ArticlePDF Available

Screening and predicted value of potential biomarkers for breast cancer using bioinformatics analysis

Springer Nature
Scientific Reports
Authors:

Abstract and Figures

Breast cancer is the most common cancer and the leading cause of cancer-related deaths in women. Increasing molecular targets have been discovered for breast cancer prognosis and therapy. However, there is still an urgent need to identify new biomarkers. Therefore, we evaluated biomarkers that may aid the diagnosis and treatment of breast cancer. We searched three mRNA microarray datasets (GSE134359, GSE31448 and GSE42568) and identified differentially expressed genes (DEGs) by comparing tumor and non-tumor tissues using GEO2R. Functional and pathway enrichment analyses of the DEGs were performed using the DAVID database. The protein–protein interaction (PPI) network was plotted with STRING and visualized using Cytoscape. Module analysis of the PPI network was done using MCODE. The associations between the identified genes and overall survival (OS) were analyzed using an online Kaplan–Meier tool. The redundancy analysis was conducted by DepMap. Finally, we verified the screened HUB gene at the protein level. A total of 268 DEGs were identified, which were mostly enriched in cell division, cell proliferation, and signal transduction. The PPI network comprised 236 nodes and 2132 edges. Two significant modules were identified in the PPI network. Elevated expression of the genes Discs large-associated protein 5 ( DLGAP5 ), aurora kinase A ( AURKA ), ubiquitin-conjugating enzyme E2 C ( UBE2C ), ribonucleotide reductase regulatory subunit M2( RRM2 ), kinesin family member 23( KIF23 ), kinesin family member 11( KIF11 ), non-structural maintenance of chromosome condensin 1 complex subunit G ( NCAPG ), ZW10 interactor ( ZWINT ), and denticleless E3 ubiquitin protein ligase homolog( DTL) are associated with poor OS of breast cancer patients. The enriched functions and pathways included cell cycle, oocyte meiosis and the p53 signaling pathway. The DEGs in breast cancer have the potential to become useful targets for the diagnosis and treatment of breast cancer.
This content is subject to copyright. Terms and conditions apply.
1
Vol.:(0123456789)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports
Screening and predicted value
of potential biomarkers for breast
cancer using bioinformatics
analysis
Xiaoyu Zeng, Gaoli Shi, Qiankun He* & Pingping Zhu*
Breast cancer is the most common cancer and the leading cause of cancer-related deaths in women.
Increasing molecular targets have been discovered for breast cancer prognosis and therapy. However,
there is still an urgent need to identify new biomarkers. Therefore, we evaluated biomarkers that
may aid the diagnosis and treatment of breast cancer. We searched three mRNA microarray datasets
(GSE134359, GSE31448 and GSE42568) and identied dierentially expressed genes (DEGs) by
comparing tumor and non-tumor tissues using GEO2R. Functional and pathway enrichment analyses
of the DEGs were performed using the DAVID database. The protein–protein interaction (PPI) network
was plotted with STRING and visualized using Cytoscape. Module analysis of the PPI network was
done using MCODE. The associations between the identied genes and overall survival (OS) were
analyzed using an online Kaplan–Meier tool. The redundancy analysis was conducted by DepMap.
Finally, we veried the screened HUB gene at the protein level. A total of 268 DEGs were identied,
which were mostly enriched in cell division, cell proliferation, and signal transduction. The PPI network
comprised 236 nodes and 2132 edges. Two signicant modules were identied in the PPI network.
Elevated expression of the genes Discs large-associated protein 5 (DLGAP5), aurora kinase A (AURKA),
ubiquitin-conjugating enzyme E2 C (UBE2C), ribonucleotide reductase regulatory subunit M2(RRM2),
kinesin family member 23(KIF23), kinesin family member 11(KIF11), non-structural maintenance of
chromosome condensin 1 complex subunit G (NCAPG), ZW10 interactor (ZWINT), and denticleless
E3 ubiquitin protein ligase homolog(DTL) are associated with poor OS of breast cancer patients. The
enriched functions and pathways included cell cycle, oocyte meiosis and the p53 signaling pathway.
The DEGs in breast cancer have the potential to become useful targets for the diagnosis and treatment
of breast cancer.
Breast cancer has now overtaken lung cancer as the leading cause of cancer incidence worldwide, with an esti-
mated 2.3 million new cases, accounting for 11.7% of all cancer cases1. In China, more than 300,000 women are
diagnosed with breast cancer each year. About 70–80% of breast cancer patients with early stage non-metastatic
disease can be cured, while advanced breast cancers with distant organ metastases are considered untreatable
with currently available therapies2.
e global death rate from breast cancer is declining because of new therapeutic strategies, especially the
targeted therapy. Increasing molecular targets have been discovered for breast cancer prognosis and therapy.
In 2000, Perou and Sorlie reported that breast cancer could be divided into three subtypes according to the
enrichment of three genes, luminal (estrogen receptor [ER]-positive), human epidermal growth factor receptor
2 (HER2, encoded byERBB2)-positive and ER-negative, and basal subtypes3. Since then, several genes have
been identied as predictive and prognostic biomarkers for breast cancer, which play important roles in targeted
therapy. e most commonly used molecular-targeted drugs for HER2-positive breast cancer include tucatinib4,
trastuzumab5, pertuzumab, lapatinib, neratinib and trastuzumab emtansine (T-DM1)6,7. Several drugs target the
phosphoinositide 3-kinase (PI3K)/serine/threonine kinase(AKT) /mammalian target of rapamycin (mTOR)
signaling pathway, including GDC-0068, Bez235, bupacoxib, abencoxib and alpelisib810. Vascular endothelial
growth factor (VEGF) has also been identied as a key target for anti-angiogenic therapy, and its inhibitors beva-
cizumab, sorafenib, and sunitinib are also used for breast cancer therapy11,12. Androgen receptor (AR)-targeted
OPEN
School of Life Sciences, Zhengzhou University, Zhengzhou, China. *email: qiankunhe@zzu.edu.cn; zhup@zzu.
edu.cn
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
Vol:.(1234567890)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
therapies, including AR agonists and AR antagonists, have shown promising results in clinical trials for breast
cancer patients, and combinations of AR-targeted therapies with other reagents (eg. PI3K inhibitor) have been
studied to overcome resistance to AR-targeted therapies13. In addition, targeted therapies have been developed
for epidermal growth factor receptor (EGFR), BRCA1/2-mutated polyadenosine diphosphate ribose polymerase
(PARP), cyclin-dependent kinase 4/6 (CDK4/6), BTB and CNC homology1 (BACH1), and so on1418. However,
because of tumor heterogeneity, low ratios of responders, relapse and drug resistance, there is still an urgent need
to identify new biomarkers that may aid the diagnosis and treatment of breast cancer.
Bioinformatics analysis is a valuable strategy for the comprehensive analysis of large databases, including
complicated genetic information. In our study, we used sophisticated bioinformatics methods to screen poten-
tial biomarkers that may be useful for breast cancer. e Gene Expression Omnibus (GEO) [https:// www. ncbi.
nlm. nih. gov/ geo/] database is an open database that allows researchers to select appropriate mRNA expression
proles. Online analysis tools are available to detect DEGs between tumors and normal tissues. In our study, we
obtained three mRNA microarray datasets from the GEO (GSE134359, GSE31448, and GSE42568) and searched
for DEGs using GEO2R. We then performed functional and pathway enrichment analyses of the identied DEGs
using the DAVID database. PPI networks were constructed using STRING and visualized using Cytoscape. Con-
duct module analyses of the PPI network were performed using MCODE. e associations of these genes with
OS were determined using an online Kaplan–Meier analysis tool. Finally, several breast cancer-related molecules
were selected to investigate their potential role in a breast cancer diagnostic system.
Methods
Subjects and gene information. e GEO database is a national repository of genetic information
databases, including microarray and next-generation sequencing data19. GEO2R is an online tool that can be
used to detect DEGs from two or more GEO datasets. In this study, we retrieved three gene expression proles
(GSE134359, GSE31448 and GSE42568) from the GEO database. GSE134359 (Platform GPL17586) comprised
74 breast cancer samples and 12 noncancerous samples, GSE31448 (Platform GPL570) comprised 29 breast
cancer samples and 4 noncancerous samples, and GSE42568 (Platform GPL570) comprised 104 breast cancer
samples and 17 noncancerous samples. e characteristics of these datasets are shown in Table1. More detailed
patient information is provided in supplementary material 1. e “Reporting recommendations for tumor
marker prognostic studies (REMARK)” were followed20.
Data analysis. We used GEO2R with screening criteria of adj. P < 0.05, log2 FC (fold change) > 1.5, or log2
FC < − 1.5 to detect DEGs in breast cancer tissues compared with normal samples. We also used an online tool
(http:// bioin forma tics. psb. ugent. be/ webto ols/ Venn/) to plot Venn diagrams of the DEGs of three datasets.
Gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) pathway analy-
sis. DAVID (http:// david. ncifc rf. gov; version 6.8) is an open database that integrates biological data and ana-
lytical tools for functional annotation of genes and pathways21. GO is a bioinformatics tool for annotating genes
and analyzing the biological processes they are involved in. KEGG is a database for analyzing relevant signaling
pathways in largescale molecular datasets generated by high-throughput experimental techniques22. DAVID was
used for GO enrichment analysis of the DEGs in terms of the molecular function, cell composition and biologi-
cal process for each gene. KEGG pathway enrichment analysis was performed to clarify the function of the DEGs
and the cell signaling pathways.
