Next Article in Journal
The Role of lncRNAs in Pig Muscle in Response to Cold Exposure
Previous Article in Journal
The Genome of the Korean Island-Originated Perilla citriodora ‘Jeju17’ Sheds Light on Its Environmental Adaptation and Fatty Acid and Lipid Production Pathways
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Survival and Enrichment Analysis of Epithelial–Mesenchymal Transition Genes in Bladder Urothelial Carcinoma

1
Albert Einstein College of Medicine, New York, NY 10461, USA
2
Professor of Pathology, University of Illinois at Chicago College of Medicine, Chicago, IL 60607, USA
*
Author to whom correspondence should be addressed.
Genes 2023, 14(10), 1899; https://doi.org/10.3390/genes14101899
Submission received: 28 August 2023 / Revised: 16 September 2023 / Accepted: 26 September 2023 / Published: 30 September 2023
(This article belongs to the Section Human Genomics and Genetic Diseases)

Abstract

:
The escalating prevalence of bladder cancer, particularly urothelial carcinoma, necessitates innovative approaches for prognosis and therapy. This study delves into the significance of genes related to epithelial–mesenchymal transition (EMT), a process inherently linked to carcinogenesis and comparatively better studied in other cancers. We examined 1184 EMT-related gene expression levels in bladder urothelial cancer cases through the TCGA dataset. Genes shown to be differentially expressed in relation to survival underwent further network and enrichment analysis to uncover how they might shape disease outcomes. Our in silico analysis revealed a subset of 32 genes, including those significantly represented in biological pathways such as VEGF signaling and bacterium response. In addition, these genes interact with genes involved in the JAK-STAT signaling pathway. Additionally, some of those 32 genes have been linked to immunomodulators such as chemokines CCL15 and CCL18, as well as to various immune cell infiltrates. Our findings highlight the prognostic utility of various EMT-related genes and identify possible modulators of their effect on survival, allowing for further targeted wet lab research and possible therapeutic intervention.

1. Introduction

Bladder cancer is the 10th most common malignancy worldwide with 573,278 new cases and 212,536 deaths in 2020 [1]. Urothelial carcinoma accounts for over 90% of bladder cancers, which costs the U.S. alone $4 billion annually [2]. The prevalence is predicted to continue to rise due to the increasing industrialization and urbanization in developing countries, and the aging population [3,4]. Well-studied risk factors include cigarette smoking and occupational exposures especially in metal workers, painters, and chemical process workers [5,6]. Various altered genes have been implicated in amplifying the effect of these environmental exposures including carcinogen detoxification genes like UDP Glucuronosyltransferase Family 1 Member A Complex Locus (UGT1A) and N-acetyltransferase 2 (NAT2) [7,8,9], fibroblast growth factor receptor 3 (FGFR 3) [10], p16, p53, retinoblastoma (RB), matrix metalloproteinases, genes involved in folate metabolism, and high activity metabolic activators like high activity P450 cytochrome enzymes [11,12,13,14,15,16,17]. Within the TNM staging used, T1 represents tumor invasion up to the muscular layer of the bladder, with further stages T2,3,4 representing further progression into the muscle, perivesicular layer, and adjacent structures and organs, respectively. Currently, treatment options include transurethral resection of bladder tumor (TURBT) and intravesical therapy for non-muscle invasive, while options for metastatic disease include radial cystectomy, neoadjuvant chemotherapy, and newer immunotherapies [2,18,19,20,21,22].
Several genes have been implicated in the development of urothelial cancer and metastases and used as targets for therapy. Some of these include LRP1B [23], ERRC2, FANCC, ATM, RB1 [24], p53 [25], and SLC14A1 [26]. There are currently no widely accepted bladder cancer screening programs, though biannual cystoscopies have been found to be efficacious in vulnerable subpopulations [27,28,29]. Currently follow-up is time-consuming and expensive, consisting of cystoscopy, imaging, and surgery; urine biomarkers are being studied to supplement those options [18,19].
Cells involved in the invasion of bladder cancer alter their surroundings and can also become transiently and reversibly plastic, turning into mesenchymal stem cells. This is the epithelial–mesenchymal transition (EMT), which is among the most relevant paradigm shifts in how we view cancer progression and can combat its growth. During EMT, epithelial cells lose their polarized, adhesive characteristics and gain a mesenchymal phenotype, enabling them to migrate and invade surrounding tissues [30,31]. Transcription factors including Snail, Zeb, and Twist aid in this process by repressing E-cadherin, an epithelial transmembrane protein [32]. In contrast to epithelial cells, mesenchymal carcinoma cells exhibit specific metabolic needs. As they undergo EMT, cancer cells finely regulate multiple metabolic pathways to support the demands of rapid cell proliferation [33]. The molecular pathways shown to be associated with EMT include Snail/Slug, Twist, Six1, Cripto, TGF-β, and Wnt/β-catenin [34]. The literature shows how genes such as CDH1, ZEB1, TGFB, CDH2, VIM, and TIMP1 have been linked to inducing the EMT phenotype, driving cell migration, and adapting to changing demands on the primary tumor [33,35,36].
In bladder cancer, various microRNAs (miRNAs) have been found to regulate proteins such as Smad7 or Twist1, either promoting or disrupting EMT and metastasis [37]. Understanding the mechanisms underlying EMT is crucial for developing targeted therapies to control cancer metastasis and may prove useful in treatment options going forward. Comparatively, there has been less work in this field in bladder cancer than in other cancers. This paper examines a multitude of EMT-related genes in relation to not only outcomes, but also the biologic networks and pathways which allow these genes to influence carcinogenesis and affect these outcomes.

2. Materials and Methods

2.1. Selection of Genes

To have a comprehensive overview of genes involved in the epithelial–mesenchymal transition, dbEMT 2.0 (http://dbemt.bioinfo-minzhao.org/ (accessed on 1 September 2022)), a database curated for focus on EMT-related genes, was utilized. A spreadsheet was generated with 1184 genes listed on the database, obtained from an initial PubMed abstract query for “Epithelial Mesenchymal Transition Genes” with the results mined for unique genes linked to EMT (see Supplemental Materials).

