
Statistical analysis
Statistical analyses were conducted using R (v. 4.3.2, R Foundation for
Statistical Computing, Vienna, Austria). All p-values were derived from
two-sided tests. Detailed descriptions of the statistical methodologies
are available in Additional file 2.
Comparison of baseline characteristics
Bivariate associations between OS status and patients’ baseline
characteristics in the three datasets were tested. Pearson’s
chi-squared test was used for differential analysis of categorical
variables, and Yates’ continuity correction was applied if at least one
expected cell count was less than 5 but greater than or equal to 1 (for
2 × 2 contingency table only). Fisher’s exact test was used when at
least one expected cell count was less than 1.
Identification of DEGs and co-expressed genes
The FRGs and CRGs are detailed in Additional file 3: Tables S1 and S2.
After running voom on TCGA count matrix, differentially expressed genes
(DEGs) between 17 MIBC biopsies and 17 NAT tissues were detected using
the “limma” package. DEGs were defined based on an absolute
log[2](fold-change) [log[2](FC)] greater than 2 and a false discovery
rate (FDR)-adjusted p-value below 0.001.
Co-expression relationships were assessed using Spearman’s rank
correlation coefficient from the “Hmisc” package [[68]22]. This
analysis was performed on a log[2]-transformed matrix of tumour
samples, with significance defined as a p-value below 0.05 and an
absolute correlation coefficient (|r|) above 0.4.
Establishment of ferroptosis/cuproptosis-related signature
To screen for hub genetic features, univariate and multivariate Cox’s
proportional hazards (PH) regression and machine learning (ML)
techniques were employed to analyse the normalized TPM expression
matrix through a rigorous selection process. The ML methods included
the least absolute shrinkage and selection operator (LASSO)-penalized
Cox’s model, support vector machine-recursive feature elimination
(SVM-RFE), and bidirectional stepwise regression. Details are shown in
Additional file 2.
Expression data of hub genes were used to perform a multivariate Cox’s
regression analysis to compute an individualized genetic risk score for
each patient in the training cohort and test set. The risk score was
calculated using the following equation:
[MATH: Riskscorei=∑j-1n
munderover>βij×exprβij :MATH]
1
where risk score[i] represents the risk score for sample i, j
represents the number of hub gene, β[ij] represents the multivariate
Cox’s regression coefficient value of each hub gene, and expr[ij]
stands for the normalized TPM value of each gene in sample i.
Correlations between the expression levels of hub BMGs and the risk
scores were examined using the “Hmisc” package [[69]22]. Patients were
then categorized into low and high risk score level groups based on the
median value. Expression of hub genes between the two groups was
compared using independent t-test. This method was also applied to
external validation datasets ([70]GSE13507 and [71]GSE32894), adjusting
the cutoff points due to differences in sequencing platform.
Development and validation of nomogram-based prognostic model
Univariate Cox’s regression analysis was utilized to assess the
associations between the ferroptosis/cuproptosis-related signature and
patients’ demographic and clinical characteristics. Significant
variables were then selected for further evaluation using bidirectional
stepwise regression from the “stats” package [[72]23]. TNM stage was
excluded after that to prevent multicollinearity. These significant
predictors, alongside the ferroptosis/cuproptosis-related signature,
informed the development of a nomogram-based prognostic model for
predicting OS.
The associations between the expression level of each hub gene and the
risk score and independent demographic and clinical predictors were
examined. Two-sample independent t-test was employed for variables with
normal distribution, while Wilcoxon’s rank-sum test was utilized for
variables that did not follow a normal distribution. The performance of
the nomogram-based predictive model was assessed using various metrics,
including C-index for discrimination, integrated Brier score (IBS) for
prediction accuracy, and calibration curves for model calibration.
Additionally, decision curve analysis (DCA) [[73]24] was used to
evaluate utility of models.
Construction of biological interaction networks
To seek potential interactions between DEGs, a protein–protein
interaction (PPI) network was constructed using the STRING (Search Tool
for the Retrieval of Interacting Genes/Proteins) database (v. 12.0,
[74]https://string-db.org/) [[75]25]. In the network, the nodes stand
for the proteins and the edges represent the interactions. The minimum
interaction score was set to a medium confidence level of 0.400 to
ensure the reliability of the interactions. Additionally, the STRING
database was used to perform overrepresentation analysis (ORA) using
KEGG pathway gene sets (herein referred to as “ORA–KEGG”). Significant
pathways associated with cancer (FDR-adjusted p < 0.05) were visually
distinguished in the PPI network by assigning distinct colours to the
corresponding nodes for clear annotation. In addition to the PPI
networks, resultant links between pairs of co-expressed DEFRGs and
DECRGs from correlation analysis were integrated to generate a
co-expression network.
Functional enrichment analysis
To evaluate the enrichment of DEGs among functional terms, ORA was
conducted using Gene Ontology (GO) annotation (hereinafter referred to
as “ORA–GO”). The Reduce and VIsualize Gene Ontology (REVIGO) web tool
([76]http://revigo.irb.hr/) was used to remove any redundancy of top 30
significantly enriched GO terms [[77]26]. Based on semantic
similarities, after merging and replacing the representative subset,
non-redundant GO terms were displayed in a semantic space with a cutoff
value C of 0.7.
Moreover, gene set enrichment analysis (GSEA) was carried out on
Hallmark gene sets (hereinafter referred to as “GSEA–Hallmark”) and
KEGG gene sets (hereinafter referred to as “GSEA–KEGG”), with genes
ranked either by log[2](FC) or Spearman’s r for single-gene GSEA. The
top 20 enriched Hallmark terms, ordered by their |enrichment scores|,
were visualized as a facet dot plot using the “ggplot2” package
[[78]27]. The KEGG pathway database comprises a set of pathway maps
drawn by hand. For human (Homo sapiens) species, these maps are divided
into six distinct types: metabolism, genetic information processing,
environmental information processing, cellular processes, organismal
systems, and human diseases [[79]28]. KEGG pathways categorized under
the human diseases category were excluded from this analysis.
Subsequently, the “aPEAR” package [[80]23] was used to leverage
similarities between the significantly enriched KEGG pathways.
Afterwards, the “ggraph” package [[81]29] and “ggforce” package
[[82]30] was utilized to represent the result as a network of
interconnected clusters. Enrichment analysis was conducted utilizing
the “clusterProfiler” package [[83]31]. A functional term or pathway
was deemed statistically significant with an FDR-adjusted p-value less
than 0.25 and p-value less than 0.05.
GSVA
The gene set variation analysis (GSVA) algorithm provided by the “GSVA”
package [[84]32] was used to measure the activity of the top 20
significant gene sets from GSEA–Hallmark (hereinafter referred to as
GSVA–Hallmark). Additionally, the immune infiltration score (IIS) was
determined based on 24 immune cell types, which included various
subsets of macrophages, dendritic cells (DCs), B cells, cytotoxic
cells, eosinophils, mast cells (MCs), neutrophils, nature killer (NK)
cells, and T cells. This score, derived from the research of
Senbabaoglu et al. [[85]33]., aimed to estimate the abundance and
infiltration of these immune cells within the tumour environment. The
GSVA scores for these 24 immune cells were calculated using the
“GSVAutils” package [[86]34]. Furthermore, the antigen processing and
presenting machinery (APM) score was calculated from mRNA expression
levels of APM genes to evaluate the efficiency of antigen processing
and presentation, a critical factor influencing ICI response. Finally,
the tumour immunogenicity score (TIGS) [[87]34] was computed by
integrating the APM score with the tumour mutational burden (TMB),
providing a comprehensive assessment of the tumour’s potential
responsiveness to immunotherapy.