PPI network visualization. We used the online analysis tool STRING (http:// www. string- db. org/), with a
condence of 0.4, to construct the PPI network diagram for the identied DEGs. Cytoscape soware23 was then
used to construct the interaction network map, and the MCODE plug-in was used to screen the key gene mod-
ules in the network map. Cytoscape (version 3.8.0) is an open-source bioinformatics tool used to generate visual
molecular interaction networks and the plug-in Molecular Complex Detection (MCODE) can develop key gene
modules in the network. For this, we set the following parameters in MCODE: Degree Cut-o= 2, Node Score
Cut-o = 0.2, K-Core = 2 and Max D epth = 10024.
Table 1. Summary of patient characteristics of three GEO datasets.
GEO accession GSE134359 GSE31448 GSE42568
Sample
Health 12 4 17
Tum or 74 29 104
Histological type
Luminal A 24 1 NA
Luminal B 23 0 NA
HER2+ 14 1 NA
Basal 13 25 NA
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
Vol.:(0123456789)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
Kaplan–Meier survival and redundancy analyses of DEGs. A Kaplan–Meier plotter has been devel-
oped to evaluate the eects of 54,000 transcripts (mRNA, miRNA, protein) on survival for 21 types of can-
cer, including breast cancer (n = 6234), ovarian cancer (n = 2190), lung cancer (n = 3452), and stomach cancer
(n = 1440)25. is database collates data from the GEO, European Genome-phenome Archive, and e Cancer
Genome Atlas (TCGA) databases. e website was used to plot the OS for breast cancer patients for each gene.
By selecting the best cuto, a survival analysis was performed and False-Discovery Rate (FDR) was computed
using the Benjamini–Hochberg method to correct for multiple hypothesis testing26. e hazard ratio (HR) and
log-rank P values with 95% condence intervals (CI) were calculated and displayed on the graph.
Cancer Dependency Map (Cancer DepMap, https:// depmap. org/ portal/), a RNAi and CRISPR-Cas9 knockout
database27,28, was used to identied the functional target genes those are essential for breast cancer survival. e
essential genes are potential therapeutic targets for breast cancer.
Protein level verication. We visualized the selected hub-gene through ualcan29, and the protein expres-
sion data with 18 normal and 125 breast cancer samples were from CPTAC (Oce of Cancer Clinical Proteom-
ics Research, https:// prote omics. cancer. gov/ progr ams/ cptac).
Results
Screening of DEGs. A total of 1529, 1550, and 2188 DEGs were identied from the GSE134359, GSE31448,
and GSE42568 datasets, respectively. Of these, 268 genes were present in all three datasets (Fig.1A). 89 genes
consistently showed high expression and 179 genes showed low expression in all three databases. e top 22
DEGs are shown on the heatmap, based on the criteria |log2 FC|> 3 and adj.P < 0.05 (Fig.1B).
GO and KEGG pathway enrichment analysis. GO enrichment and KEGG pathway analysis were per-
formed on the DEGs using the DAVID database. GO enrichment analysis covers three aspects: biological pro-
cesses, cell composition and molecular function (Fig.2A). e upregulated genes were mainly related to mitotic
cytokinesis, mitotic spindle assembly and microtubule-based movement; while the downregulated genes were
mainly involved in cell adhesion, the response to mechanical stimuli and the response to glucose. e KEGG
pathway analysis showed that the genes upregulated in tumors were enriched in cell cycle, oocyte meiosis and
the P53 signaling pathway, while the downregulated genes were enriched in PPAR signaling pathway, AMPK
signaling pathway, tyrosine metabolism, pathways in cancer and so on (Fig.2B).
PPI network construction and module selection. Considering the critical role of protein interactions
in protein function, we used the STRING database and Cytoscape soware to generate PPI network once we had
Figure1. Identication of DEGs in the indicated breast cancer datasets.(A) ree online-available expression
proling datasets (GSE134359, GSE31448, GSE42568) were analyzed using GEO2R, and genes dierentially
expressed in breast tumor and peri-tumor samples (adj. P < 0.05 and |log2 FC |> 1.5) were dened as DEGs,
followed by Venn diagram of DEGs. (B) Heatmap of top DEGs (adj.P < 0.05 and |log2 FC|> 3) in datasets
GSE134359, GSE31448 and GSE42568.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
Vol:.(1234567890)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
identied the 268 DEGs. e results showed that there were dense regions in PPI, that is, genes closely related to
breast cancer (HUB genes) modules.
A total of 236 nodes and 2132 edges were selected to plot the PPI network, which consisted of 87 up-regulated
genes and 149 down-regulated genes (Fig.3A). Subsequently, a pivotal module of 53 genes (CDK1, KIF11,
DLGAP5, KIF4A and so on) was identied with the degree ≥ 10 as the cut-o value by using MCODE (Fig.3B).
Another important module of 8 genes including both up-regulated and down-regulated genes was also identi-
ed (Fig.3C). e top 10 HUB genes were identied by cytoHubba (Top 10 genes ranked in MCC). GO and
KEGG analysis of these ten genes were conducted. HUB genes are related with cell division, mitotic cytokinesis
in Biologycal Process; spindle, nucleus, spindle microtubule in Cellular Component; protein kinase binding,
ATP binding in Molecular Function (Fig.3D). ey are also enriched in cell cycle, oocyte meiosis, p53 signaling
pathway and so on (Fig.3E).
Survival and redundancy analyses. Ten HUB genes in PPI network were evaluated for their prognostic
value on the Kaplan–Meier plotter. All 10 genes exhibited their potential in the prediction of survival based on
their expression. e OS for breast cancer patients was determined based on the expression level of each gene
(low vs. high). As shown in Fig.4, high mRNA expression of ZWINT (HR 1.6, 95% CI: 1.31–1.94, P < 2.9E−6,
FDR 1%) was associated with a poorer OS for breast cancer patients, and this association also works for DLGAP5
(HR 2.25, 95% CI: 1.74–2.92, P = 2.8e−10, FDR 1%), DTL (HR 1.61, 95% CI: 1.32–1.96, P < 1.5E−6, FDR 1%),
NCAPG(HR 1.6, 95% CI: 1.48–2.19, P < 1.9E−9, FDR 1%), CCNB1 (HR 1.66, 95% CI: 1.27–2.17, P < 0.00019,
FDR 10%), AURKA (HR 1.73, 95% CI: 1.42–2.11, P < 2.9E−8, FDR 1%), KIF23 (HR 1.59, 95% CI: 1.3–1.93,
P < 2.9E−6, FDR 1%), KIF11 (HR 1.64, 95% CI: 1.33–2.03, P < 3.2E−6, FDR 1%), RRM2 (HR 2.09, 95% CI:
1.63–2.68, P < 2.3E−9, FDR 1%) and UBE2C (HR 1.74, 95% CI: 1.43–2.12, P < 2.3E−8, FDR 1%). Among them,
FDR of CCNB1 was 10%. Maybe the relationship between CCNB1 and survival is not obvious.
It is of great signicance to analyze the role of HUB genes in breast cancer cell survival, and the essential genes
are potential therapeutic targets. Here we analyzed the function of HUB genes using online-available DepMap
tool, which was established based on CRISPR screening and siRNA screening data. ere are 2 genes (KIF11,
RRM2) that are common essential in both CRIAPR knockout and RNAi; 6 genes (AURKA, CCNB1, DTL, KIF23,
NCAPG, ZWINT) that are common essential only in CRISPR knockout, indicating that these genes are not only
diagnosis markers but also potential therapeutic targets (Fig.5).
Figure2. GO and KEGG analyses of DEGs. (A) GO analysis with up-regulated (red) and down-regulated
(green) DEGs. Enriched GO items with P < 0.01 are shown, including biological process, cellular component,
and molecular function. (B) KEGG analysis with up-regulated (red) and down-regulated (green) DEGs.
Enriched KEGG pathways (P < 0.01) are shown.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
Vol.:(0123456789)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
Figure3. PPI and MCODE analyses of DEGs. (A) Protein–protein interaction network of 268 DEGs. (B) A
signicant module, containing 53 up-regulated proteins, was selected from protein–protein interaction network.
(C) Another module selected from protein–protein interaction network. (D) GO analysis of MCODE genes.
Enriched GO items with P < 0.01 are shown. (E) KEGG pathway analysis of MCODE genes. Enriched pathways
with P < 0.05 are shown. For (AC), red nodes are up-regulated proteins, and green nodes are down-regulated
proteins. e lines represent the interaction relationship between nodes.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
Vol:.(1234567890)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
Protein level verication. Finally, we veried the screened HUB genes at the protein level (Fig.6). e sta-
tistical signicance of ZWINT (< 1E-12), DLGAP5 (< 1E−12), DTL (1.335246E-04), NCAPG (2.391440E−04),
CCNB1 (3.434030E−03), AURKA (< 1E−12), KIF11 (5.4090702551948E−13), RRM2 (4.27904E−03) and
UBE2C (1.99866742542919E−06) was less than 0.05, except KIF23 (9.61308686E−01).
Discussion
Regardless of recent progress in the treatment of breast cancer, it has remained the most common cause of
cancer-related deaths in the past few years. e high mortality rate of breast cancer is partly due to the lack of
adequate screening methods with high sensitivity and specicity. erefore, it is necessary to identify potential
biomarkers for screening and early diagnosis of breast cancer. Microarray technologies and next-generation
sequencing have become key tools for providing comprehensive genetic information on breast cancer samples
and revealing the changes in disease progression. In this study, we used proven online bioinformatics tools to
investigate possible biomarkers for diagnosis of breast cancer. We identied a total of 268 DEGs common to all
three GEO datasets, which included 89 upregulated genes and 179 downregulated genes.
e upregulated genes were mainly involved in the three pathways, namely cell cycle, oocyte meiosis and the
P53 signaling pathway, which are closely associated with cancer. e downregulated genes were mainly enriched
in three other pathways: cell adhesion, the response to mechanical stimuli and the response to hormonal hypoxia.