2.2. Survival Analysis

Publicly available cases from the NIH-funded “The Cancer Genome Atlas” (TCGA) project were utilized to examine gene expression pertaining to survival in bladder urothelial cancer (data portal: https://portal.gdc.cancer.gov/projects/TCGA-BLCA (accessed on 1 September 2022)). Kaplan–Meier plots were generated through the R2 platform (https://hgserver1.amc.nl/cgi-bin/r2/main.cgi (accessed on 1 September 2022)) using the TCGA dataset for “Bladder Urothelial Carcinoma”, n = 407. The built-in “KaplanScan” algorithm was used to divide mRNA gene expression into “high” versus “low” categories (n values for each based on KaplanScan groupings of expression). Overall survival was compared to follow-up time in months being analyzed. For multiple hypothesis testing, p-values were adjusted to a false discovery rate (FDR) of 0.05.

2.3. Expression Analysis

To compare normal versus tumor levels of those EMT genes which showed differential expression regarding survival, mRNA levels for the “Bladder Urothelial Carcinoma” TCGA dataset were analyzed with a Welch’s t-test through the UCSC Xena platform. This platform is a genome browser and visualization tool of genomic and phenotypic data for both public and private datasets (https://xena.ucsc.edu/ (accessed on 1 September 2022)). An FDR cutoff of 0.05 was used for significance. Violin plots were generated to visualize expression values.
Through the aforementioned R2 platform, the expression levels of the previously mentioned genes which showed differential expression in regard to survival were analyzed. mRNA gene levels were compared in respect to pathologic staging and, given the small n associated with cases of stage 1 bladder urothelial cancer in the TCGA dataset (n = 2), Kruskal–Wallis analysis with corresponding pairwise Welch’s t-tests were used. Again, p-values were adjusted and an FDR cutoff of 0.05 was used.

2.4. Network & Enrichment Analysis

GeneMANIA (http://www.genemania.org (accessed on 1 September 2022)) serves as a platform for visualizing diverse biological interactions encompassing co-expression, co-localization, and domain similarity. In this study, GeneMANIA, R (https://www.r-project.org/ (accessed on 1 September 2022)), and the Cytoscape platform (https://cytoscape.org/ (accessed on 1 September 2022)) were utilized to construct a gene–gene interaction network focusing on EMT-related genes exhibiting significant differential expression based on survival data.
Subsequently, the network analysis highlighted certain genes alongside the aforementioned EMT genes. These genes underwent enrichment analysis to shed light on their potential involvement in specific biological processes, using annotations from the Gene Ontology (GO), Kyoto Encyclopedia of Genes, and Genomes (KEGG) databases. It was carried out through the Metascape platform (http://metascape.org (accessed on 1 October 2022)). The criteria for this analysis included a minimum overlap of 3 genes and an enrichment threshold of 1.5. Statistical significance was set at p < 0.05.

2.5. Tumor Immune Microenvironment Analysis

The relationship between the EMT-related genes differentially expressed in connection with survival and the immune system in cases of bladder urothelial cancer was examined through the TISIDB and TIMER platform. Spearman’s correlations between mRNA gene expression of the EMT genes and clinically relevant immunoinhibitor and cytokine gene expressions were calculated and visualized. Those with a rho of >|0.40| were considered meaningful with a p-value of >0.01. Devolution methods were used to estimate the immune infiltration of a wide variety of immune cells based on gene expression on the TIMER platform. The Spearman’s correlation was adjusted based on tumor purity, with a rho of >|0.30| visualized.

3. Results

3.1. Survival Analysis

A myriad of EMT-related genes showed differential expression in normal versus cancer tissues. Kaplan–Meier plots (Figure 1) were generated using TCGA data on “Bladder Urothelial Carcinoma” for each of the 1184 genes mined from the EMTdb, with only 32 meeting significant cutoff following FDR correction (Table 1).
Genes with “high” mRNA expression leading to worse prognosis include those in Table 2.
On the other hand, genes which showed “low” mRNA expression leading to worse prognosis include those in Table 3.

3.2. Expression Analysis

Out of the 32 genes highlighted in the survival analysis, some of them also showed statistically significant expression levels when comparing normal versus tumor samples. This can be seen in the density plot (Figure 2) or in more detail in the sample violin plots (Figure 3); the rest of the genes and p-values from the violin plots in Supplemental Materials can be seen in Table 4. Some genes also showed significant differential expression based on tumor stage (Figure 4).
From the XENA browser, 19 normal tissues were compared with 407 TCGA cases. Following FDR correction (cut-off p-value is 0.003143), 10 genes were shown to be differentially expressed when considering overall survival (Table 5).
Several genes approach significance post-correction (Table 6).
Out of the same set of EMT-genes regarding survival, 10 out of the 32 showed differential expression when considering pathologic staging (Table 7).

3.3. Identification of Further Gene Interactions and Enriched Biological Processes

3.3.1. Network Analysis

Network analysis was employed to identify genes with interconnected relationships, utilizing factors such as physical interactions, co-localization, and co-expression data (Figure 5). The genes chosen for constructing each network analysis were the previously mentioned EMT-associated genes that displayed distinct expression patterns in survival outcomes.

3.3.2. Enrichment Analysis

Enrichment analysis was executed on two distinct gene sets: first, on the genes pinpointed in the network analysis, and additionally, on the genes situated at the periphery (“outer rim”) of the network analysis, apart from the initial EMT genes (Figure 6). Enrichment analysis of the initial 32 highlighted EMT genes showed numerous pathways significantly overrepresented in the cohort of genes, such as endothelial cell migration and regulation of cell shape, in line with the known physiologic processes involved with the epithelial–mesenchymal transition. However, other less canonically associated ones such as defense response to bacterium and carbohydrate response were also uncovered through the analysis. Regarding the genes listed in the network analysis, similar biologic processes such as the VEGFA signaling pathway was highlighted alongside other related ones such as HIF-1 survival signaling.