Exploration of gene mutation pattern
Analysing MAF (MAF) files is crucial in cancer genomics to encapsulate
and summarize mutation data across various samples. Oncoplots were
created to visualize the somatic mutation landscape of the top 20
mutated genes within cohorts stratified by risk score level using the
“ggplot2” package [[88]27]. Tumour mutation burden (TMB) was calculated
by summing up all missense, insertion/deletion, and frameshift variants
within each tumour sample using the following equation [[89]34]:
[MATH:
TMB=1nwholeexomemutation38<
mo>+1 :MATH]
2
CNVs are pivotal in the oncogenesis and progression of malignant
tumours [[90]35], necessitating detailed analyses of gene-level and
segmental CNVs across 384 samples. To depict the genomic landscapes of
CNVs and their associations with OS, a lollipop was visualized via the
“ggplot2” package [[91]27] and a circos diagram using the circos tool
(v. 0.69–9; tools version: 0.23) [[92]36]. Moreover, the GISTIC
(Genomic Identification of Significant Targets in Cancer) 2.0 algorithm
[[93]37], provided by the GenePattern tool
([94]https://cloud.genepattern.org/) [[95]38], was applied to pinpoint
significant genomic regions underpinning cancer, highlighting amplified
or deleted segments through GISTIC scores and q-values. These focal
regions were further intersected with the Gencode GTF file
(gencode.v43.annotation.gtf.gz) using Bedtools (v. 2.27.1) [[96]39] to
align genomic data with gene annotations effectively. Gene mapping to
position on the chromosome was performed using the genome reference
consortium human build 38 (GRCh38/hg38).
Analysis of tumour microenvironment
Cells in tumour microenvironment consist of tumour cells, stromal cells
and immune cells [[97]40]. The Estimation of STromal and Immune cells
in MAlignant Tumours using Expression data (ESTIMATE) method was
applied using the “tidyestimate” package [[98]41] to analyse these
components in 384 MIBC samples from the TCGA database. The ESTIMATE
method provided scores reflecting immune and stromal cell infiltration,
overall ESTIMATE scores, and tumour purity estimates. Additionally, the
Cell-type Identification By Estimating Relative Subsets Of RNA
Transcripts (CIBERSORT) algorithm was used to analyse the gene
expression matrix constructed from TPM data, employing the CIBERSORT R
script (v. 1.04, Stanford University, CA, USA) [[99]42] with 1000
permutations, offering a detailed profile of immune cell composition
within the TME.
These scores and immune cell profiles were statistically analysed
between two risk score groups using Wilcoxon’s rank sum test via the
“rstatix” package [[100]43]. The association between expression levels
of hub genes and immune cell fractions differing in two risk score
level groups was assessed using Spearman’s rank correlation analysis
with the help of “Hmisc” package [[101]22]. All statistical analyses
were performed with a significance threshold set at a p-value of less
than 0.05 to ensure robustness of the findings.
Prediction of immunotherapy efficacy and drug activity
Tumours that respond well to ICIs are referred to as immunologically
“hot” tumours, while those that do not respond favourably are termed
immunologically “cold” tumours. The Tumour Immune Dysfunction and
Exclusion (TIDE) score for each sample in the TCGA cohort was
calculated using the TIDE database ([102]http://tide.dfci.harvard.edu/)
[[103]44]. TIDE and TIGS scores were compared between two risk groups
using Wilcoxon’s rank sum test provided by the “rstatix” package
[[104]43]. Correlations between the risk score, immune infiltration
score (IIS), and 17 immune checkpoints (detailed in Additional file 4:
Table S3) were examined using the “correlation” package [[105]45], with
a significance level set at a p-value below 0.05.
Additionally, drug activity prediction was performed based on the data
from the RNAactDrug database
([106]http://bio-bigdata.hrbmu.edu.cn/RNAactDrug), an extensive
resource for exploring drug activity and RNA expression level
correlations, integrating data from GDSC, CellMiner, and CCLE databases
[[107]46]. The analysis focused on the association between RNA
molecules and drug activity, selecting records with an FDR-adjusted
p-value below 0.25 and a p-value less than 0.05. Compounds showing at
least two significant correlations with hub genes were visualized as
bubble plots using the “ggplot2” package [[108]27].
Differential analysis of RT-qPCR data
Gene expression was presented using a modification of the 2^−ΔΔCt
method. To analyse the differential relative expression of the hub
genes and si-SCD in cancer and normal cell lines, p-values from
pairwise t-test for independent groups were calculated and then
adjusted using the FDR method by the “rstatix” package [[109]43]. Ct
values in samples for each mRNA were visualized as a bar plot using the
“ggplot2” package [[110]27]. An FDR-adjusted p-value less than 0.05 was
considered significant for mRNA, and less than 0.0001 for siRNA.
Analysis of repeated-measures data
Linear mixed-effects model (LMM) provided by the “mmrm” [[111]47] was
introduced to fit linear mixed-effects models. Then, “emmeans”
[[112]48] package was utilized to assess the differential impact on
cell viability (%) exerted by three siRNAs targeting SCD in cancer cell
lines against that of si-NC. An FDR-adjusted p-value less than 0.0001
at the time point of 72 indicated a statically significant effect of
the SCD-targeting siRNAs on cell viability compared to the control.
Results
Baseline characteristics of participants
The TCGA cohort consisted of 384 participants diagnosed with MIBC,
including 102 females (26.56%) and 282 males (73.44%). The median age
of the participants was 68.00 [IQR: 60.00, 76.00] y/o. Of these, 213
participants (55.47%) were alive at the time of analysis, while 171
(44.53%) had succumbed to the disease. The analysis of survival rates
revealed no statistically significant difference between female and
male participants (p > 0.05). Additional file 5: Tables S4–6 provide a
comprehensive summary of the demographic and pathological details for
MIBC patients in the TCGA cohort (training set), as well as for the
[113]GSE13507 dataset (test set 1) and the [114]GSE32894 dataset (test
set 2).
Identification of DEFRGs and DECRGs between tumour samples and NATs
Differential expression analysis (DEA) between 17 MIBC specimen and 17
paired NAT tissues from the TCGA cohort revealed 833 down-regulated and
470 up-regulated genes (p < 0.001 and |log[2](FC)|> 2). Out of these,
21 DEFRGs were up-regulated and 15 were down-regulated (Fig. [115]1A
and Additional file 6: Table S7). Three DECRGs exhibited
down-regulation (Fig. [116]1B and Additional file 6: Table S8). In
Figs. [117]1A and 1B, volcano plots depict the results of DEA for FRGs
and CRGs respectively. In Fig. [118]1C, the heatmap colours show the
expression of each DEG across 34 samples.