Among the identied DEGs, 87 showed high degrees in the PPI network. Further analysis revealed that the
following 10 DEGs within these modules were closely associated with a shorter survival time of breast cancer
patients: DLGAP5, AURKA, UBE2C, CCNB1, RRM2, KIF23, KIF11, NCAPG, ZWINT and DTL.
DLGAP5 is involved in Aurora A signaling and its neurogenic locus notch homolog protein 3 (NOTCH3)
intracellular domain regulates transcription. DLGAP5 overexpression is associated with poor prognosis of breast
Figure4. Prognostic estimation of the top 10 HUB genes. e top 10 HUB genes including ZWINT, DLGAP5,
DTL, NCAPG, CCNB1, AURKA, KIF23, KIF11, RRM2 and UBE2C, were identied by cytoHubba, followed by
survival analysis. Breast cancer patients were divided into two groups according to auto select best cuto. Low,
patients with gene expression lower than best cuto; high, patients with gene expression higher than best cuto.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
Vol.:(0123456789)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
cancer30. DLGAP5 is also associated with the prognosis of colorectal cancer, prostate cancer, and non-small cell
lung cancer (NSCLC)3135. A study identied a critical target of NOTCH3 signaling was the mitotic apparatus
organizing protein DLGAP5 (HURP/DLG7)36. DLGAP5, which is regulated by nucleolar and spindle associated
protein 1 (NUSAP1), is associated with the proliferation, migration and invasion of invasive breast cancer37.
DLGAP5, required for AURKA-dependent, centrosome-independent mitotic spindle assembly, is essential for
the survival and proliferation ofSMARCA4/BRG1 mutant38.One subpopulation of prostate cancerwas associated
with enhanced expression ofDLGAP5 and decreased dependence upon androgen receptor signaling39.
AURKA plays an important role in cell cycle progression by promoting cell entry into mitosis, and is associ-
ated with increased risk of developing breast cancer. AURKA can translocate to the nucleus and enhance the
phenotype of breast cancer stem cells, promoting unique oncogenic properties in malignant cells40. It has been
reported that AURKA regulates the phenotype of breast cancer tumor stem cells by modifying and stabilizing
Drosha mRNA with M6A41. In addition, AURKA plays an important role in the treatment of drug-resistant breast
cancer42, and Aurora kinase A inhibitor has been in a ve-arm phase 2 study for safety and activity43.
Figure5. Redundancy analysis of the top 10 HUB genes. e essential role of indicated HUB genes in breast
cancer cell survival was analyzed via DepMap, (https:// depmap. org/ portal/), which was established from
CRISPR and RNAi screening data. (A) Redundancy analysis of ten genes in total cells lines. (B) e CERES
dependency score of ten genes in breast cancer cells. A lower CERES score indicates a higher likelihood that the
gene of interest is essential in a given cell line. A score of 0 indicates a gene is not essential (dotted line); − 1 is
comparable to the median of all pan-essential genes (red line).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
Vol:.(1234567890)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
UBE2C can ubiquitinate Anaphase-Promoting Complex/Cyclosome (APC/C) (Ub)44. e high expression
of UBE2C in breast cancer was reported to be an independent prognostic factor associated with increased risk
of disease recurrence and death. us, it is considered as a potential therapeutic target for breast cancer4547.
Cyclin B1, the protein encoded by CCNB1, is a regulatory protein involved in mitosis. It is necessary for
proper control of the G2/M transition phase of the cell cycle. A study showed cyclin B1 and B2 transgenic mice
are highly prone to tumors, including tumor types where B-type cyclins serve as prognosticators48. CCNB1 is
associated with radiosensitivity in colorectal cancer49. CCNB1 can also aect cavernous sinus invasion in pituitary
adenomas through the epithelial-mesenchymal transition50.
e gene RRM2 encodes ribonucleotide reductase regulatory subunit M2, one of two non-identical subunits
of ribonucleotide reductase. In a study that reported RRM2 acetylation at K95 suppresses tumor cell growth
invitro and invivo, and is therefore a potentially attractive strategy for cancer therapy51. In a study that searched
Figure6. Protein expression of the top 10 HUB genes. e top 10 HUB genes, including ZWINT, DLGAP5,
DTL, NCAPG, CCNB1, AURKA, KIF23, KIF11, RRM2 and UBE2C, were identied by cytoHubba, veried at
the protein level by Ualcan. Z-values represent standard deviations from the median across samples for the given
cancer type. Log2 Spectral count ratio values from CPTAC were rst normalized within each sample prole,
then normalized across samples.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
Vol.:(0123456789)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
the GEO database for miRNA-mRNA or lncRNA-mRNA as novel biomarkers for breast cancer, the miR-21/
RRM2 axis was identied as a candidate biomarker for the diagnosis and treatment of breast cancer52. In another
study that showed a lincRNA, lincNMR, regulates tumor cell proliferation through a YBX1-RRM2-TYMS-TK1
axis governing nucleotide metabolism53. In addition, RRM2 was reported to be associated with the prognosis
of prostate cancer54.
Kinesin family member 23, the protein encoded by KIF23 is a member of the kinesin-like protein family, also
known as MKLP1. MKLP1/KIF23 is the kinesin component of the centralspindlin complex55. It was reported
that KIF23 expression is high in the majority of primary and metastatic lung cancer tissues or cell lines, and it is
associated with poor survival56. In a study that examined the association between members of the kinesin fam-
ily and breast cancer, KIF23 and KIF11 were found to be associated with poor prognosis57. KIF23 is regulated
through wnt signaling pathway and associated with recurrence of hepatocellular carcinoma58.
Kinesin family member 11, the protein encoded by KIF11, is another member of the kinesin-like protein
family. According to an Oncomine analysis of GEO and TCGA databases, KIF11 is a proto-oncogene associated
with breast cancer and is signicantly associated with poor prognosis59. KIF11 is also regulated through wnt
signaling pathway and associated with recurrence of hepatocellular carcinoma.
NCAPG is a potential prognostic marker in HER2+ breast cancer, anda therapeutic target to eectively over-
come trastuzumab resistance as well60. NCAPG has also been identied as a key gene in triple-negative breast
cancer61 as well as hepatocellular carcinoma62. Furthermore, it was reported that high expression of NCAPG is
associated with poor prognosis of various tumor types, and its overexpression may play an important role in the
regulation of tumor-related pathways in tumor growth63.
Currently, little is known about the role of ZW10 interactor (ZWINT) in breast cancer. Denticleless E3 ubiq-
uitin protein ligase homolog (DTL) is associated with proliferation and appears to be a promising molecular
therapeutic target in breast cancer64. DTL may also be associated with poor prognosis of acral melanoma and
gastric carcinoma65,66.
Based on redundancy analysis, two genes, KIF11 and RRM2, may serve as therapeutic targets or prognostic
indicators. e two genes are also dierentially expressed by protein level verication. ere are many dierences
between the predicted data and the clinical data, and the survival data derived from the Kaplan–Meier tool need
to be validated. In future studies, more attention should be paid to breast cancer patients. ere are many tumor
subtypes for breast cancer, and it is necessary to dene the biomarker characteristics of each subtypes. In our
future study, we intend to recruit a cohort of breast cancer patients to investigate the sensitivity and specicity
of these biomarkers for early screening of breast cancer; the results should facilitate the clinical application of
these biomarkers for the diagnosis of breast cancer.
Data availability
e datasets are available from the GEO database.
Received: 9 June 2021; Accepted: 8 October 2021
References
1. Sung, H. G. et al. Global caner statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185
countries. CA Cancer J. Clin. https:// doi. org/ 10. 3322/ caac. 21660 (2020).
2. Harbeck, N, et al. Breast cancer. Nat Rev Dis Primers 5, 1–66. https:// doi. org/ 10. 1038/ s41572- 019- 0111-2 (2019).
3. Perou, C. M. et al. Molecular portraits of human breast tumours. Nature 406, 747–752. https:// doi. org/ 10. 1038/ 35021 093 (2000).
4. Murthy, R. K. et al. Tucatinib, trastuzumab, and capecitabine for HER2-positive metastatic breast cancer. N. Engl. J. Med. 382,
597–609. https:// doi. org/ 10. 1056/ NEJMo a1914 609 (2020).
5. von Minckwitz, G. et al. Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N. Engl. J. Med. 380, 617–628.
https:// doi. org/ 10. 1056/ NEJMo a1814 017 (2019).
6. Oh, D. Y. & Bang, Y. J. HER2-targeted therapies - a role beyond breast cancer. Nat. Rev. Clin. Oncol. 17, 33–48. https:// doi. org/ 10.
1038/ s41571- 019- 0268-3 (2020).
7. Swain, S. M. et al. Pertuzumab, trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA): end-of-
study results from a double-blind, randomised, placebo-controlled, phase 3 study. Lancet Oncol. 21, 519–530. https:// doi. org/ 10.
1016/ S1470- 2045(19) 30863-0 (2020).
8. Ippen, F. M. et al. Targeting the PI3K/Akt/mTOR pathway with the pan-Akt inhibitor GDC-0068 in PIK3CA-mutant breast cancer
brain metastases. Neuro Oncol. 21, 1401–1411. https:// doi. org/ 10. 1093/ neuonc/ noz105 (2019).
9. Verret, B., Cortes, J., Bachelot, T., Andre, F. & Arnedos, M. Ecacy of PI3K inhibitors in advanced breast cancer. Ann. Oncol.
30(Suppl 10), x12–x20. https:// doi. org/ 10. 1093/ annonc/ mdz381 (2019).
10. Vas an, N. et al. Double PIK3CA mutations in cis increase oncogenicity and sensitivity to PI3Kalpha inhibitors. Science 366,
714–723. https:// doi. org/ 10. 1126/ scien ce. aaw90 32 (2019).