3.4. Correlation to Inflammation Mediators

3.4.1. Immunomodulator, Cytokine

Through examining for correlation between various immunomodulators and chemokines (see Appendix A, Table A1 for list), the following plots were generated (Figure 7), with the statistically and clinically significant ones (p < 0.05 post correction, |rho| > 0.4) being shown. Out of the 32 genes, 12 were shown to have at least significant correlation with an immunomodulator or chemokines (TBX3, NRP2, FN1, FOXA1, FBP1, ANXA1, LAMC2, HOOK1, NES, PTPN6, RUNX2), with the first four having at least 25 different immunomodulators or cytokines to be significantly correlated with (Table 8).

3.4.2. Immune Cell Infiltrate

Through the TIMER platform, deconvolution methods were used to estimate the amount of various immune cells (T cells CD4+, Tregs, B cells, Neutrophils, Monocytes, Macrophages, DCs, NK and Mast Cells; see Figure 8). Out of the 32 initial genes, 14 (ADAM17, AGER, ANXA1, ARMC8, FBP1, FN1, FOXA1, LAMC2, MAP2K1, NRP2, PTPN6, RUNX2, STIM2, TBX3) showed to have at least significant correlation (Spearman’s correlation > 0.05 following adjustment based on tumor purity, with a rho of >|0.30|; see Table 9).

4. Discussion

Overall, we see a robust cohort of EMT-related genes that are differentially expressed as pertaining to survival in bladder urothelial carcinoma. RUNX2, a gene most associated with cartilage production, has been linked to pancreatic cancer and to breast cancer progression through modulation of MicroRNAs and the metastasis-associated 1 (MTA1)/NuRD complex [38,39]. By activating the Wnt signaling pathway, ARMC8 has been linked to increased invasion in cutaneous squamous cell carcinoma and lung cancer [40,41,42]. Also implicated in the Wnt pathway, as well as the VEGF pathway, CEMIP (formerly known as KIA1199) has emerged via immunohistochemical studies as a possible biomarker for a variety of cancers [43,44,45]. The phosphatase INPP4B is an inhibitor of the Wnt pathway; its knockout has been linked to increased proliferation [46]. Knockout or downregulation of the transcription factor gene FOXA1 has also been linked to worse prognosis, altering the carcinogenic activity of the Snail/Twist1 axis in breast cancer as well as prostate cancer [47,48]. TBX3 has been linked to breast and cervical cancer proliferation, but it also inhibits the activity of the YAP/TAZ signaling pathway involved with cellular regeneration and growth [49,50,51].
Of note, PTPN6, a tyrosine phosphatase, affects cell growth and carcinogenesis in both bladder and colon cancer. Interestingly, overexpression of PTPN6 has been associated with worse prognosis and increased metastasis, while the opposite was seen in other studies [52,53]. Additionally, there were cases where our findings were incongruent with other cancers. PEBP4 expression was correlated with metastasis in colorectal and breast cancer but is significantly associated with better outcomes in our analysis. AGER (advanced glycosylation end-product-specific receptor) was shown to be associated with increased cell migration in cervical cancer, but the opposite as per our analysis [54]. The pro-carcinogenic effect that ART1 has in colorectal cancer was not seen in our Bladder TCGA analysis [55].
Interestingly enough, some of the genes shown to be differentially expressed in regard to survival were not differentially expressed when comparing normal tissue to tumor; nor did their expression correlate to pathologic staging. For the pathologic staging, only FN1, NRP2, FOXA1, NES, AGER, RUNX2, PTPN6, FBP1, TBX3, and STIM2 were shown to be significantly different.
While there may be differences in Stage I expression versus the other stages, our sample size (n = 2) was too small to reliably detect any except for in the expression levels of FOXA1. Instead, many of the differences were seen between Stage 2 versus 3 and 4. Histopathologically, this correlates with the expansion of the cancer through the muscle with possible lymph node and systemic metastasis. During this process, the pro-mobility changes which accompany the EMT would be fully evident. Our findings were mostly consistent with the survival analysis results, such as with RUNX2 which showed worse prognosis with higher levels, increasing expression from Stages 1 to 4.
Through the network analysis, we were able to see more genes through which the EMT-related genes can modulate and affect carcinogenesis. For example, VEGFA was indicated, a well-known member of a family of growth factors which has proliferative and anti-apoptotic effects [56]. It has been found to be highly expressed in hepatocellular carcinoma (HCC) and triple-negative breast cancer (TNBC). It is associated with worse prognosis in HCC [57] and significantly lower progression-free survival in TNBC treated with chemotherapy [58]. Additionally, SFRP2 was implicated, and recent immunohistochemistry work has linked this gene as a possible early biomarker of pancreatic and colon cancer [59,60]. Another gene highlighted in our analysis, CAV1, codes for a scaffolding protein and often predicts poor prognosis [61].
Enrichment analysis of the EMT-related genes further highlights the multiple modalities through which the EMT process itself encourages cancer growth and metastasis. Included among the highlighted cellular processes in our analysis are those regarding the establishment or maintenance of epithelial cell apical/basal polarity and positive regulation of cell migration. The VEGFA-VEGFR2 signaling pathway and associated angiogenesis, among the seminal hallmarks of cancer, were highlighted as the most overly enriched biologic process (p < 5.0 × 10−7). Other biological processes less typically associated with EMT such as “defense response to bacterium” and “signaling by interleukins” were picked up by the analysis. However, the same cytokine and inflammatory-mediated response to bacteria can help explain another way EMT-related genes play a role in cancer progression. The increased angiogenic and metabolic changes associated with certain proinflammatory states, which would help fend off a bacterial infection, can serve as fertile ground for initiation of cancer metastasis.
Among the most salient advances in our understanding of cancer progression is the complex role the immune system plays. Notable mediators of the immune system repeatedly occurred in our analysis. TGFBR1, a receptor for the TBFB growth factor, and CSF1R, a cytokine receptor, have been linked to pro-survival activity [62,63,64,65]. In other cancers, the chemokine CCL15 has been linked to tumor-associated macrophage recruitment and CCL18 has been linked to an immunosuppressive tumor microenvironment, allowing for evasion of the host’s immune system [66,67,68,69]. The most common immune cell type with a significant correlation was CD4+ T cells. While classically thought of as having an anti-tumor role, the varied subtypes not completely covered in our current deconvolution methods may play a paradoxical pro-tumor role. It is possible they do so by decreasing activation of other immune cells such as macrophages, warranting further study into the specific gene to immune cell interaction [70].
While this research represents a step forward in investigating the expression patterns of EMT-related genes, there exist certain limitations to these discoveries. These assessments rely on the levels of mRNA in a stable state as a proxy for protein levels. To partially address this, proteomic analysis of the highlighted genes in our study were examined in bladder cancer models through the depmap.org portal (https://depmap.org/portal/ (accessed on 12 September 2023)) and Cancer Cell Line Encyclopedia (CCLE), verifying expression of highlighted genes. Moreover, the functional behavior of these mRNA molecules could be subject to additional regulation by post-translational elements. Additionally, the techniques used to decipher immune cell infiltrations might be influenced by the tumor’s purity, contingent upon the specific formula employed. Ongoing efforts are dedicated to refining methods for accurately estimating cellular infiltrations based on mRNA levels. Overall, this study potentially serves as a starting point for future investigations aimed at directly scrutinizing the highlighted relationship between gene expression and prognosis, extending to the protein or enzymatic realm.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14101899/s1.