Fig. 1.
[119]Fig. 1
[120]Open in a new tab
Identification of differentially expressed ferroptosis-related (DEFRGs)
and cuproptosis-related genes (DECRGs). A Volcano plot depicting
DEFRGs. B Volcano plot showing DEFRGs. DEGs (down-regulated and
up-regulated genes) were determined in 17 MIBC specimens vs. 17 paired
normal adjacent tumour (NAT) tissues using a threshold of
|log[2](FC)|> 2 and a false discovery rate (FDR)-adjusted
p-value < 0.001. C Heatmap illustrating expression pattern of 36 DEFRGs
and 3 DECRGs across 17 MIBC specimens and 17 paired NAT tissues. The
rows of the heatmap represent genes, and the columns represent samples.
Each cell is colourized based on the level of expression of that DEFRG
or DECRG in that sample. D Bubble plot showing the GO cluster
representatives in a two-dimensional space. Each circle indicates a
representative cluster. The colour of the circles represents
-log[10](FDR) value of the GO analysis. The size of each circle
indicates the count of genes involved in the GO term. E Co-expression
network of DEFRGs and DECRGs (blue: down-regulated, red: up-regulated,
size: degree; sky blue: negative, tomato red: positive correlation). F
PPI network composed of 37 nodes (proteins) and 75 edges (interactions)
from STRING (confidence: 0.400, p < 0.0001; sky blue halo:
down-regulated, tomato red halo: up-regulated).*** p < 0.001, ****
p < 0.0001
REVIGO was applied to summarize long lists of GO terms from DEGs in the
training cohort. Relevant biological processes identified include
regulation of keratinocyte differentiation, response to hypoxia,
regulation of collagen metabolic process, and positive regulation of
cell cycle G1/S phase transition (Fig. [121]1D).
Spearman’s correlation analysis (Fig. [122]1E and Additional file 7:
Table S9) explored the interconnections among these genes, revealing a
co-expression network of 32 genes, including 29 differentially
expressed FRG (DEFRGs) and 3 differentially expressed CRG (DECRGs). All
correlation pairs exhibited positive correlations, with genes such as
ZEB1 (DECRG, 17 partners) and IL6 (DEFRG, 15 partners) demonstrating
higher connectivity.
The PPIs were analysed for the known and predicted interactions between
DEGs. The PPI network (Fig. [123]1F and Additional file 8: Table S10)
comprised 37 nodes (proteins) and 75 edges (interactions). ATF3 (FRG)
demonstrates the highest number of significant correlations with 10
partners. Notably, the connection between AURKA (DEFRG, 9 partners) and
MYCN (DEFRG, 6 partners) had an exceptionally high combined score,
indicating a potentially crucial role in cellular ferroptosis. Besides,
the interaction between MT1A (DECRG, 1 partner) and MT1M (DECRG, 1
partner) may reflect a co-evolved function in cuproptosis. PPI analysis
also revealed significant enriched KEGG pathways, including the
microRNAs in cancer and transcriptional misregulation cancer.
Selection of hub genes
Univariate Cox’s regression analysis was performed on DECRGs and DEFRGs
in the training set. Genes with statistical significance (p < 0.05) and
a C-index greater than 0.5 were identified as DECRGs and DEFRGs
associated with OS. Ten genes, including AOC3, MEG3, EGR1, SCD, CAV1,
CDO1, CREB5, DDR2, MT1A, and ZEB1, were found to have significant
impacts on OS (Additional file 9: Table S11).
Chromosomal distribution and CNV frequencies of the ten DECRGs and
DEFRGs associated with OS were analysed (Figs. [124]2A and 2B).
Notably, DDR2, located on chromosome 1, had the highest copy number
gain at 66.93% and the lowest loss at 0.78%. CREB5, located on
chromosome 7, also demonstrated a substantial gain of 63.54% with a
loss of 1.30%. Interestingly, MEG3, located on chromosome 14, showed no
copy number variations, indicating stability in its genomic copy
number.
Fig. 2.
[125]Fig. 2
[126]Open in a new tab
Selection of hub genes. A Lollipop chart illustrating CNV frequencies
of DEFRGs and DECRGs in association with overall survival (OS). Bar
length shows copy number gain/loss percentage. B Circos plot
demonstrating chromosomal CNV distribution of 21 DEFRGs and DECRGs in
association with OS. Tracks from outer to inner: genomic locations,
percentage of copy number gain, percentage of copy number loss,
-log[10](p-value) from univariate Cox’s analysis. C Plot indicating the
best penalty parameter (optimal λ = 0.032) selection by
cross-validation (CV) partial likelihood deviance (PLD) of the least
absolute shrinkage and selection operator (LASSO)-penalized Cox’s
regression. D Plots showing LASSO-penalized Cox’s regression
coefficients over different values of ln(λ). E Mean discriminatory
performance of feature subsets of each size selected by linear support
vector machine recursive feature elimination (SVM-RFE) regression
model. CV mean area under the curve (AUC) was plotted against subset
size. The red dot represents that the optimal number of selected
variables is six. F Lollipop plot showing the normalized importance
value of each of the six selected features in the linear SVM model. y/o
Years old
Two ML methods with fivefold cross-validation were used for gene
selection, enhancing robustness and preventing overfitting.
LASSO-penalized Cox’s regression identified nine genes with non-zero
coefficients at the optimal penalty parameter (λ = 0.032)
(Figs. [127]2C and 2D). The SVM-RFE algorithm further refined the
selection to six genes based on their maximal mean AUC value
(Figs. [128]2E and 2F). The genetic features identified by both ML
methods were subsequently considered as candidates in a multivariate
Cox’s PH regression analysis. Bidirectional stepwise selection
identified SCD, DDR2, and MT1A as hub genes, which are the most
contributive predictors for OS (Additional file 10: Table S12).
Establishment of ferroptosis/cuproptosis-related prognostic signature
The development of a ferroptosis/cuproptosis-related prognostic
signature was confirmed through a multivariate Cox’s PH model using
data from the TCGA cohort. The risk score calculation was based on a
linear weighted combination of hub gene expression levels:
Risk score = (0.18362 × SCD expr) + (0.16029 × DDR2
expr) + (0.12655 × MT1A expr). The multivariate Cox’s regression
coefficients for each hub gene were displayed using a forest plot in
Fig. [129]3A. The links between risk score and gene expression were
visualized through circos charts in Fig. [130]3B. All of three hub
genes had a strong correlation with the risk score.
Fig. 3.