11. Baselga, J. et al. Sorafenib in combination with capecitabine: an oral regimen for patients with HER2-negative locally advanced
or metastatic breast cancer. J. Clin. Oncol. 30, 1484–1491. https:// doi. org/ 10. 1200/ JCO. 2011. 36. 7771 (2012).
12. Uribesalgo, I. et al. Apelin inhibition prevents resistance and metastasis associated with anti-angiogenic therapy. EMBO Mol. Med.
11, e9266. https:// doi. org/ 10. 15252/ emmm. 20180 9266 (2019).
13. Kono, M. et al. Androgen receptor function and androgen receptor-targeted therapies in breast cancer: a review. JAMA Oncol. 3,
1266–1273. https:// doi. org/ 10. 1001/ jamao ncol. 2016. 4975 (2017).
14. Esteva, F. J., Hubbard-Lucey, V. M., Tang, J. & Pusztai, L. Immunotherapy and targeted therapy combinations in metastatic breast
cancer. Lancet Oncol. 20, e175–e186. https:// doi. org/ 10. 1016/ S1470- 2045(19) 30026-9 (2019).
15. Baselga, J. et al. Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with
cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer. J. Clin. Oncol. 31, 2586–2592. https:// doi.
org/ 10. 1200/ JCO. 2012. 46. 2408 (2013).
16. O’Shaughnessy, J. et al. Iniparib plus chemotherapy in metastatic triple-negative breast cancer. N. Engl. J. Med. 364, 205–214.
https:// doi. org/ 10. 1056/ NEJMo a1011 418 (2011).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
Vol:.(1234567890)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
17. Salazar, L. G. et al. Topical imiquimod plus nab-paclitaxel for breast cancer cutaneous metastases: a phase 2 clinical trial. JAMA
Oncol. 3, 969–973. https:// doi. org/ 10. 1001/ jamao ncol. 2016. 6007 (2017).
18. Lee, J. et al. Eective breast cancer combination therapy targeting BACH1 and mitochondrial metabolism. Nature 568, 254–258.
https:// doi. org/ 10. 1038/ s41586- 019- 1005-x (2019).
19. Barrett, T. et al. NCBI GEO: archive for functional genomics data sets–update. Nucleic Acids Res. 41, D991-995. https:// doi. org/
10. 1093/ nar/ gks11 93 (2013).
20. McShane, L. M. et al. REporting recommendations for tumor MARKer prognostic studies (REMARK). Breast Cancer Res. Treat.
100, 229–235. https:// doi. org/ 10. 1007/ s10549- 006- 9242-8 (2006).
21. Huang, D. W. et al. DAVID Bioinformatics resources: expanded annotation database and novel algorithms to better extract biology
from large gene lists. Nucleic Acids Res. 35, W169-175. https:// doi. org/ 10. 1093/ nar/ gkm415 (2007).
22. Kanehisa, M., Furumichi, M., Tanabe, M., Sato, Y. & Morishima, K. KEGG: new perspectives on genomes, pathways, diseases and
drugs. Nucleic Acids Res. 45, D353–D361. https:// doi. org/ 10. 1093/ nar/ gkw10 92 (2017).
23. Shannon, P. et al. Cytoscape: a soware environment for integrated models of biomolecular interaction networks. Genome Res.
13, 2498–2504. https:// doi. org/ 10. 1101/ gr. 12393 03 (2003).
24. Yao, Q. et al. Identication of potential genomic alterations and the circRNA-miRNA-mRNA regulatory network in primary and
recurrent synovial sarcomas. Front. Mol. Biosci. https:// doi. org/ 10. 3389/ fmolb. 2021. 707151 (2021).
25. Nagy, A., Lanczky, A., Menyhart, O. & Gyory, B. Validation of miRNA prognostic power in hepatocellular carcinoma using
expression data of independent datasets. Sci. Rep. 8, 9227. https:// doi. org/ 10. 1038/ s41598- 018- 27521-y (2018).
26. Győry, B. Survival analysis across the entire transcriptome identies biomarkers with the highest prognostic power in breast
cancer. Comput. Struct. Biotechnol. J. 19, 4101–4109. https:// doi. org/ 10. 1016/j. csbj. 2021. 07. 014 (2021).
27. Dwane, L. et al. Project Score database: a resource for investigating cancer cell dependencies and prioritizing therapeutic targets.
Nucleic Acids Res. 49, D1365–D1372. https:// doi. org/ 10. 1093/ nar/ gkaa8 82 (2021).
28. Tsherniak, A. et al. Dening a cancer dependency map. Cell 170, 564-576 e516. https:// doi. org/ 10. 1016/j. cell. 2017. 06. 010 (2017).
29. Chandrashekar, D. S. et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia
19, 649–658. https:// doi. org/ 10. 1016/j. neo. 2017. 05. 002 (2017).
30. Xu, T., Dong, M., Li, H., Zhang, R. & Li, X. Elevated mRNA expression levels of DLGAP5 are associated with poor prognosis in
breast cancer. Oncol. Lett. 19, 4053–4065. https:// doi. org/ 10. 3892/ ol. 2020. 11533 (2020).
31. Branchi, V. et al. Prognostic value of DLGAP5 in colorectal cancer. Int. J. Colorectal. Dis. 34, 1455–1465. https:// doi. org/ 10. 1007/
s00384- 019- 03339-6 (2019).
32. Yamamoto, S. et al. Identication of new octamer transcription factor 1-target genes upregulated in castration-resistant prostate
cancer. Cancer Sci. 110, 3476–3485. https:// doi. org/ 10. 1111/ cas. 14183 (2019).
33. Shi, Y. X. et al. Genome-scale analysis identies NEK2, DLGAP5 and ECT2 as promising diagnostic and prognostic biomarkers
in human lung cancer. Sci. Rep. 7, 8072. https:// doi. org/ 10. 1038/ s41598- 017- 08615-5 (2017).
34. Wang, Q., Chen, Y., Feng, H., Zhang, B. & Wang, H. Prognostic and predictive value of HURP in nonsmall cell lung cancer. Oncol.
Rep. 39, 1682–1692. https:// doi. org/ 10. 3892/ or. 2018. 6280 (2018).
35. Fragoso, M. C. et al. Combined expression of BUB1B, DLGAP5, and PINK1 as predictors of poor outcome in adrenocortical
tumors: validation in a Brazilian cohort of adult and pediatric patients. Eur. J. Endocrinol. 166, 61–67. https:// doi. org/ 10. 1530/
EJE- 11- 0806 (2012).
36. Chen, X. et al. Dening NOTCH3 target genes in ovarian cancer. Cancer Res. 72, 2294–2303. https:// doi. org/ 10. 1158/ 0008- 5472.
CAN- 11- 2181 (2012).
37. Zhang, X., Pan, Y., Fu, H. & Zhang, J. Nucleolar and spindle associated protein 1 (NUSAP1) inhibits cell proliferat ion and enhances
susceptibility to epirubicin in invasive breast cancer cells by regulating cyclin D kinase (CDK1) and DLGAP5 expression. Med.
Sci. Monit. 24, 8553–8564. https:// doi. org/ 10. 12659/ MSM. 910364 (2018).
38. Tagal, V. et al. SMARCA4-inactivating mutations increase sensitivity to Aurora kinase A inhibitor VX-680 in non-small cell lung
cancers. Nat. Commun. 8, 14098. https:// doi. org/ 10. 1038/ ncomm s14098 (2017).
39. Horning, A. M. et al. Single-Cell RNA-seq reveals a subpopulation of prostate cancer cells with enhanced cell-cycle-related tran-
scription and attenuated androgen response. Cancer Res. 78, 853–864. https:// doi. org/ 10. 1158/ 0008- 5472. CAN- 17- 1924 (2018).
40. Zheng, F. et al. Nuclear AURKA acquires kinase-independent transactivating function to enhance breast cancer stem cell pheno-
type. Nat. Commun. 7, 10180. https:// doi. org/ 10. 1038/ ncomm s10180 (2016).
41. Peng, F. et al. Oncogenic AURKA-enhanced N(6)-methyladenosine modication increases DROSHA mRNA stability to transac-
tivate STC1 in breast cancer stem-like cells. Cell Res. https:// doi. org/ 10. 1038/ s41422- 020- 00397-2 (2020).
42. Donnella, H. J. et al. Kinome rewiring reveals AURKA limits PI3K-pathway inhibitor ecacy in breast cancer. Nat. Chem. Biol.
14, 768–777. https:// doi. org/ 10. 1038/ s41589- 018- 0081-9 (2018).
43. Melichar, B. et al. Safety and activity of alisertib, an investigational aurora kinase A inhibitor, in patients with breast cancer, small-
cell lung cancer, non-small-cell lung cancer, head and neck squamous-cell carcinoma, and gastro-oesophageal adenocarcinoma:
a ve-arm phase 2 study. Lancet Oncol. 16, 395–405. https:// doi. org/ 10. 1016/ S1470- 2045(15) 70051-3 (2015).
44. Martinez-Chacin, R. C. et al. Ubiquitin chain-elongating enzyme UBE2S activates the RING E3 ligase APC/C for substrate prim-
ing. Nat. Struct. Mol. Biol. 27, 550–560. https:// doi. org/ 10. 1038/ s41594- 020- 0424-6 (2020).
45. Psyrri, A. et al. Prognostic signicance of UBE2C mRNA expression in high-risk early breast cancer: a hellenic cooperative oncol-
ogy group (HeCOG) study. Ann. Oncol. 23, 1422–1427. https:// doi. org/ 10. 1093/ annonc/ mdr527 (2012).
46. Qin, T. et al. Exceptionally high UBE2C expression is a unique phenomenon in basal-like type breast cancer and is regulated by
BRCA1. Biomed. Pharmacother. 95, 649–655. https:// doi. org/ 10. 1016/j. biopha. 2017. 08. 095 (2017).