Author Contributions

Conceptualization, W.A. and A.K.-B.; methodology, W.A.; software, A.K.-B.; validation, W.A., W.X. and D.J.; formal analysis, W.A. and D.J.; resources, W.A.; data curation, W.X.; writing—original draft preparation, W.A.; writing—review and editing, W.A. and W.X.; visualization, D.J.; supervision, A.K.-B.; project administration, A.K.-B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Publicly available datasets were analyzed in this study. This data can be found through https://www.cancer.gov/ccg/research/genome-sequencing/tcga (accessed on 1 September 2022) or on the R2 platform as described above.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Below is the complete list of genes listed as “immunomodulators” in the aforementioned analysis.
Table A1. Complete list of genes included as “immunomodulators”.
Table A1. Complete list of genes included as “immunomodulators”.
ImmunomodulatorsChemokinesChemokines
ADORA2ACCL1CX3CL1
BTLACCL2CXCL1
CD160CCL3CXCL2
CD244CCL4CXCL3
CD274CCL5CXCL5
CD96CCL7CXCL6
CSF1RCCL8CXCL8
CTLA4CCL11CXCL9
HAVCR2CCL13CXCL10
IDO1CCL14CXCL11
IL10CCL15CXCL12
IL10RBCCL16CXCL13
KDRCCL17CXCL14
KIR2DL1CCL18CXCL16
KIR2DL3CCL19CXCL17
LAG3CCL20XCL1
LGALS9CCL21XCL2
PDCD1CCL22
PDCD1LG2CCL23
PVRL2CCL24
TGFB1CCL25
TGFBR1CCL26
TIGITCCL27
VTCN1CCL28
Table A2. Spearman’s correlation data for all EMT-related genes, grouped by EMT-gene of interest. Only correlations shown to be statistically significant (p < 0.05 post multiple hypothesis correction) and deemed clinically significant |rho| > 0.4 are depicted.
Table A2. Spearman’s correlation data for all EMT-related genes, grouped by EMT-gene of interest. Only correlations shown to be statistically significant (p < 0.05 post multiple hypothesis correction) and deemed clinically significant |rho| > 0.4 are depicted.
Gene of InterestImmunomodulator Generho
TBX3
CXCL16−0.434
CXCL13−0.401
CXCL11−0.482
CXCL10−0.512
CXCL9−0.458
CXCL5−0.474
CXCL3−0.470
CXCL2−0.453
CXCL1−0.424
CCL26−0.489
CCL23−0.415
CCL18−0.469
CCL150.474
CCL13−0.411
CCL8−0.535
CCL7−0.544
CCL5−0.443
CCL4−0.570
CCL3−0.571
TIGIT−0.416
TGFBR1−0.45
PDCD1LG2−0.654
PDCD1−0.4
LAG3−0.528
IL10−0.449
IDO1−0.464
HAVCR2−0.570
CTLA4−0.423
CSF1R−0.562
CD274−0.507
NRP2
CXCL130.454
CXCL120.647
CXCL110.426
CXCL100.453
CXCL90.490
CXCL20.408
CCL260.579
CCL230.461
CCL210.57
CCL190.426
CCL180.519
CCL130.488
CCL110.479
CCL80.525
CCL70.531
CCL50.432
CCL40.507
CCL30.501
CCL20.553
LAG30.434
TIGIT0.435
TGFBR10.550
PDCD1LG20.676
PDCD10.438
IL100.633
HAVCR20.633
CTLA40.460
CSF1R0.679
BTLA0.433
ADORA2A0.402
FOXA1
CXCL12−0.419
CXCL11−0.401
CXCL10−0.446
CXCL9−0.415
CXCL5−0.450
CXCL3−0.425
CXCL2−0.471
CCL26−0.555
CCL23−0.426
CCL21−0.408
CCL18−0.474
CCL150.467
CCL13−0.469
CCL8−0.581
CCL7−0.589
CCL5−0.461
CCL4−0.547
CCL3−0.594
CCL2−0.498
TGFBR1−0.442
TGFB1−0.414
PDCD1LG2−0.687
LAG3−0.518
IL10−0.520
HAVCR2−0.572
CTLA4−0.425
CSF1R−0.599
CD274−0.492
FN1
TGFBR10.570
TGFB10.410
PDCD1LG20.716
LAG30.431
IL100.648
HAVCR20.620
CSF1R0.657
CD2740.416
CXCL130.415
CXCL120.625
CXCL110.404
CXCL100.448
CXCL90.458
CXCL50.418
CXCL20.401
CCL260.554
CCL230.459
CCL210.539
CCL180.551
CCL130.522
CCL110.561
CCL70.540
CCL50.462
CCL40.505
CCL30.511
CCL20.524
FBP1
CCL150.592
CCL4−0.404
TGFBR1−0.455
PDCD1LG2−0.491
CD274−0.474
ANXA1
PDCD1LG20.504
CD2740.451
CCL15−0.414
CCL70.407
LAMC2
CXCL80.440
CXCL10.422
TGFB10.433
HOOK1
TGFB1−0.423
CSF1R−0.410
CCL23−0.403
NES
CXCL120.423
KDR0.405
SPRR2A
CXCL10.423
CXCL80.412
PTPN6
LGALS90.422
RUNX2
PDCD1LG20.451