[131]Fig. 3
[132]Open in a new tab
Establishment of the ferroptosis/cuproptosis-related signature and
validation of its prognostic value. A Bar plot showing multivariate
Cox’s regression coefficient for each hub gene. B Chord diagram of
Spearman’s correlation between expression level of hub genes and risk
score. The distribution of each patient’s risk score ordered from low
to high in (C) TCGA cohort, (H) [133]GSE13507 dataset, and (M)
[134]GSE32894 dataset. Patients in each dataset were divided into two
risk score level groups based on each dataset’s median. Scatter diagram
of overall survival (OS) time against patients’ rank of risk score in
(D) TCGA cohort, (I) [135]GSE13507 dataset, and (N) [136]GSE32894
dataset. Boxplot depicting the expression level of each hub gene was
significantly different between the two risk score level groups in (E)
TCGA cohort, (J) [137]GSE13507 dataset, and (O) [138]GSE32894 dataset.
The independent sample t-test was applied for comparing the
differences, and the p-values were FDR-adjusted. Low risk score level
tissues marked in blue colour and high risk score level samples marked
in red. Kaplan–Meier (K–M) plot demonstrating elevation in OS
probability in (F) TCGA cohort, (K) [139]GSE13507 dataset, and (P)
[140]GSE32894 dataset. Plots of the time-dependent receiver-operating
characteristics (ROC) curves for the risk score prognostic model for
1-, 3-, and 5-year OS and the corresponding area under the curve (AUC)
values in (G) TCGA cohort, (L) [141]GSE13507 dataset, and (Q)
[142]GSE32894 dataset. (R) Plots of the time-dependent ROC curves for
the risk score prognostic model in each age and gender subgroups of the
TCGA cohort for 1-year and the corresponding AUC values. Each AUC value
is represented in the legend as the estimated value [95% CI]. *
p < 0.05, *** p < 0.001, **** p < 0.0001
Performance evaluation of the prognostic signature
The performance evaluation of the prognostic signature was conducted
across multiple datasets, including TCGA and two GEO datasets
([143]GSE13507 and [144]GSE32894). The distribution of risk scores is
presented in an ascending order, aligned with patient rankings in the
training set (Fig. [145]3C) and the two test sets (Figs. [146]3H, and
3M). Risk score cutoff was set based on the median risk score of each
cohort, classifying samples into low and high risk score level groups.
Samples with a risk score below the threshold were categorized into the
low risk score level group, and those above into the high risk level
group. The model’s discrimination capability between different OS
outcomes was effectively showcased through scatter diagrams, which
plotted the OS-time against patients’ risk score rankings. This
demonstrated strong discrimination within the TCGA cohort, as shown in
(Fig. [147]3D), and was further validated in the datasets represented
in (F[148]igs. [149]3I and 3N). Significant differences in the
expression levels of the three hub genes between high and low risk
score level groups were consistently observed across all datasets, as
shown in Figs. [150]3E, 3J, and 3O.
K–M survival analysis confirmed the effectiveness of the risk score
level in predicting survival across all datasets (Figs. [151]3F, 3K,
and 3P). The performance of the univariate model incorporating risk
score performance was further assessed using time-dependent ROC curves
(Figs. [152]3G, 3L, and 3Q), demonstrating strong predictive efficacy.
Additionally, this signature also exhibited good discriminatory ability
across demographic subgroups of MIBC patients, as illustrated in
Fig. [153]3R.
Development and validation of nomogram-based model
In the establishment and validation of a nomogram-based model,
univariate and bidirectional stepwise multivariate Cox’s PH regression
analyses identified variables significantly associated with OS. The
multivariate analyses revealed that older age (≥ 60 y/o vs. < 60 y/o,
HR = 1.608 [95% CI: 1.030–2.512], p = 0.0367), higher T stage (High vs.
Low, HR = 1.531 [95% CI: 1.068–2.194], p = 0.0205), lymph node
metastasis (Positive vs. Negative, HR = 1.776 [95% CI: 1.301–2.423],
p = 0.0003), and high risk score level (High vs. Low, HR = 1.655 [95%
CI: 1.211–2.261], p = 0.016) were significantly associated with worse
OS in the TCGA cohort. The results of univariate and multivariate Cox’s
PH regression analyses are summarized in Fig. [154]4A.
Fig. 4.
[155]Fig. 4
[156]Open in a new tab
Development, and internal and external validation of nomogram-based
prognostic model incorporating the ferroptosis/cuproptosis-related
signature. A Forest plot of uni- and multi multivariate Cox’s model for
overall survival (OS) based on the TCGA cohort (training set). B A
nomogram for predicting 1, 3, and 5-year OS possibilities in the MIBC
patients relying on the TCGA population. The steps for using the
nomogram are (1) determine the individual patient’s point for each
predictor (T stage, Lymph node status, and risk score level), (2) draw
a straight line upwards from each predictive value to the top point
reference line, (3) sum the points from each predictive variable, (4)
locate the sum on the total points reference line, and (5) draw a
straight line from total points line down to the bottom probability
lines to obtain the patient’s likelihood of 1-, 3-, and 5-year OS. The
diagram also prints how many linear predictor units there are per point
and the number of points per unit change in the linear predictor (lp).
Calibration curves of Kaplan–Meier (K–M) vs nomogram predicted (C) 1-,
(D) 3-, and (E) 5-year OS for TCGA cohort (red) and [157]GSE13507
dataset (green). (F) Time-dependent c-index curves for the “Clinical”,
“Risk Score Level”, and “Nomogram” models. DCA for the “Clinical”,
“Risk Score Level”, and “Nomogram” models built to predict (G) 1-, (H)
3-, and (I) 5-year OS probability based on records of patients in TCGA
cohort (Orange bar: range where the Nomogram model performs better
others, deep blue bar: range not relevant to the Nomogram model). ns
not significant, * p < 0.05, ** p < 0.01, *** p < 0.001, ****
p < 0.0001
A nomogram (Fig. [158]4B) was developed utilizing these independent
prognostic factors to predict 1-, 3-, and 5-year OS probabilities for
MIBC patients. This nomogram demonstrated excellent discrimination and
calibration in both training and test datasets, with a C-index of 0.660
[95% CI: 0.639–0.681] for the training cohort and an integrated Brier
score (IBS) of 0.206 [95% CI: 0.185–0.227]. The performance in the test
dataset was consistent, with a C-index of 0.752 [95% CI: 0.724–0.781]
and an IBS of 0.166 [95% CI: 0.151–0.181]. The 1000-resampling
calibration plots for the training cohort and validation set 1
([159]GSE13057) at 1, 3, and 5 years demonstrated minimal deviations
from the 45° reference line, indicating robust model performance
(Figs. [160]4C–E).
Further internal and external validation showed that the nomogram
provided better predictive accuracy than other established Cox’s PH
regression models. The time-independent C-index of the nomogram model
consistently outperformed other models at any time point, indicating
superior discrimination (Fig. [161]4F). Decision curves confirmed that
the nomogram model had the highest net benefit for predicting 1-, 3-,
and 5-year OS probability, surpassing all other options
(F[162]igs. [163]4G–I). The orange bar in each decision curve analysis
(DCA) plot represents a threshold probability range where the nomogram
model outperforms other models, disregarding random noise [[164]24].