47. Chou, C. P. et al. Ubiquitin-conjugating enzyme UBE2C is highly expressed in breast microcalcication lesions. PLoS ONE 9,
e93934. https:// doi. org/ 10. 1371/ journ al. pone. 00939 34 (2014).
48. Nam, H. J. & van Deursen, J. M. Cyclin B2 and p53 control proper timing of centrosome separation. Nat. Cell Biol. 16, 538–549.
https:// doi. org/ 10. 1038/ ncb29 52 (2014).
49. Song, W. et al. Silencing PSME3 induces colorectal cancer radiosensitivity by downregulating the expression of cyclin B1 and
CKD1. Exp. Biol. Med. (Maywood) 244, 1409–1418. https:// doi. org/ 10. 1177/ 15353 70219 883408 (2019).
50. Li, B. et al. CCNB1 aects cavernous sinus invasion in pituitary adenomas through the epithelial-mesenchymal transition. J. Transl.
Med. 17, 336. https:// doi. org/ 10. 1186/ s12967- 019- 2088-8 (2019).
51. Chen, G. et al. Acetylation regulates ribonucleotide reductase activity and cancer cell growth. Nat. Commun. 10, 3213. https:// doi.
org/ 10. 1038/ s41467- 019- 11214-9 (2019).
52. Quan, D. et al. Identication of lncRNA NEAT1/miR-21/RRM2 axis as a novel biomarker in breast cancer. J. Cell Physiol. 235,
3372–3381. https:// doi. org/ 10. 1002/ jcp. 29225 (2020).
53. Gandhi, M. et al. e lncRNA lincNMR regulates nucleotide metabolism via a YBX1—RRM2 axis in cancer. Nat. Commun. 11,
3214. https:// doi. org/ 10. 1038/ s41467- 020- 17007-9 (2020).
54. Mazzu, Y. Z. et al. A novel mechanism driving poor-prognosis prostate cancer: overexpression of the DNA repair gene, ribonu-
cleotide reductase small subunit M2 (RRM2). Clin. Cancer Res. 25, 4480–4492. https:// doi. org/ 10. 1158/ 1078- 0432. CCR- 18- 4046
(2019).
55. Capalbo, L. et al. e midbody interactome reveals unexpected roles for PP1 phosphatases in cytokinesis. Nat. Commun. 10, 4513.
https:// doi. org/ 10. 1038/ s41467- 019- 12507-9 (2019).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
11
Vol.:(0123456789)
Scientic Reports | (2021) 11:20799 | https://doi.org/10.1038/s41598-021-00268-9
www.nature.com/scientificreports/
56. Kato, T. et al. Overexpression of KIF23 predicts clinical outcome in primary lung cancer patients. Lung Cancer 92, 53–61. https://
doi. org/ 10. 1016/j. lungc an. 2015. 11. 018 (2016).
57. Li, T. F. et al. Overexpression of kinesin superfamily members as prognostic biomarkers of breast cancer. Cancer Cell Int. 20, 123.
https:// doi. org/ 10. 1186/ s12935- 020- 01191-1 (2020).
58. Chen, J. et al. e microtubule-associated protein PRC1 promotes early recurrence of hepatocellular carcinoma in association
with the Wnt/beta-catenin signalling pathway. Gut 65, 1522–1534. https:// doi. org/ 10. 1136/ gutjnl- 2015- 310625 (2016).
59. Zhou, J. et al. KIF11 functions as an oncogene and is associated with poor outcomes from breast cancer. Cancer Res. Treat. O. J.
Korean Cancer Assoc. 51, 1207–1221. https:// doi. org/ 10. 4143/ crt. 2018. 460 (2019).
60. Jiang, L. et al. NCAPG confers trastuzumab resistance via activating SRC/STAT3 signaling pathway in HER2-positive bre ast cancer.
Cell Death Dis. 11, 547. https:// doi. org/ 10. 1038/ s41419- 020- 02753-x (2020).
61. Chen, J., Qian, X., He, Y., Han, X. & Pan, Y. Novel key genes in triple-negative breast cancer identied by weighted gene co-
expression network analysis. J. Cell Biochem. 120, 16900–16912. https:// doi. org/ 10. 1002/ jcb. 28948 (2019).
62. Bayo, J. et al. A comprehensive study of epigenetic alterations in hepatocellular carcinoma identies potential therapeutic targets.
J. Hepatol. 71, 78–90. https:// doi. org/ 10. 1016/j. jhep. 2019. 03. 007 (2019).
63. Xiao, C. et al. NCAPG is a promising therapeutic target across dierent tumor types. Front. Pharmacol. 11, 387. https:// doi. org/
10. 3389/ fphar. 2020. 00387 (2020).
64. Ueki, T. et al. Involvement of elevated expression of multiple cell-cycle regulator, DTL/RAMP (denticleless/RA-regulated nuclear
matrix associated protein), in the growth of breast cancer cells. Oncogene 27, 5672–5683. https:// doi. org/ 10. 1038/ onc. 2008. 186
(2008).
65. Yang, L. et al. Identication of a functional polymorphism within the 3’-untranslated region of denticleless E3 ubiquitin protein
ligase homolog associated with survival in acral melanoma. Eur. J. Cancer 118, 70–81. https:// doi. org/ 10. 1016/j. ejca. 2019. 06. 006
(2019).
66. Kobayashi, H. et al. Overexpression of denticleless E3 ubiquitin protein ligase homolog (DTL) is related to poor outcome in gastric
carcinoma. Oncotarget 6, 36615–36624. https:// doi. org/ 10. 18632/ oncot arget. 5620 (2015).
Acknowledgements
e authors thank all individuals who contributed sample information and research personnel who have been
responsible for data acquisition. Data are available from GEO database. is work was supported by Ministry of
Science and Technology of the People’s Republic of China (2020YFA0803500), National Natural Science Foun-
dation of China (31922024). We thank the supporting grants from Zhengzhou University to Pingping Zhu, and
the technical support from Modern Analysis and Computer Center of Zhengzhou University.
Author contributions
X.Z. wrote the main manuscript text and prepared Figs.1, 2, 3, 4, and 5. G.S. prepared Fig.1. All authors reviewed
the manuscript.
Funding
This work was supported by Ministry of Science and Technology of the People’s Republic of China
(2020YFA0803500), National Natural Science Foundation of China (31922024).
Competing interests
e authors declare no competing interests.
Additional information
Supplementary Information e online version contains supplementary material available at https:// doi. org/
10. 1038/ s41598- 021- 00268-9.
Correspondence and requests for materials should be addressed to Q.H.orP.Z.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.
© e Author(s) 2021
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... The Aurora-A kinase (AURKA) is an important cell cycle regulatory protein found to be overexpressed in various cancers, including PCa [14][15][16]. Studies have indicated that the overexpression of AURKA is closely associated with the malignant progression, metastasis, and development of resistance to treatment in PCa [17,18]. ...
... Studies have indicated that the overexpression of AURKA is closely associated with the malignant progression, metastasis, and development of resistance to treatment in PCa [17,18]. AURKA plays a crucial role in the growth and survival of cancer cells by regulating cell division and proliferation [14,19,20]. Targeting AURKA can inhibit cancer cell proliferation and induce apoptosis [20][21][22]. ...
Article
Full-text available
Castration-resistant prostate cancer (CRPC) presents significant therapeutic challenges due to its aggressive nature and poor prognosis. Targeting Aurora-A kinase (AURKA) has shown promise in cancer treatment. This study investigates the efficacy of ART-T cell membrane-encapsulated AMS@AD (CM-AMS@AD) nanoparticles (NPs) in a photothermal–chemotherapy–immunotherapy combination for CRPC. Bioinformatics analysis of the Cancer Genome Atlas-prostate adenocarcinoma (TCGA-PRAD) dataset revealed overexpression of AURKA in PCa, correlating with poor clinical outcomes. Single-cell RNA sequencing data from the GEO database showed a significant reduction in immune cells in CRPC. Experimentally, T cell membrane-biomimetic NPs loaded with the AURKA inhibitor Alisertib and chemotherapy drug DTX were synthesized and characterized by dynamic light scattering and transmission electron microscopy, showing good stability and uniformity (average diameter: 158 nm). In vitro studies demonstrated that these NPs inhibited CRPC cell proliferation, increased the G2/M cell population, and elevated apoptosis, confirmed by γH2AX expression. In vivo, CM-AMS@AD NPs accumulated in tumor tissues, significantly slowed tumor growth, decreased proliferation, increased apoptosis, and improved the immune environment, enhancing dendritic cell (DC) maturation and increasing CD8 + /CD4 + ratios. These findings suggest that CM-AMS@AD NPs offer a promising triple-combination therapy for CRPC, integrating photothermal, chemotherapy, and immunotherapy, with significant potential for future clinical applications. Graphical Abstract
... Initially, UBE2C was integrated into a highly sensitive and specific multi-marker assay for detecting circulating tumor cells (CTCs) in breast cancer patients. Researchers used high-throughput membrane arrays to detect a set of mRNA Table 1 Summarizing the Role and Impact of UBE2C in Different Types of Malignant Tumors UBE2C as a prognostic factor in lung adenocarcinoma [20] Overexpression of UBE2C and AGGF1 is associated with angiogenesis and poor prognosis [21] Lung cancer UBE2C+ cancer cell subpopulations are increasing during LUAD invasion [22] The UBE2C/CDH1/DEPTOR axis forms an oncogene and tumor suppressor cascade that regulates cell cycle progression and autophagy [23] UBE2C Protein May Serve as a Prognostic Marker in N+ Breast Cancer [26] UBE2C may serve as a prognostic marker for breast cancer [28] Breast cancer UBE2S and UBE2C downregulate Numb and enhance breast cancer malignancy [29] UBE2C can promote BC proliferation by activating the AKT/mTOR signaling pathway [30] ALKBH5 promotes stemness, growth and metastasis of triple negative breast cancer (TNBC) cells by regulating m6A modification of UBE2C to upregulate UBE2C expression and decrease p53 expression [31] UBE2C overexpression enhances cell proliferation and UBE2C depletion inhibits cell proliferation in colon cancer cells [33,34] Colorectal cancer UBE2C inhibits colorectal cancer cell growth in vitro and in vivo [35] UBE2C promotes rectal carcinoma via miR-381 [39] High levels of UBE2C are significantly associated with prognosis in patients with esophageal squamous cell carcinoma [40] This suggests that ECRG4 down-regulates the expression of UBE2C in ESCC cells through NF-κB signaling, and UBE2C ...