References

  1. Bladder cancer statistics| World Cancer Research Fund International. Available online: https://www.wcrf.org/cancer-trends/bladder-cancer-statistics/ (accessed on 7 June 2023).
  2. Thompson, D.B.; Siref, L.E.; Feloney, M.P.; Hauke, R.J.; Agrawal, D.K. Immunological basis in the pathogenesis and treatment of bladder cancer. Expert. Rev. Clin. Immunol. 2015, 11, 265–279. [Google Scholar] [CrossRef] [PubMed]
  3. Ploeg, M.; Aben, K.K.H.; Kiemeney, L.A. The present and future burden of urinary bladder cancer in the world. World J. Urol. 2009, 27, 289. [Google Scholar] [CrossRef] [PubMed]
  4. Wong, M.C.S.; Fung, F.D.H.; Leung, C.; Cheung, W.W.L.; Goggins, W.B.; Ng, C.F. The global epidemiology of bladder cancer: A joinpoint regression analysis of its incidence and mortality trends and projection. Sci. Rep. 2018, 8, 1129. [Google Scholar] [CrossRef] [PubMed]
  5. Purdue, M.P.; Hutchings, S.J.; Rushton, L.; Silverman, D.T. The proportion of cancer attributable to occupational exposures. Ann. Epidemiol. 2015, 25, 188. [Google Scholar] [CrossRef]
  6. Cumberbatch, M.G.; Windsor-Shellard, B.; Catto, J.W.F. The contemporary landscape of occupational bladder cancer within the United Kingdom: A meta-analysis of risks over the last 80 years. BJU Int. 2017, 119, 100–109. [Google Scholar] [CrossRef]
  7. Figueroa, J.D.; Ye, Y.; Siddiq, A.; Garcia-Closas, M.; Chatterjee, N.; Prokunina-olsson, L.; Cortessis, V.K.; Kooperberg, C.; Cussenot, O.; Benhamou, S.; et al. Genome-wide association study identifies multiple loci associated with bladder cancer risk. Human Mol. Genet. 2014, 23, 1387–1398. [Google Scholar] [CrossRef]
  8. Garcia-Closas, M.; Rothman, N.; Figueroa, J.D.; Prokunina-Olsson, L.; Han, S.S.; Baris, D.; Jacobs, E.J.; Malats, N.; De Vivo, I.; Albanes, D.; et al. Common genetic polymorphisms modify the effect of smoking on absolute risk of bladder cancer. Cancer Res. 2013, 73, 2211–2220. [Google Scholar] [CrossRef]
  9. Engel, L.S.; Taioli, E.; Pfeiffer, R.; Garcia-Closas, M.; Marcus, P.M.; Lan, Q.; Boffetta, P.; Vineis, P.; Autrup, H.; Bell, D.A.; et al. Pooled analysis and meta-analysis of glutathione S-transferase M1 and bladder cancer: A HuGE review. Am. J. Epidemiol. 2002, 156, 95–109. [Google Scholar] [CrossRef]
  10. Figueroa, J.D.; Koutros, S.; Colt, J.S.; Kogevinas, M.; Garcia-Closas, M.; Real, F.X.; Friesen, M.C.; Baris, D.; Stewart, P.; Schwenn, M.; et al. Modification of Occupational Exposures on Bladder Cancer Risk by Common Genetic Polymorphisms. JNCI J. Natl. Cancer Inst. 2015, 107, 223. [Google Scholar] [CrossRef]
  11. Carlo, M.I.; Ravichandran, V.; Srinavasan, P.; Bandlamudi, C.; Kemel, Y.; Ceyhan-Birsoy, O.; Mukherjee, S.; Mandelker, D.; Chaim, J.; Knezevic, A.; et al. Cancer Susceptibility Mutations in Patients with Urothelial Malignancies. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 406–414. [Google Scholar] [CrossRef]
  12. Malats, N.; Bustos, A.; Nascimento, C.M.; Fernandez, F.; Rivas, M.; Puente, D.; Kogevinas, M.; Real, F.X. P53 as a prognostic marker for bladder cancer: A meta-analysis and review. Lancet Oncol. 2005, 6, 678–686. [Google Scholar] [CrossRef] [PubMed]
  13. Shariat, S.F.; Tokunaga, H.; Zhou, J.H.; Kim, J.H.; Ayala, G.E.; Benedict, W.F.; Lerner, S.P. p53, p21, pRB, and p16 expression predict clinical outcome in cystectomy with bladder cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2004, 22, 1014–1024. [Google Scholar] [CrossRef] [PubMed]
  14. Benedict, W.F.; Lerner, S.P.; Zhou, J.; Shen, X.; Tokunaga, H.; Czerniak, B. Level of retinoblastoma protein expression correlates with p16 (MTS-1/INK4A/CDKN2) status in bladder cancer. Oncogene 1999, 18, 1197–1203. [Google Scholar] [CrossRef]
  15. Kader, A.K.; Shao, L.; Dinney, C.P.; Schabath, M.B.; Wang, Y.; Liu, J.; Gu, J.; Grossman, H.B.; Wu, X. Matrix metalloproteinase polymorphisms and bladder cancer risk. Cancer Res. 2006, 66, 11644–11648. [Google Scholar] [CrossRef] [PubMed]
  16. Lin, J.; Spitz, M.R.; Wang, Y.; Schabath, M.B.; Gorlov, I.P.; Hernandez, L.M.; Pillow, P.C.; Grossman, H.B.; Wu, X. Polymorphisms of folate metabolic genes and susceptibility to bladder cancer: A case-control study. Carcinogenesis 2004, 25, 1639–1647. [Google Scholar] [CrossRef]