The differential analysis performed on the TCGA cohort, investigating
the disparities in risk scores and the expression of hub genes across
demographic and clinical subgroups, yielded substantial findings
(Additional file 11: Tables S13–15). Firstly, gender did not
significantly impact the risk score or gene expression levels. In
contrast, age demonstrated a marked influence, with patients under 60
showing lower risk score levels and reduced expression of SCD and MT1A
genes compared to those aged 60 and above. Higher T stage was
associated with elevated risk scores and increased expression of DDR2
and MT1A in advanced T stages. Lastly, the presence of lymph node
metastasis correlated with elevated risk scores and DDR2 expression but
not with SCD and MT1A.
Enriched Hallmark Terms and KEGG Pathways
Figure [165]5A shows the top 20 enriched terms from the GSEA-Hallmark
analysis (FDR-adjusted p < 0.25 and p < 0.05). All of these terms were
activated in the high risk score level group. The GSVA method was used
to estimate the pathway activity by transforming the input gene-sample
expression data matrix into a corresponding gene set-sample expression
data matrix (i.e., pathway expression score matrix). Figure [166]5B is
the heatmap illustrating the GSVA scores of the top 20 enriched
Hallmark pathways across 384 TCGA samples. Both figures indicate the
upregulation of these terms in relation to higher risk of death for
MIBC patients. Among them, angiogenesis, IL6-JAK-STAT3, IL2/STAT5,
complement system, interferon gamma response, and G2M checkpoint, are
crucial for tumour growth, inflammation, and immune system evasion.
Fig. 5.
[167]Fig. 5
[168]Open in a new tab
Enriched gene sets associated with risk score level in the gene set
enrichment analysis (GSEA) performed on TCGA data. A Dot plot depicted
the 20 most significantly enriched Hallmark terms ranked by gene ratio.
Dot size is proportional to the number of overlapping genes. p-values
are colour-coded according to the colour scale. B Heatmap plots of gene
set variation analysis (GSVA) scores across 384 TCGA samples for the 20
most significantly enriched Hallmark gene sets. Red label: activated
Hallmark terms. C Clustering network of significantly enriched KEGG
pathways in the GSEA analysis. The nodes represent the significant KEGG
pathways and the edges represent similarity between them and are
coloured by normalized enrichment score (NES). The lines connected
similar pathways are coloured by similarity
The clustering of significant pathways from the GSEA–KEGG in
Fig. [169]5C has identified several groups based on their functional
similarities. This analysis highlights the interconnected nature of
biological pathways, underscoring how various aspects of cellular
functioning are closely related. For instance, pathways involving
PI3K-Akt signaling, ECM-receptor interactions, and focal adhesions
cluster together, suggesting a shared role in processes such as cell
migration, proliferation, and survival. Similarly, the close grouping
of the cytokine-cytokine receptor interaction, chemokine signaling
pathway, and viral protein interactions with cytokines and their
receptors indicates a concerted involvement in immune response and
inflammation.
Somatic mutation and segmental CNV in patients with different risk score
levels
In the low risk score level group, 93.75% samples displayed somatic
mutation (Fig. [170]6A). The most frequently mutated gene was TP53, a
well-known tumour suppressor gene, followed by TTN. Other significant
genes affected by somatic mutation include KMT2D, KDM6A, and MUC16,
which are involved in chromatin modification, histone demethylation,
and cell adhesion, respectively. In the high risk score level group,
genetic changes were evident in 92.19% of the samples (Fig. [171]6B).
These mutations encompassed missense mutations, nonsense mutations,
splice site alterations, as well as frame shift insertions and
deletions. The TP53 gene had the highest frequency of mutation,
followed by TTN and ARID1A. Other genes that frequently underwent
mutations include MUC16, KMT2D, KDM6A, PIK3CA, and RB1.
Fig. 6.
[172]Fig. 6
[173]Open in a new tab
Somatic mutation and copy-number alteration in association with risk
score level. A MAF mutation oncoplot showing top 20 altered in 180
(93.75%) of 192 low risk score level samples from TCGA cohort. B MAF
mutation oncoplot showing top 20 altered in 177 (92.19%) of 192 high
risk score level samples from TCGA cohort. C GISTIC amplification (up,
red) and deletion (down, steel blue) plot of 191 TCGA low risk score
level samples. D GISTIC amplification (up, red) and deletion (down,
steel blue) plot of 189 TCGA high risk score level samples
The data from chromosomal CNVs for both high and low risk score level
groups reveal distinct genomic alterations. Participants in the low
risk score level group primarily displayed deletions, including genes
such as PEX14, RPS6KA1, and ARID1A (Fig. [174]6C and Additional file
12: Table S16). Conversely, patients in the high risk score level group
exhibited a mix of deletions and amplifications across various
chromosomal segments, including genes like SLC9A1, HSPA5P1, and MACF1
(Fig. [175]6D and Additional file 12: Table S17).
Immune cell landscape and immunotherapy response prediction
Differential analysis was conducted on the stromal score
(Fig. [176]7A), immune score (Fig. [177]7B), ESTIMATE score
(Fig. [178]7C), and tumour purity (Fig. [179]7D) calculated using the
ESTIMATE algorithm. The high risk score level group exhibited
significantly increased levels of stromal score, immune score, and
ESTIMATE score. Conversely, tumour purity was observed to be lower in
this group.
Fig. 7.
[180]Fig. 7
[181]Open in a new tab
Immune cell landscape. Boxplot reporting the different distributions of
(A) stromal score, (B) immune score, (C) Estimation of STromal and
Immune cells in MAlignant Tumours using Expression data (ESTIMATE)
score, and (D) tumour purity between high (n = 192, yellow) and low
risk score level groups (n = 192, green). E Bar plot showing
composition of 22 infiltrating immune cells in two risk score level
subgroups. Fraction values of CIBERSORT immune cells were determined
for each patient; each bar represents one patient. F Box plot showing
the differences in CIBERSORT fractions between tissues from the high
risk score level patients (n = 192, yellow) and low risk score level
participants (n = 192, green). G Heatmap showing the Spearman’s
correlation coefficient between the expression of the three hub genes
and immune cell fractions differing in two risk score level groups. ns
not significant, * p < 0.05, ** p < 0.01, *** p < 0.001, ****
p < 0.0001
CIBERSORT was also applied to the gene expression matrix in order to
infer the relative abundance of 22 tumour-infiltrating immune cells for
each sample in the TCGA cohort. Significant differences in the
fractions of various immune cells were also observed between the two
risk score level groups (Fig. [182]7E). Notably, the high risk score
level group exhibits a significantly lower proportion of B cells
memory, T cells follicular helper, T cells regulatory (Tregs), T cells
gamma delta, NK cells activated, monocytes, DCs resting, DCs activated,
and eosinophils (all p < 0.05). Conversely, a higher proportion of B
cells naïve, T cells CD4 memory activated, macrophages M0, macrophages
M1, macrophages M2, MCs activated, and neutrophils (all p < 0.05).