... 27 Recent bioinformatics studies have identified UBE2C as a prognostic marker for breast cancer, highlighting its functional enrichment in the cell cycle, oocyte meiosis, and p53 signaling pathway. 28 However, several studies have offered varying insights into the specific mechanisms. Studies have demonstrated that UBE2S and UBE2C downregulate Numb and promote breast cancer malignancy. ...
Article
Full-text available
UBE2C (Ubiquitination-binding enzyme 2C), one of the E2 enzymes encoded in the human genome, is a component of the ubiquitin proteasome system and plays a pivotal role in regulating cell cycle progression. Moreover, UBE2C is highly expressed and may play a pivotal role in both high-incidence and high-mortality malignancies, including lung cancers, breast cancers, and esophageal cancers. UBE2C influences a number of key processes, including cell cycle progression, tumor invasion and metastasis, proliferation, and drug resistance. However, few articles have systematically summarized the role of UBE2C in cancer. The aim of this review is to describe the structure and function of UBE2C, focusing on the current status of UBE2C research in malignant tumors. Furthermore, this review presents the potential of UBE2C as a new therapeutic target and a diagnostic and prognostic biomarker. Finally, future research directions for UBE2C are proposed. It is of great value to explore the mechanism of action of UBE2C in the tumor microenvironment (TME). A comprehensive and coherent comprehension of UBE2C will undoubtedly facilitate its transition from fundamental research to clinical applications.
... In multiple cancer types, including non-small cell lung cancer (NSCLC) and liver cancer, DLGAP5 has been linked to poor prognosis and is considered a diagnostic and prognostic biomarker [19,20]. Interestingly, research has shown that the inactivation of the DLGAP5 gene can inhibit the growth of breast cancer cells, potentially through effects on the cell cycle and mitochondrial autophagy [21]. Prior research has underscored the strong association of DLGAP5 with breast cancer development [22,23]. ...
... DLGAP5 is overexpressed in TNBC and a variety of other tumors [21]. This finding suggests it could be a prevalent oncogenic driver [67]. ...
Article
Full-text available
Triple-negative breast cancer (TNBC) is a challenging subtype with unclear biological mechanisms. Recently, the transcription factor androgen receptor (AR) and its regulation of the DLGAP5 gene have gained attention in TNBC pathogenesis. In this study, we found a positive correlation between high AR expression and TNBC cell proliferation and growth. Furthermore, we confirmed DLGAP5 as a critical downstream regulator of AR with high expression in TNBC tissues. Knockdown of DLGAP5 significantly inhibited TNBC cell proliferation, migration, and invasion. AR was observed to directly bind to the DLGAP5 promoter, enhancing its transcriptional activity and suppressing the activation of the p53 signaling pathway. In vivo experiments further validated that downregulation of AR or DLGAP5 inhibited tumor growth and enhanced CD8⁺T cell infiltration. This study highlights the crucial roles of AR and DLGAP5 in TNBC growth and immune cell infiltration. Taken together, AR inhibits the p53 signaling pathway by promoting DLGAP5 expression, thereby impacting CD8⁺T cell infiltration in TNBC.
... These findings could aid in understanding BC development at the molecular level and identifying potential biomarkers for BC diagnosis and treatment. While numerous studies have focused on BC biomarkers (Zeng et al., 2021;Tian et al., 2020;Bankhead et al., 2017), none have specifically combined mutation analysis of hub genes with identifying alternative treatments tailored to these mutations for personalized medicine. ...
Article
Full-text available
Background The proposed study integrates several advanced computational techniques to unravel the molecular mechanisms underlying breast cancer progression and drug resistance. Methods We investigated HER2-L755S mutation through a multi-step approach, including gene expression analysis, molecular docking, and molecular dynamics simulations. Results and Discussion By conducting a network-based analysis of gene expression data from breast cancer samples, key hub genes such as MYC, EGFR, CDKN2A, ERBB2, CDK1, E2F1, TOP2A, MDM2, TGFB1, and FOXM1 were identified, all of which are critical in tumor growth and metastasis. The study mainly focuses on the ERBB2 gene, which encodes the HER2 protein, and its common mutation HER2-L755S, associated with breast cancer and resistance to the drug lapatinib. The HER2-L755S mutation contributes to both tumorigenesis and therapeutic failure. To address this, alternative therapeutic strategies were investigated using combinatorial computational approaches. The stability and flexibility of the HER2-L755S mutation were evaluated through comparative molecular dynamics simulations over 1000 ns using Gromacs in the unbound (Apo) state. Virtual screening with Schrodinger Glide identified ibrutinib as a promising alternative to lapatinib for targeting the HER2-L755S mutant. Detailed docking and molecular dynamics simulations in the bound (Holo) state demonstrated that the HER2-L755S-ibrutinib complex exhibited higher binding affinity and lower binding energy, indicating more stable interactions compared to other complexes. MM-PBSA analysis revealed that the HER2-L755S-ibrutinib complex had more negative binding energy than the HER2-L755S-afatinib, HER2-L755S-lapatinib, and HER2-L755S-neratinib complexes, suggesting that ibrutinib forms the most stable complex with favorable binding interactions. Conclusion These results provide in-depth atomic-level insights into the binding mechanisms of these inhibitors, highlighting ibrutinib as a potentially effective inhibitor for the clinical treatment of breast cancer.
... The integration of genomic data and other emerging biomarkers into the nomogram could offer deeper insights into the mechanisms driving lymph node metastasis, potentially improving predictive accuracy and clinical utility. 26 In summary, our clinical-radiomics nomogram represents a significant step forward in personalizing the management of young onset breast cancer. It provides a scientifically validated, non-invasive tool that enhances the prediction of ALNM, enabling more precise and tailored treatment strategies. ...
Article
Full-text available
Background Young onset breast cancer, diagnosed in women under 50, is known for its aggressive nature and challenging prognosis. Precisely forecasting axillary lymph node metastasis (ALNM) is essential for customizing treatment plans and enhancing patient results. Objective This research sought to create and verify a clinical-radiomics nomogram that combines radiomic features from Dynamic Contrast-Enhanced Magnetic Resonance Imaging (DCE-MRI) with standard clinical predictors to improve the accuracy of predicting ALNM in young breast cancer patients. Methods We performed a retrospective analysis at one facility, involving the creation and validation of a nomogram in two stages.At first, a medical model was developed utilizing conventional indicators like tumor dimensions, molecular classifications, multifocal presence, and MRI-determined ALN status.A more detailed clinical-radiomics model was subsequently developed by integrating radiomic characteristics derived from DCE-MRI images.These models were created using logistic regression analyses on a training dataset, and their effectiveness was assessed by measuring the area under the receiver operating characteristic curve (AUC) in a separate validation dataset. Results The clinical-radiomics nomogram surpassed the clinical-only model, recording an AUC of 0.892 in the training dataset and 0.877 in the validation dataset.Significant predictors included MRI-reported ALN status and select radiomic features, which markedly enhanced the model’s predictive capacity. Conclusion Integrating radiomic features with clinical predictors in a nomogram significantly improves ALNM prediction in young onset breast cancer, providing a valuable tool for personalized treatment planning. This study underscores the potential of merging advanced imaging data with clinical insights to refine oncological predictive models. Future research should expand to multicentric studies and include genomic data to boost the nomogram’s generalizability and precision.
... STRING()is" title = "https://cn.string-db.org/)is">https://cn.string-db.org/)is a protein interaction database that can be used to analyze both known and predicted interactions between proteins. Protein-protein interaction (PPI) refers to a process in which two or more protein molecules form a protein complex through non-covalent bonds [17]. Protein interactions are a key element of the biochemical response network within cells and are essential for regulating cellular functions and their signaling pathways. ...
Article
Full-text available
Background Non-small cell lung cancer (NSCLC), which accounts for about 85 % of all lung cancers, currently exhibits insensitivity to most treatment regimens. Therefore, the identification of new and effective biomarkers for NSCLC is crucial for the development of treatment strategies. Immunogenic cell death (ICD), a form of regulated cell death capable of activating adaptive immune responses and generating long-term immune memory, holds promise for enhancing anti-tumor immunity and offering promising prospects for immunotherapy strategies in NSCLC. Methods Clinical information and expressive profiles of NSCLC genes were retrieved from the GEO and TCGA databases. By combining these databases, the researchers were able to identify the appropriate genes for use in forecasting outcomes of patients with this type of cancer. We further performed functional enrichment, gene variants and immune privilege correlation analysis to determine the underlying mechanisms. This was followed by univariate and multivariate Cox regression and LASSO regression analyses, we developed a prognostic risk model based on the TCGA cohort, which included 17 gene labels. The results of the external validation were then used to identify the appropriate genes for use in predicting the survival outcome of patients with this type of cancer. In addition, a nomogram was created to help visualise the clinical presentation of the patients. For the analyses, we performed 50 functional and immunoinfiltration assessments for two risk groups. Results Using 17 genes (AIRE, APOH, CDKN2A, CEACAM4, COL4A3, CPA, DBH, F10, FCGRB, FGFR4, MMP1, PGLYRP1, SCGB2A2, SLC9A3, UGT2B17 and VIP), The researchers then created a gene signature that could be used to identify patients with an increased risk of contracting cancer. They divided the patients into two groups based on their risk score. The low-risk group exhibited a better prognosis (P < 0.01). The survival curve demonstrated that ICD-related models could accurately predict patient prognosis. Conversely, high-risk subgroups were closely associated with immune-related signaling pathways. The analysis of immune infiltration also showed that the infiltration levels of most immune cells were higher in the high risk sub-group than in the low risk sub-group. In comparison to the low-risk group, the high-risk group was more susceptible to the immune-checkpoint blockade (ICB) treatment. Conclusion Our researchers utilized a gene model to analyze the immune inflammation and prognosis of patients with non-small-cell lung cancer (NSCLC). The discovery of new ICD-related genes could lead to the development of new targeted treatments for this condition.