  17. Kaderlik, K.R.; Kadlubar, F.F. Metabolic polymorphisms and carcinogen-DNA adduct formation in human populations. Pharmacogenetics 1995, 5, S108–S117. [Google Scholar] [CrossRef]
  18. Dobruch, J.; Oszczudłowski, M. Bladder Cancer: Current Challenges and Future Directions. Medicina 2021, 57, 749. [Google Scholar] [CrossRef]
  19. Advanced Bladder Cancer Meta-analysis Collaboration. Neo-adjuvant chemotherapy for invasive bladder cancer. Cochrane Database Syst. Rev. 2004, 2004. [Google Scholar] [CrossRef]
  20. Pham, A.; Ballas, L.K. Trimodality therapy for bladder cancer: Modern management and future directions. Curr. Opin. Urol. 2019, 29, 210–215. [Google Scholar] [CrossRef]
  21. Ott, P.A.; Hu-Lieskovan, S.; Chmielowski, B.; Govindan, R.; Naing, A.; Bhardwaj, N.; Margolin, K.; Awad, M.M.; Hellmann, M.D.; Lin, J.J.; et al. A Phase Ib Trial of Personalized Neoantigen Therapy Plus Anti-PD-1 in Patients with Advanced Melanoma, Non-small Cell Lung Cancer, or Bladder Cancer. Cell 2020, 183, 347–362.e324. [Google Scholar] [CrossRef]
  22. Magers, M.J.; Lopez-Beltran, A.; Montironi, R.; Williamson, S.R.; Kaimakliotis, H.Z.; Cheng, L. Staging of bladder cancer. Histopathology 2019, 74, 112–134. [Google Scholar] [CrossRef] [PubMed]
  23. Langbein, S.; Szakacs, O.; Wilhelm, M.; Sukosd, F.; Weber, S.; Jauch, A.; Lopez Beltran, A.; Alken, P.; Kälble, T.; Kovacs, G. Alteration of the LRP1B gene region is associated with high grade of urothelial cancer. Lab. Investig. 2002, 82, 639–643. [Google Scholar] [CrossRef] [PubMed]
  24. Damrauer, J.S.; Beckabir, W.; Klomp, J.; Zhou, M.; Plimack, E.R.; Galsky, M.D.; Grivas, P.; Hahn, N.M.; O’Donnell, P.H.; Iyer, G.; et al. Collaborative study from the Bladder Cancer Advocacy Network for the genomic analysis of metastatic urothelial cancer. Nat. Commun. 2022, 13, 6658. [Google Scholar] [CrossRef]
  25. Habuchi, T.; Takahashi, R.; Yamada, H.; Ogawa, O.; Kakehi, Y.; Ogura, K.; Hamazaki, S.; Toguchida, J.; Ishizaki, K.; Fujita, J.; et al. Influence of cigarette smoking and schistosomiasis on p53 gene mutation in urothelial cancer. Cancer Res. 1993, 53, 3795–3799. [Google Scholar]
  26. Hou, R.; Kong, X.; Yang, B.; Xie, Y.; Chen, G. SLC14A1: A novel target for human urothelial cancer. Clin. Transl. Oncol. 2017, 19, 1438–1446. [Google Scholar] [CrossRef]
  27. Zlotta, A.R.; Roumeguere, T.; Kuk, C.; Alkhateeb, S.; Rorive, S.; Lemy, A.; Van Der Kwast, T.H.; Fleshner, N.E.; Jewett, M.A.S.; Finelli, A.; et al. Select screening in a specific high-risk population of patients suggests a stage migration toward detection of non-muscle-invasive bladder cancer. Eur. Urol. 2011, 59, 1026–1031. [Google Scholar] [CrossRef] [PubMed]
  28. PubMed Health. A service of the National Library of Medicine, N.I.o.H. In Bladder Cancer Treatment (PDQ®). Available online: https://www.ncbi.nlm.nih.gov/books/NBK65962/ (accessed on 7 June 2023).
  29. Sylvester, R.J.; Van Der Meijden, A.P.M.; Oosterlinck, W.; Witjes, J.A.; Bouffioux, C.; Denis, L.; Newling, D.W.W.; Kurth, K. Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: A combined analysis of 2596 patients from seven EORTC trials. Eur. Urol. 2006, 49, 466–477. [Google Scholar] [CrossRef]
  30. Friedl, P.; Mayor, R. Tuning Collective Cell Migration by Cell-Cell Junction Regulation. Cold Spring Harb. Perspect. Biol. 2017, 9, a029199. [Google Scholar] [CrossRef]
  31. Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. EMT: 2016. Cell 2016, 166, 21–45. [Google Scholar] [CrossRef]
  32. Friedl, P. Prespecification and plasticity: Shifting mechanisms of cell migration. Curr. Opin. Cell Biol. 2004, 16, 14–23. [Google Scholar] [CrossRef]
  33. Ramesh, V.; Brabletz, T.; Ceppi, P. Targeting EMT in Cancer with Repurposed Metabolic Inhibitors. Trends Cancer 2020, 6, 942–950. [Google Scholar] [CrossRef] [PubMed]
  34. Micalizzi, D.S.; Farabaugh, S.M.; Ford, H.L. Epithelial-mesenchymal transition in cancer: Parallels between normal development and tumor progression. J. Mammary Gland. Biol. Neoplasia 2010, 15, 117–134. [Google Scholar] [CrossRef] [PubMed]