The correlation analysis between gene expression and CIBERSORT immune
cell fractions reveals complex interactions within the tumour
microenvironment (Fig. [183]7F). The correlation analysis between gene
expression and immune cells reveals significant interactions for hub
genes. For SCD, notable correlations include a positive correlation
with macrophages M0 and MCs activated (all p < 0.05), and negative
correlations with monocytes, NK cells activated, T cells gamma delta,
and T cells regulatory (Tregs) (all p < 0.05). For DDR2, significant
positive correlations are seen with B cells naïve, macrophages M0,
macrophages M1, macrophages M2, neutrophils, and T cells CD4 memory
activated (all p < 0.05), while negative correlations include B cells
memory, DCs activated, DCs resting, Monocytes, NK cells activated, and
T cells follicular helper (all p < 0.05). For MT1A, positive
correlations are found with B cells naïve, macrophages M0, macrophages
M1, Macrophages M2, MCs activated, neutrophils, and T cells CD4 memory
activated (all p < 0.05), while negative correlations are observed with
B cells memory, DCs activated, DCs resting, eosinophils, NK cells
activated, T cells follicular helper, T cells gamma delta, and Tregs
(all p < 0.05).
Additionally, the risk score positively correlated with several immune
checkpoint molecules (IDO1, TNFRSF18 [GITR], TNFRSF4 [OX40], PDCD1
[PD-1], CD274 [PD-L1], LAG3, CTLA4 [CD152], CD27 [TNFRSF7], CD86
[B7-2], TNFRSF9 [CD137], PVR [CD155], CD28, HAVCR2 [TIM-3], and
PDCD1LG2 [PD-L2]) (Fig. [184]8A) and IIS (Fig. [185]8B). The
correlation analysis also revealed a positive association between the
IIS and tumour purity (Fig. [186]8C). Meanwhile, the differential
analysis revealed that the low risk score level group displayed lower
TIDE and TIGS scores (Figs. [187]8D and E).
Fig. 8.
[188]Fig. 8
[189]Open in a new tab
Spearman’s correlation analysis and differential analysis with regards
to immune checkpoint inhibitor (ICI) therapy response prediction and
tumour immune microenvironment. A Lollipop plot showing immune
checkpoints that significantly correlate with risk score in TCGA
patients. Scatter plot showing (B) risk score and (C) tumour purity,
and immune infiltration score (IIS) correlate among TCGA patients.
Beemswarm plot showing significant differences in (D) Tumour
immunogenicity score (TIGS) and (E) Tumour Immune Dysfunction and
Exclusion (TIDE) score between the high risk score level (n = 192,
yellow) and low risk score level groups (n = 192, green). * p < 0.05,
** p < 0.01, *** p < 0.001, **** p < 0.0001
Candidate chemical drugs
The study extracted statistically significant correlations between
specific chemical compounds and RNA molecules from the RNAactDrug
database, which have implications for chemical drug activity
(Fig. [190]9). The data primarily originated from the GDSC database,
with an exception for PF2341066, where information was derived from the
CCLE. Noteworthy associations were found between the chemotherapy agent
5-fluorouracil and RNA molecules DDR2 and MT1A, suggesting a potential
link in drug response. Similarly, the compound AR-42 exhibited
significant correlations with MT1A and SCD RNA molecules. The histone
deacetylase inhibitor belinostat (PXD101) [[191]49] and PDK1 inhibitor
BX-912 also displayed correlations with these RNA molecules. The
microtubule inhibitor Docetaxel and the kinase inhibitor FR-180204 were
significantly associated with DDR2 and SCD, as well as DDR2 and MT1A,
respectively. Innovative compounds, such as Genentech Cpd 10 and
GSK1070916, along with the PI3K inhibitor idelalisib, showed
significant associations with MT1A and SCD. Notably, PF2341066
demonstrated significant correlations with MT1A and SCD.
Fig. 9.
Fig. 9
[192]Open in a new tab
Bubble plot illustrating the correlation between hub genes (y-axis) and
compounds (x-axis). Each point’s size is proportional to the false
discovery rate (FDR), while the colour gradient indicates the
Spearman’s r, representing the correlation between the two variables.
The correlation data between drug activity and mRNA expression in cell
lines was sourced from RNAactDrug
Biological experiments and single-gene GSEA
The expression patterns of SCD, DDR2, and MT1A genes in both normal and
cancerous human urothelial cells were investigated based on
differential analysis of data from the qRT-PCR test. The results
revealed that the expression level of SCD was significantly elevated in
the T24 and UM-UC-3 cell lines compared to SV-HUC-1 (Fig. [193]10A). In
contrast, the expression levels of DDR2 (Fig. [194]10B) and MT1A
(Fig. [195]10C) were lower in the T24 and UM-UC-3 cell lines than in
SV-HUC-1. These RT-qPCR results align completely with the differential
expression patterns observed between MIBC samples and NAT tissues in
the TCGA cohort.
Fig. 10.
[196]Fig. 10
[197]Open in a new tab
Biological experimental validation and the impact of SCD on cell
proliferation, migration, and invasion of human bladder cells. A–C Bar
plot showing the relative expression level of the three hub genes in a
normal cell line (SV-HUC-1) and two bladder cancer cell lines (T24 and
UM-UC-3). (A) SCD was up-regulated in two cancer cell lines compared to
the normal cell line, while (B) M1TA and (C) DDR2 were down-regulated.
The RNA transcription levels of hub genes were evaluated by using the
2.^−ΔΔCt method. GAPDH was used as an internal control. Time course of
(F) T24 and (G) UM-UC-3 cell viability after acute treatment with
CCK-8. Morphology of migrating (H) T24 and (I) UM-UC-3 cells after
transfecting siRNA#1, siRNA#2, siRNA#3, and si-NC from the microscopy
of the Transwell assay. Fluorescent microscope photos to evaluate wound
healing in vitro in the scratch assay using (J) T24 and (K) UM-UC-3
cells after transfecting siRNA#1, siRNA#2, siRNA#3, and si-NC.
Knockdown of SCD may inhibit the proliferation, invasion, and migration
of bladder cancer cells. Error bars indicate SD. * p < 0.05,
** p < 0.01, *** p < 0.001, **** p < 0.0001
In MIBC tissues from the TCGA cohort, single-gene GSEA–Hallmark
(Additional file 1: Figure S2a) and GSEA–KEGG (Additional file 1:
Figure S2b) analyses for the SCD gene revealed significant activation
of multiple pathways, including E2F targets, G2M checkpoint, mTORC1
signaling, MYC targets, unfolded protein response, protein secretion,
cholesterol homeostasis, oxidative phosphorylation, PI3K-Akt-mTOR
signaling, lipid metabolism, and glycolysis. Notably, both analyses
highlighted the enrichment of the mTOR signaling pathway, fatty acid
metabolism, protein processing/secretion, and cell cycle regulation. To
further explore the role of SCD in UC, RNA interference experiments
targeting SCD were performed on the T24 and UM-UC-3 cell lines. The
efficiency of gene silencing was verified by RT-qPCR, demonstrating
that three distinct siRNA sequences successfully knocked down the
expression of SCD. The impact of SCD knockdown on cellular
proliferation was assessed using CCK-8 assays. The results indicated a
significant reduction in cell proliferation rates in both T24
(Figs. [198]10D and F) and UM-UC-3 cell lines (Figs. [199]10E and G)
following SCD silencing. Additionally, the effects of SCD knockdown on
cell migration abilities were evaluated through Transwell and scratch
assays. In both the T24 (Figs. [200]10H and J) and UM-UC-3 cell lines
(Figs. [201]10I and K), the migratory capacity of cells was
significantly impaired after SCD silencing, suggesting a critical role
of SCD in the regulation of cell motility in UC.