... The occurrence and development of breast cancer is thought to be the result of the combined effects of environmental and genetic factors (4). Improving breast cancer screening in high-risk groups, reducing the incidence of breast cancer and improving the cure rate are challenges that need to be addressed (5). The risk of breast cancer has been confirmed to be closely associated with mutations and expression changes in genes such as BRCA1, BRCA2, P53, epidermal growth factor receptor (EGFR), and Ki-67 (6)(7)(8). ...
Article
Full-text available
Introduction Breast cancer, as the most prevalent malignancy among women globally, continues to exhibit rising incidence rates, particularly in China. The disease predominantly affects women aged 40 to 60 and is influenced by both genetic and environmental factors. This study focuses on the role of H19 gene polymorphisms, investigating their impact on breast cancer susceptibility, clinical outcomes, and response to treatment. Methods We engaged 581 breast cancer patients and 558 healthy controls, using TaqMan assays and DNA sequencing to determine genotypes at specific loci (rs11042167, rs2071095, rs2251375). We employed in situ hybridization and immunohistochemistry to measure the expression levels of LincRNA H19, miR-675, MRP3, HOXA1, and MMP16 in formalin-fixed, paraffin-embedded samples. Statistical analyses included chi-squared tests, logistic regression, and Kaplan-Meier survival curves to evaluate associations between genetic variations, gene expression, and clinical outcomes. Results Genotypes AG at rs11042167, GT at rs2071095, and AC at rs2251375 were significantly associated with increased risk of breast cancer. Notably, the AA genotype at rs11042167 and TT genotype at rs2071095 were linked to favorable prognosis. High expression levels of LincRNA H19, miR-675, MRP3, HOXA1, and MMP16 in cancer tissues correlated with advanced disease stages and poorer survival rates. Spearman correlation analysis revealed significant positive correlations between the expression of LincRNA H19 and miR-675 and specific genotypes, highlighting their potential regulatory roles in tumor progression. Discussion The study underscores the critical roles of LincRNA H19 and miR-675 as prognostic biomarkers in breast cancer, with their overexpression associated with disease progression and adverse outcomes. The H19/LincRNA H19/miR-675/MRP3-HOXA1-MMP16 axis offers promising targets for new therapeutic strategies, reflecting the complex interplay between genetic markers and breast cancer pathology. Conclusion The findings confirm that certain H19 SNPs are associated with heightened breast cancer risk and that the expression profiles of related genetic markers can significantly influence prognosis and treatment response. These biomarkers hold potential as targets for personalized therapy and early detection strategies in breast cancer, underscoring the importance of genetic research in understanding and managing this disease.
... These algorithms are designed to handle common tasks such as feature extraction, classification, and regression, streamlining the model development process. This accelerates the pace of healthomics research, enabling scientists to focus more on the biological interpretation of results rather than the intricacies of model development [28]. Moreover, AWS supports the integration of ML models with other AWS services, enhancing the overall analytical capabilities in healthomics. ...
Article
Full-text available
Breast cancer presents a profound global health challenge, compounded by unique intricacies within the Indian demographic, necessitating bespoke research methodologies. This abstract delineates the profound impact of Amazon Web Services (AWS) Cloud Solutions on advancing multi-omics breast cancer biomarker research, with a particular focus on Indian patient cohorts. It initiates with an exposition of the inherent challenges encountered during the transition from raw data acquisition to clinical diagnosis, emphasizing the indispensable role of cloud-based infrastructures in expediting this complex trajectory. Harnessing the comprehensive capabilities of AWS, this study elucidates how cloud solutions facilitate the seamless integration and analysis of multifaceted omics datasets, encompassing genomics, transcriptomics, proteomics, and metabolomics. Central to this endeavor is a meticulous exploration of region-specific molecular markers germane to breast cancer within the Indian populace, illuminating their diagnostic and therapeutic ramifications. By capitalizing on AWS Cloud's scalability and computational acumen, this research underscores notable efficiency enhancements in processing voluminous datasets and distilling salient patterns therein. Furthermore, the discourse extends to the broader ramifications of these technological advancements within the precision medicine landscape, emphasizing the potential for tailored therapeutic interventions. This research heralds a paradigmatic shift in the application of cloud-based infrastructures to unravel the intricate tapestry of breast cancer, transcending geographical confines. Through its provision of insights poised to augment diagnostic precision and therapeutic efficacy on a global scale, this study marks a seminal stride towards fully harnessing the potential of precision oncology in combating breast malignancies.
Article
Full-text available
Angiotensin II (Ang II) plays an important role in regulating various physiological processes. However, little is known about the existence of intracellular Ang II (iAng II), whether iAng II would regulate the automaticity of early differentiating cardiomyocytes, and the underlying mechanism involved. Here, iAng II was detected by immunocytochemistry and ultra-high performance liquid chromatography combined with electrospray ionization triple quadrupole tandem mass spectrometry in mouse embryonic stem cell–derived cardiomyocytes (mESC-CMs) and neonatal rat ventricular myocytes. Expression of AT1R-YFP in mESC-CMs revealed that Ang II type 1 receptors were located on the surface membrane, while immunostaining of Ang II type 2 receptors (AT2R) revealed that AT2R were predominately located on the nucleus and the sarcoplasmic reticulum. While extracellular Ang II increased spontaneous action potentials (APs), dual patch clamping revealed that intracellular delivery of Ang II or AT2R activator C21 decreased spontaneous APs. Interestingly, iAng II was found to decrease the caffeine-induced increase in spontaneous APs and caffeine-induced calcium release, suggesting that iAng II decreased spontaneous APs via the AT2R- and ryanodine receptor–mediated pathways. This is the first study that provides evidence of the presence and function of iAng II in regulating the automaticity behavior of ESC-CMs and may therefore shed light on the role of iAng II in fate determination.
Article
Full-text available
Introduction: Synovial sarcoma (SS) is one of the most invasive soft tissue sarcomas, prone to recurrence and metastasis, and the efficacy of surgical treatment and chemotherapy for SS remains poor. Therefore, the diagnosis and treatment of SS remain a significant challenge. This study aimed to analyze the mutated genes of primary SS (PSS) and recurrent SS (RSS), discover whether these sarcomas exhibit some potential mutated genes, and then predict associated microRNAs (miRNA) and circular RNAs (circRNA) by analyzing the mutated genes. We focused on the regulation mechanism of the circRNA-miRNA-mutated hub gene in PSS and RSS. Methods: We performed a comprehensive genomic analysis of four pairs of formalin-fixed paraffin-embedded samples of PSS and RSS, using Illumina human exon microarrays. The gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) function, and pathway enrichment of the mutated genes were analyzed, and the protein-protein interaction (PPI) network was forecast using String software 11.0. The hub genes were then obtained using the Molecular Complex Detection (MCODE) plug-in for Cytoscape 3.7.2 and were used to analyze overall survival (OS) using the Gene Expression Profiling Interactive Analysis (GEPIA) database. The corresponding miRNAs were obtained from the miRDB 5.0 and TargetScan 7.2 databases. The corresponding circRNAs of the hub genes were found through the miRNAs from these databases: Circbank, CircInteractome, and StarBase v2.0. Thereafter we set up a competing endogenous RNA (ceRNA) network with circRNA-miRNA and miRNA-messenger RNA (mRNA) pairs. Results: Using the chi-squared test, 391 mutated genes were screened using a significance level of p -values < 0.01 from the four pairs of PSS and RSS samples. A GO pathway analysis of 391 mutated genes demonstrated that differential expression mRNAs (DEmRNAs) might be bound up with the “positive regulation of neurogenesis,” “cell growth,” “axon part,” “cell−substrate junction,” or “protein phosphatase binding” of SS. The PPI network was constructed using 391 mutated genes, and 53 hub genes were identified ( p < 0.05). Eight variant hub genes were discovered to be statistically significant using the OS analysis ( p < 0.05). The circRNA-miRNA-mRNA (ceRNA) network was constructed, and it identified two circRNAs (hsa_circ_0070557 and hsa_circ_0070558), 10 miRNAs (hsa-let-7a-3p, hsa-let-7b-3p, hsa-let-7f-1-3p, hsa-let-7f-2-3p, hsa-mir-1244, hsa-mir-1197, hsa-mir-124-3p, hsa-mir-1249-5p, hsa-mir-1253, and hsa-mir-1271-5p) and five hub genes (CENPE, ENPP3, GPR18, MDC1, and PLOD2). Conclusion: This study screened novel biological markers and investigated the differentiated circRNA-miRNA-mutated hub gene axis, which may play a pivotal role in the nosogenesis of PSS and RSS. Some circRNAs may be deemed new diagnostic or therapeutic targets that could be conducive to the future clinical treatment of SS.