  35. Zhu, W.; Leber, B.; Andrews, D.W. Cytoplasmic O-glycosylation prevents cell surface transport of E-cadherin during apoptosis. EMBO J. 2001, 20, 5999–6007. [Google Scholar] [CrossRef]
  36. Parsana, P.; Amend, S.R.; Hernandez, J.; Pienta, K.J.; Battle, A. Identifying global expression patterns and key regulators in epithelial to mesenchymal transition through multi-study integration. BMC Cancer 2017, 17, 447. [Google Scholar] [CrossRef]
  37. Sahib, A.S.; Fawzi, A.; Zabibah, R.S.; Koka, N.A.; Khudair, S.A.; Muhammad, F.A.; Hamad, D.A. miRNA/epithelial-mesenchymal axis (EMT) axis as a key player in cancer progression and metastasis: A focus on gastric and bladder cancers. Cell Signal. 2023, 110881. [Google Scholar] [CrossRef]
  38. Yin, X.; Teng, X.; Ma, T.; Yang, T.; Zhang, J.; Huo, M.; Liu, W.; Yang, Y.; Yuan, B.; Yu, H.; et al. RUNX2 recruits the NuRD(MTA1)/CRL4B complex to promote breast cancer progression and bone metastasis. Cell Death Differ. 2022, 29, 2203–2217. [Google Scholar] [CrossRef] [PubMed]
  39. Pranavkrishna, S.; Sanjeev, G.; Akshaya, R.L.; Rohini, M.; Selvamurugan, N. Regulation of Runx2 and Its Signaling Pathways by MicroRNAs in Breast Cancer Metastasis. Curr. Protein Pept. Sci. 2021, 22, 534–547. [Google Scholar] [CrossRef]
  40. Amin, A.; Bukhari, S.; Mokhdomi, T.A.; Anjum, N.; Wafai, A.H.; Wani, Z.; Manzoor, S.; Koul, A.M.; Amin, B.; Ain, Q.U.; et al. Comparative proteomics and global genome-wide expression data implicate role of ARMC8 in lung cancer. Asian Pac. J. Cancer Prev. 2015, 16, 3691–3696. [Google Scholar] [CrossRef]
  41. Xie, C.; Jiang, G.; Fan, C.; Zhang, X.; Zhang, Y.; Miao, Y.; Lin, X.; Wu, J.; Wang, L.; Liu, Y.; et al. ARMC8α promotes proliferation and invasion of non-small cell lung cancer cells by activating the canonical Wnt signaling pathway. Tumour Biol. 2014, 35, 8903–8911. [Google Scholar] [CrossRef]
  42. Li, X.; Zhang, C.; Yuan, Y.; Wang, Y.; Lu, S.; Zhou, Z.; Zhen, P.; Zhou, M. Downregulation of ARMC8 promotes tumorigenesis through activating Wnt/β-catenin pathway and EMT in cutaneous squamous cell carcinomas. J. Dermatol. Sci. 2021, 102, 184–192. [Google Scholar] [CrossRef]
  43. Domanegg, K.; Sleeman, J.P.; Schmaus, A. CEMIP, a Promising Biomarker That Promotes the Progression and Metastasis of Colorectal and Other Types of Cancer. Cancers 2022, 14, 5093. [Google Scholar] [CrossRef] [PubMed]
  44. Hua, Q.; Zhang, B.; Xu, G.; Wang, L.; Wang, H.; Lin, Z.; Yu, D.; Ren, J.; Zhang, D.; Zhao, L.; et al. CEMIP, a novel adaptor protein of OGT, promotes colorectal cancer metastasis through glutamine metabolic reprogramming via reciprocal regulation of β-catenin. Oncogene 2021, 40, 6443–6455. [Google Scholar] [CrossRef] [PubMed]
  45. Rodrigues, G.; Hoshino, A.; Kenific, C.M.; Matei, I.R.; Steiner, L.; Freitas, D.; Kim, H.S.; Oxley, P.R.; Scandariato, I.; Casanova-Salas, I.; et al. Tumour exosomal CEMIP protein promotes cancer cell colonization in brain metastasis. Nat. Cell Biol. 2019, 21, 1403–1412. [Google Scholar] [CrossRef] [PubMed]
  46. Liu, H.; Paddock, M.N.; Wang, H.; Murphy, C.J.; Geck, R.C.; Navarro, A.J.; Wulf, G.M.; Elemento, O.; Haucke, V.; Cantley, L.C.; et al. The INPP4B Tumor Suppressor Modulates EGFR Trafficking and Promotes Triple-Negative Breast Cancer. Cancer Discov. 2020, 10, 1226–1239. [Google Scholar] [CrossRef] [PubMed]
  47. Arruabarrena-Aristorena, A.; Maag, J.L.V.; Kittane, S.; Cai, Y.; Karthaus, W.R.; Ladewig, E.; Park, J.; Kannan, S.; Ferrando, L.; Cocco, E.; et al. FOXA1 Mutations Reveal Distinct Chromatin Profiles and Influence Therapeutic Response in Breast Cancer. Cancer Cell 2020, 38, 534–550.e539. [Google Scholar] [CrossRef] [PubMed]
  48. Jägle, S.; Busch, H.; Freihen, V.; Beyes, S.; Schrempp, M.; Boerries, M.; Hecht, A. SNAIL1-mediated downregulation of FOXA proteins facilitates the inactivation of transcriptional enhancer elements at key epithelial genes in colorectal cancer cells. PLoS Genet. 2017, 13, e1007109. [Google Scholar] [CrossRef] [PubMed]
  49. Dong, L.; Lyu, X.; Faleti, O.D.; He, M.L. The special stemness functions of Tbx3 in stem cells and cancer development. Semin. Cancer Biol. 2019, 57, 105–110. [Google Scholar] [CrossRef]