Discussion
Through comparative analysis of MIBC samples and paired NAT tissues, 36
DEFRGs and 3 DECRGs were identified. GO enrichment analysis was
performed on these DEGs. The results in Fig. [202]1D display
significant key biological processes shared by DEGs, which underline
the roles of cell death mechanisms, cell proliferation,
differentiation, and tumour microenvironment adaptation in MIBC’s
progression.
The interplay between ferroptosis and cuproptosis, two recently
identified forms of non-apoptotic cell death, has become an
increasingly prominent topic in the field of cancer research [[203]13,
[204]15, [205]50]. Spearman’s correlation analysis revealed that all
three DECRGs were significantly correlated with at least one DEFRG in
MIBC. Additionally, DEFRGs and DECRGs were involved in some KEGG
pathways associated with cancer.
Disruptions in their metabolic functions can lead to lethal
consequences for cancer cells, triggering both ferroptosis and
cuproptosis [[206]50]. The mechanism of copper-dependent cell death is
closely linked to mitochondrial activity. Cuproptosis often occurs when
copper binds to lipoylated proteins in the TCA cycle, leading to
protein aggregation, loss of iron-sulfur cluster proteins, and
proteotoxic stress, ultimately causing cell death [[207]11, [208]51,
[209]52]. In addition, a connection between mitochondrial copper
accumulation and the initiation of ferroptosis has been established.
The intricate interaction mechanisms between ferroptosis and
cuproptosis in MIBC indeed call for further investigation through
experimental studies [[210]50].
The application of multiple ML techniques and Cox’s regression analysis
identified SCD, DDR2, and MT1A as hub genes for MIBC patients. SCD,
also known as SCD1, is a key player in lipid metabolism. The regulation
of the saturated fatty acids (SFAs) to monounsaturated fatty acids
(MUFAs) ratio by SCD1 is not only vital for cellular homeostasis but
also implicates its role in cell proliferation and survival [[211]53].
Lipid metabolic reprogramming is considered a hallmark of cancer, and
the role of SCD reveals the potential of targeting ferroptosis, a
regulated cell death process, as a therapeutic strategy [[212]54,
[213]55]. Experimental evidence shows that targeting SCD with siRNA
significantly reduces the proliferation and migration of bladder cancer
cells, further confirming its role in cancer progression [[214]56].
DDR2 is critical in bladder cancer progression, affecting
proliferation, migration, invasion, metastasis, EMT, and chemotherapy
resistance, and its abnormal expression and mutations are linked to
aggressive cancer phenotypes, highlighting its potential as a
therapeutic target [[215]57, [216]58]. MT1A, a member of the
metallothionein (MT) family, is involved in tumour development,
progression, and drug resistance, essential for metal homeostasis and
cellular stress protection, with its variable expression across cancer
types indicating its potential as a biomarker for cancer diagnosis and
prognosis, as well as a target for therapeutic intervention [[217]59].
By utilizing the multivariate Cox’s regression coefficients for the
three hub genes and their expression data, a risk score was developed
to predict the survival probability of MIBC patients. The results from
the two validation sets also confirmed that this variable exhibits good
discriminative ability across different datasets. Subsequently, a
prognostic model incorporating the ferroptosis/cuproptosis-related
signature was developed. The internal and external validation indicated
that this model can accurately distinguish between patients with high
and low risk score levels. The ability to predict survival probability
of the nomogram-based model and its robust performance were also
confirmed. As such, the model can be used as an effective tool to
assist clinicians in assessing the prognostic risk of MIBC patients.
The differential analysis revealed a significant correlation between
elevated SCD expression and more advanced T stages in MIBC patients
from the TCGA database. This finding gains further support from the
results of single-gene GSEA–KEGG and GSEA–Hallmark analyses, which
revealed a significant enrichment of genes associated with SCD
expression in fatty acid metabolism pathways. This enrichment implies
that SCD upregulation goes beyond a simple correlation, indicating an
active role in tumourigenesis by influencing lipid metabolism in a
manner conducive to cancer proliferation.
In the TCGA cohort, both single-gene GSEA–Hallmark and GSEA–KEGG
analyses suggest that the SCD gene may influence the pathogenesis and
progression of MIBC by modulating cell cycle, lipid metabolism, and
immune response. This emphasizes the potential of SCD upregulation to
not only alter cellular metabolic processes but also engage in
signaling cascades that are quintessential for cancer progression.
SNORD88C acts as a non-invasive diagnostic biomarker for non-small cell
lung cancer (NSCLC), promoting cancer progression by enhancing SCD1
translation through specific RNA modifications, which leads to
inhibited autophagy and increased tumour growth and metastasis
[[218]60]. Furthermore, another line of research has interpreted the
role of FGFR3 in bladder tumours. The increased activity of FGFR3
augments the demand for fatty acid desaturation, a fundamental process
for maintaining membrane fluidity and effective cell signaling. This
increased requirement is satisfied through the signaling via the
PI3K-mTORC1 pathway, resulting in the upregulation of SREBP1 and,
consequently, SCD [[219]61].
The pathway enrichment analysis further elaborated on the biological
significance of the identified DEGs between the two risk score level
groups, revealing their involvement in critical pathways that govern
cancer progression and response to therapy. Notably, pathways
associated with inflammation and immune response were prominently
enriched among these genes, highlighting their integral roles in
modulating the cellular mechanisms central to MIBC pathogenesis. The
results of enrichment analyses provide a molecular blueprint, offering
the potential of targeting these pathways for therapeutic intervention.
At the same time, CNV analysis not only highlighted the mutational
landscape of these critical genes but also underscored the genetic
underpinnings that may contribute to the aggressive nature of MIBC.
Comparing the somatic mutation landscape of the two risk score level
groups, TP53 mutations are predominant in both, suggesting its
fundamental role in the disease regardless of risk level. The presence
of additional mutations in genes such as ARID1A, PIK3CA, and RB1 in the
high risk group may provide insights into potential therapeutic targets
or biomarkers for stratifying patients based on genetic risk. The
GISTIC scores further reveal the potential involvement of these genomic
regions in cancer pathogenesis. The segmental deletions in the tumour
samples with high risk score level suggest a loss of function in
pathways associated with a more aggressive cancer phenotype when
intact. The segmental CNVs in the tumour samples with high risk score
level may indicate a more aggressive tumour phenotype or reflect the
genomic instability characterizing the high risk score level group.
Stratifying patients into high and low risk score level groups based on
the ferroptosis/cuproptosis-related signature not only provides a wider
perspective of the tumour’s biological behaviours and heterogeneity in
mutational landscape, but also guides tailored therapeutic
interventions. Tumour purity refers to the proportion of cancer cells
within a given tumour tissue sample. Accurate assessment of tumour
purity is essential for understanding the respective roles of malignant
and non-malignant cells within the tumour microenvironment [[220]62].