Article
Full-text available
Introduction Extensive research is directed to uncover new biomarkers capable to stratify breast cancer patients into clinically relevant cohorts. However, the overall performance ranking of such marker candidates compared to other genes is virtually absent. Here, we present the ranking of all survival related genes in chemotherapy treated basal and estrogen positive / HER2 negative breast cancer. Methods We searched the GEO repository to uncover transcriptomic datasets with available follow-up and clinical data. After quality control and normalization, samples entered an integrated database. Molecular subtypes were designated using gene expression data. Relapse-free survival analysis was performed using Cox proportional hazards regression. False discovery rate was computed to combat multiple hypothesis testing. Kaplan-Meier plots were drawn to visualize the best performing genes. Results The entire database includes 7,830 unique samples from 55 independent datasets. Of those with available relapse-free survival time, 3,382 samples were estrogen receptor-positive and 696 were basal. In chemotherapy treated ER positive / ERBB2 negative patients the significant prognostic biomarker genes achieved hazard rates between 1.76 and 3.33 with a p value below 5.8E-04. The significant prognostic genes in adjuvant chemotherapy treated basal breast cancer samples reached hazard rates between 1.88 and 3.61 with a p value below 7.2E-04. Our integrated platform was extended enabling the validation of future biomarker candidates. Conclusions A reference ranking for all genes in two chemotherapy treated breast cancer cohorts is presented. The results help to neglect those with unlikely clinical significance and to focus future research on the most promising candidates.
Article
Full-text available
This article provides an update on the global cancer burden using the GLOBOCAN 2020 estimates of cancer incidence and mortality produced by the International Agency for Research on Cancer. Worldwide, an estimated 19.3 million new cancer cases (18.1 million excluding nonmelanoma skin cancer) and almost 10.0 million cancer deaths (9.9 million excluding nonmelanoma skin cancer) occurred in 2020. Female breast cancer has surpassed lung cancer as the most commonly diagnosed cancer, with an estimated 2.3 million new cases (11.7%), followed by lung (11.4%), colorectal (10.0 %), prostate (7.3%), and stomach (5.6%) cancers. Lung cancer remained the leading cause of cancer death, with an estimated 1.8 million deaths (18%), followed by colorectal (9.4%), liver (8.3%), stomach (7.7%), and female breast (6.9%) cancers. Overall incidence was from 2‐fold to 3‐fold higher in transitioned versus transitioning countries for both sexes, whereas mortality varied <2‐fold for men and little for women. Death rates for female breast and cervical cancers, however, were considerably higher in transitioning versus transitioned countries (15.0 vs 12.8 per 100,000 and 12.4 vs 5.2 per 100,000, respectively). The global cancer burden is expected to be 28.4 million cases in 2040, a 47% rise from 2020, with a larger increase in transitioning (64% to 95%) versus transitioned (32% to 56%) countries due to demographic changes, although this may be further exacerbated by increasing risk factors associated with globalization and a growing economy. Efforts to build a sustainable infrastructure for the dissemination of cancer prevention measures and provision of cancer care in transitioning countries is critical for global cancer control.
Article
Full-text available
CRISPR genetic screens in cancer cell models are a powerful tool to elucidate oncogenic mechanisms and to identify promising therapeutic targets. The Project Score database (https://score.depmap.sanger.ac.uk/) uses genome-wide CRISPR–Cas9 dropout screening data in hundreds of highly annotated cancer cell models to identify genes required for cell fitness and prioritize novel oncology targets. The Project Score database currently allows users to investigate the fitness effect of 18 009 genes tested across 323 cancer cell models. Through interactive interfaces, users can investigate data by selecting a specific gene, cancer cell model or tissue type, as well as browsing all gene fitness scores. Additionally, users can identify and rank candidate drug targets based on an established oncology target prioritization pipeline, incorporating genetic biomarkers and clinical datasets for each target, and including suitability for drug development based on pharmaceutical tractability. Data are freely available and downloadable. To enhance analyses, links to other key resources including Open Targets, COSMIC, the Cell Model Passports, UniProt and the Genomics of Drug Sensitivity in Cancer are provided. The Project Score database is a valuable new tool for investigating genetic dependencies in cancer cells and the identification of candidate oncology targets.
Article
Full-text available
HER2+ breast cancer (BC) is characterized by rapid growth, early recurrence, early metastasis, and chemoresistance. Trastuzumab is the most effective treatment for HER2+ BC and effectively reduces the risk of recurrence and death of patients. Resistance to trastuzumab results in cancer recurrence and metastasis, leading to poor prognosis of HER2+ BC. In the present study, we found that non-structural maintenance of chromosome condensin 1 complex subunit G (NCAPG) expression was highly upregulated in trastuzumab-resistant HER2+ BC. Ectopic NCAPG was positively correlated with tumor relapse and shorter survival in HER2+ BC patients. Moreover, overexpression of NCAPG promoted, while silencing of NCAPG reduced, the proliferative and anti-apoptotic capacity of HER2+ BC cells both in vitro and in vivo, indicating NCAPG reduces the sensitivity of HER2+ BC cells to trastuzumab and may confer trastuzumab resistance. Furthermore, our results suggest that NCAPG triggers a series of biological cascades by phosphorylating SRC and enhancing nuclear localization and activation of STAT3. To summarize, our study explores a crucial role for NCAPG in trastuzumab resistance and its underlying mechanisms in HER2+ BC, and suggests that NCAPG could be both a potential prognostic marker as well as a therapeutic target to effectively overcome trastuzumab resistance.
Article
Full-text available
Long intergenic non-coding RNA-Nucleotide Metabolism Regulator (lincNMR) is a long non-coding RNA (lncRNA) which is induced in hepatocellular carcinoma. Its depletion invokes a proliferation defect, triggers senescence and inhibits colony formation in liver, but also breast and lung cancer cells. Triple-label SILAC proteomics profiles reveal a deregulation of key cell cycle regulators in lincNMR-depleted cells like the key dNTP synthesizing enzymes RRM2, TYMS and TK1, implicating lincNMR in regulating nucleotide metabolism. LincNMR silencing decreases dNTP levels, while exogenous dNTPs rescues the proliferation defect induced by lincNMR depletion. In vivo RNA Antisense Purification (RAP-MS) identifies YBX1 as a direct interaction partner of lincNMR which regulates RRM2, TYMS and TK1 expression and binds to their promoter regions. In a Chick Chorioallantoic Membrane (CAM) in vivo model, lincNMR-depleted tumors are significantly smaller. In summary, we discover a lincRNA, lincNMR, which regulates tumor cell proliferation through a YBX1-RRM2-TYMS-TK1 axis governing nucleotide metabolism. Despite some well-characterized functions in cancer, the impact of most long non-coding RNAs remains unknown. Here, the authors discover the lncRNA lincNMR which is upregulated in cancer and drives cell proliferation by interacting with YBX1 and controlling nucleotide metabolism.
Article
Full-text available
The interplay between E2 and E3 enzymes regulates the polyubiquitination of substrates in eukaryotes. Among the several RING-domain E3 ligases in humans, many utilize two distinct E2s for polyubiquitination. For example, the cell cycle regulatory E3, human anaphase-promoting complex/cyclosome (APC/C), relies on UBE2C to prime substrates with ubiquitin (Ub) and UBE2S to extend polyubiquitin chains. However, the potential coordination between these steps in ubiquitin chain formation remains undefined. While numerous studies have unveiled how RING E3s stimulate individual E2s for Ub transfer, here we change perspective to describe a case where the chain-elongating E2 UBE2S feeds back and directly stimulates the E3 APC/C to promote substrate priming and subsequent multiubiquitination by UBE2C. Our work reveals an unexpected model for the mechanisms of RING E3-dependent ubiquitination and for the diverse and complex interrelationship between components of the ubiquitination cascade.
Article
PTEN tumor suppressor opposes the PI3K/Akt signaling pathway in the cytoplasm and maintains chromosomal integrity in the nucleus. Nucleus–cytoplasm shuttling of PTEN is regulated by ubiquitylation, SUMOylation and phosphorylation, and nuclear PTEN has been proposed to exhibit tumor-suppressive functions. Here we show that PTEN is conjugated by Nedd8 under high glucose conditions, which induces PTEN nuclear import without effects on PTEN stability. PTEN neddylation is promoted by the XIAP ligase and removed by the NEDP1 deneddylase. We identify Lys197 and Lys402 as major neddylation sites on PTEN. Neddylated PTEN accumulates predominantly in the nucleus and promotes rather than suppresses cell proliferation and metabolism. The nuclear neddylated PTEN dephosphorylates the fatty acid synthase (FASN) protein, inhibits the TRIM21-mediated ubiquitylation and degradation of FASN, and then promotes de novo fatty acid synthesis. In human breast cancer tissues, neddylated PTEN correlates with tumor progression and poor prognosis. Therefore, we demonstrate a previously unidentified pool of nuclear PTEN in the Nedd8-conjugated form and an unexpected tumor-promoting role of neddylated PTEN.
Article
RNase III DROSHA is upregulated in multiple cancers and contributes to tumor progression by hitherto unclear mechanisms. Here, we demonstrate that DROSHA interacts with β-Catenin to transactivate STC1 in an RNA cleavage-independent manner, contributing to breast cancer stem-like cell (BCSC) properties. DROSHA mRNA stability is enhanced by N⁶-methyladenosine (m⁶A) modification which is activated by AURKA in BCSCs. AURKA stabilizes METTL14 by inhibiting its ubiquitylation and degradation to promote DROSHA mRNA methylation. Moreover, binding of AURKA to DROSHA transcript further strengthens the binding of the m⁶A reader IGF2BP2 to stabilize m⁶A-modified DROSHA. In addition, wild-type DROSHA, but not an m⁶A methylation-deficient mutant, enhances BCSC stemness maintenance, while inhibition of DROSHA m⁶A modification attenuates BCSC traits. Our study unveils the AURKA-induced oncogenic m⁶A modification as a key regulator of DROSHA in breast cancer and identifies a novel DROSHA transcriptional function in promoting the BCSC phenotype.