Decreased tumour purity in the high risk score level group suggested
the samples in this group had a higher proportion of non-tumour cells
within their microenvironment. Low tumour purity was also associated
with worse prognosis in MIBC [[221]63]. Research has shown low purity
tumours may signify chromosomal amplifications or deletions. Moreover,
cancer patients with low tumour purity may not benefit from adjuvant
chemotherapy [[222]64].
As indicated in the immune infiltration differential analysis,
different tumour immune microenvironments might influence tumour
progression and patient prognosis. Tumours with high risk score level
show decreased proportions of memory B cells, Tregs, T cells gamma
delta, activated NK cells, and various DCs, indicating reduced immune
surveillance. Meanwhile, these samples also exhibit increased naïve B
cells, activated CD4 memory T cells, all macrophage subtypes, activated
MCs, and neutrophils, suggesting a pro-tumorigenic environment.
Gene-immune cell correlation analysis highlights the roles of SCD,
DDR2, and MT1A in modulating immune cell infiltration. SCD correlates
with increased macrophages and activated mast cells but decreased
monocytes and NK cells. DDR2 and MT1A similarly influence a wide range
of immune cells. The three hub genes may promote a pro-tumourigenic and
immunosuppressive microenvironment.
The risk score was found to positively correlate with several immune
checkpoint molecules, like PD-1, PD-L1, and CTLA-4. This suggests that
a higher risk score might be indicative of increased immune checkpoint
activity, potentially leading to immune evasion by tumours. While high
expression of immune checkpoints often associates with poor prognosis
for cancer patients [[223]65, [224]66], this sign can indicate
potential responsiveness to ICI [[225]16, [226]17]. Moreover,
innovative strategies such as the use of reactive oxygen species
(ROS)-responsive nanoparticles combined with copper and elesclomol to
induce cuproptosis, in conjunction with αPD-L1, have shown enhanced
cancer immunotherapy outcomes, further emphasizing the importance of
targeting immune evasion mechanisms in cancer treatment [[227]67].
IIS, which quantifies the extent and type of immune cell infiltration
within the tumour, was positively correlated with risk score in MIBC.
Tumours with higher risk score level not only tended to possess
increased expression of immune checkpoints, but also exhibited a more
substantial immune cell proportion. This dual characteristic might
indicate a complex immune landscape where the immune system is actively
engaged yet simultaneously suppressed by the tumour’s immune evasion
strategies. The low risk score level group has lower TIGS and TIDE
scores compared to another group. Lower TIGS score reflects a lower
level of tumour immunogenicity, which has been associated with better
survival outcomes in patients not treated with immunotherapy [[228]34].
On the other side, positive TIDE score signifies ICI non-response
[[229]34]. It can be concluded that patients in the low risk score
level group may have a more favourable immunological profile for
responding to ICI therapy.
Through analysis with the RNAactDrug database, some chemotherapeutic
drugs were found to be significantly associated with the expression
levels of hub genes, offering targeted strategies for the potential
treatment of MIBC. Emerging biomarkers and potential therapeutics
related to cuproptosis and ferroptosis in cancer provide new
enlightenment for treatment. A recent study [[230]68] has demonstrated
that inhibiting SCD can trigger both ferroptosis and apoptosis in
ovarian cancer cells. This dual mechanism presents a significant
barrier to the development of drug resistance, making SCD a promising
target for anti-cancer therapy [[231]68]. In addition, since cancer
cells have a higher demand for copper compared to normal cells,
targeting copper metabolism and cuproptosis-related pathways could
selectively kill cancer cells without harming normal cells. Potential
therapeutic approaches could involve utilizing copper ion carriers or
chelators to regulate cellular copper levels in cells, inducing
cuproptosis in cancer cells while sparing healthy cells.
Moreover, the results of differential relative expression of hub genes,
SCD, DDR2, and MT1A, in bladder cancer cells compared to normal
urothelial cells from RT-qPCR experiments were consistent with those of
DEA on transcriptomic data from the TCGA cohort. The experimental
validation of the expression patterns of the three hub genes confirms
the reproducibility of the molecular alterations associated with
bladder cancer.
The precise molecular mechanisms and interactions of these hub genes in
MIBC remain a critical knowledge gap. Therefore, it is essential to
further elucidate the detailed molecular pathways associated with these
hub genes and investigate their potential as biomarkers for early
diagnosis and treatment response. Additionally, exploring synergistic
treatment combinations that integrate traditional therapies with
targeted approaches against FRGs and CRGs could enhance therapeutic
efficacy. The advancements in multi-omics technologies and the
development of novel targeted drugs hold promise for revolutionizing
MIBC treatment, making it more personalized and effective.
While this investigation reveals the significance of FRGs and CRGs in
the progression of MIBC, it is not without limitations. The molecular
heterogeneity of MIBC poses a challenge in identifying uniform
therapeutic targets across different subtypes. Additionally, the
utilization of public databases introduces variability in data quality
and completeness, potentially impacting study findings. Moreover, this
study was limited to experiments at the cell line level. Further
research involving human clinical samples is necessary to
comprehensively validate the expression pattern of the hub genes.
Larger, multicentric studies with diverse MIBC patient cohorts are also
needed to confirm the prognostic utility of hub genes. Additionally,
exploring the influence of the tumour microenvironment on treatment
response in different risk score level groups is crucial. Furthermore,
this study did not delve deeply into the roles of hub genes in
mediating cuproptosis and ferroptosis in MIBC. Future research should
include more in-depth biological experiments to investigate these
mechanisms.
Conclusions
This study identifies three hub ferroptosis/cuproptosis-related genes
and develops a prognostic signature based on these genes. Additionally,
it proposes a nomogram-based model to predict OS probability for MIBC
patients. The established prognostic model, along with analysis of
mutational landscape, provides a valuable tool for clinical
decision-making. Furthermore, the exploration of immune cell
infiltration and immune checkpoint expression reveals the
ferroptosis/cuproptosis-related signature’s value in predicting
responses to immunotherapy and drug therapy. This represents a
significant step forward in the personalized treatment of MIBC,
offering hope to patients facing this aggressive disease.
Supplementary Information
[232]Supplementary Material 1.^ (297.3KB, docx)
[233]Supplementary Material 2.^ (44KB, docx)
[234]Supplementary Material 3.^ (25.9KB, xlsx)
[235]Supplementary Material 4.^ (10.2KB, xlsx)
[236]Supplementary Material 5.^ (16.1KB, xlsx)
[237]Supplementary Material 6.^ (72.4KB, xlsx)
[238]Supplementary Material 7.^ (13.7KB, xlsx)
[239]Supplementary Material 8.^ (13.7KB, xlsx)
[240]Supplementary Material 9.^ (10.1KB, xlsx)
[241]Supplementary Material 10.^ (9.6KB, xlsx)
[242]Supplementary Material 11.^ (15.4KB, xlsx)
[243]Supplementary Material 12.^ (407.8KB, xlsx)
Acknowledgements