  50. Khan, S.F.; Burmeister, C.A.; Scott, D.J.; Sinkala, M.; Ramburan, A.; Wu, H.T.; Schäfer, G.; Katz, A.A.; Prince, S. TBX3 Promotes Cervical Cancer Proliferation and Migration via HPV E6 and E7 Signaling. Mol. Cancer Res. 2023, 21, 345–358. [Google Scholar] [CrossRef]
  51. Liang, B.; Zhou, Y.; Qian, M.; Xu, M.; Wang, J.; Zhang, Y.; Song, X.; Wang, H.; Lin, S.; Ren, C.; et al. TBX3 functions as a tumor suppressor downstream of activated CTNNB1 mutants during hepatocarcinogenesis. J. Hepatol. 2021, 75, 120–131. [Google Scholar] [CrossRef]
  52. Shen, C.; Liu, J.; Wang, J.; Yang, X.; Niu, H.; Wang, Y. The Analysis of PTPN6 for Bladder Cancer: An Exploratory Study Based on TCGA. Dis. Markers 2020, 2020, 4312629. [Google Scholar] [CrossRef]
  53. Liu, G.; Zhang, Y.; Huang, Y.; Yuan, X.; Cao, Z.; Zhao, Z. PTPN6-EGFR Protein Complex: A Novel Target for Colon Cancer Metastasis. J. Oncol. 2022, 2022, 7391069. [Google Scholar] [CrossRef] [PubMed]
  54. Zhu, X.; Zhou, L.; Li, R.; Shen, Q.; Cheng, H.; Shen, Z.; Zhu, H. AGER promotes proliferation and migration in cervical cancer. Biosci. Rep. 2018, 38, BSR20171329. [Google Scholar] [CrossRef] [PubMed]
  55. Tang, Y.; Li, M.; Wang, Y.L.; Threadgill, M.D.; Xiao, M.; Mou, C.F.; Song, G.L.; Kuang, J.; Yang, X.; Yang, L.; et al. ART1 promotes starvation-induced autophagy: A possible protective role in the development of colon carcinoma. Am. J. Cancer Res. 2015, 5, 498–513. [Google Scholar] [PubMed]
  56. Melincovici, C.S.; Boşca, A.B.; Şuşman, S.; Mărginean, M.; Mihu, C.; Istrate, M.; Moldovan, I.M.; Roman, A.L.; Mihu, C.M. Vascular endothelial growth factor (VEGF)-key factor in normal and pathological angiogenesis. Rom. J. Morphol. Embryol. 2018, 59, 455–467. [Google Scholar] [PubMed]
  57. Xu, K.; Wu, C.L.; Wang, Z.X.; Wang, H.J.; Yin, F.J.; Li, W.D.; Liu, C.C.; Fan, H.N. VEGF Family Gene Expression as Prognostic Biomarkers for Alzheimer’s Disease and Primary Liver Cancer. Comput. Math. Methods Med. 2021, 2021, 3422393. [Google Scholar] [CrossRef]
  58. Taha, F.M.; Zeeneldin, A.A.; Helal, A.M.; Gaber, A.A.; Sallam, Y.A.; Ramadan, H.; Moneer, M.M. Prognostic value of serum vascular endothelial growth factor in Egyptian females with metastatic triple negative breast cancer. Clin. Biochem. 2009, 42, 1420–1426. [Google Scholar] [CrossRef]
  59. Charles Jacob, H.K.; Signorelli, R.; Charles Richard, J.L.; Kashuv, T.; Lavania, S.; Middleton, A.; Gomez, B.A.; Ferrantella, A.; Amirian, H.; Tao, J.; et al. Identification of novel early pancreatic cancer biomarkers KIF5B and SFRP2 from “first contact” interactions in the tumor microenvironment. J. Exp. Clin. Cancer Res. 2022, 41, 258. [Google Scholar] [CrossRef]
  60. Müller, D.; Győrffy, B. DNA methylation-based diagnostic, prognostic, and predictive biomarkers in colorectal cancer. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188722. [Google Scholar] [CrossRef]
  61. Goetz, J.G.; Lajoie, P.; Wiseman, S.M.; Nabi, I.R. Caveolin-1 in tumor progression: The good, the bad and the ugly. Cancer Metastasis Rev. 2008, 27, 715–735. [Google Scholar] [CrossRef]
  62. Kretzschmar, M.; Doody, J.; Timokhina, I.; Massagué, J. A mechanism of repression of TGFbeta/ Smad signaling by oncogenic Ras. Genes Dev. 1999, 13, 804–816. [Google Scholar] [CrossRef]
  63. Ranganathan, P.; Agrawal, A.; Bhushan, R.; Chavalmane, A.K.; Kalathur, R.K.; Takahashi, T.; Kondaiah, P. Expression profiling of genes regulated by TGF-beta: Differential regulation in normal and tumour cells. BMC Genom. 2007, 8, 98. [Google Scholar] [CrossRef] [PubMed]
  64. Murga-Zamalloa, C.; Rolland, D.C.M.; Polk, A.; Wolfe, A.; Dewar, H.; Chowdhury, P.; Onder, O.; Dewar, R.; Brown, N.A.; Bailey, N.G.; et al. Colony-Stimulating Factor 1 Receptor (CSF1R) Activates AKT/mTOR Signaling and Promotes T-Cell Lymphoma Viability. Clin. Cancer Res. 2020, 26, 690–703. [Google Scholar] [CrossRef] [PubMed]
  65. Wen, J.; Wang, S.; Guo, R.; Liu, D. CSF1R inhibitors are emerging immunotherapeutic drugs for cancer treatment. Eur. J. Med. Chem. 2023, 245, 114884. [Google Scholar] [CrossRef] [PubMed]
  66. Gao, Y.; Zhou, Z.; Lu, S.; Huang, X.; Zhang, C.; Jiang, R.; Yao, A.; Sun, B.; Wang, X. Chemokine CCL15 Mediates Migration of Human Bone Marrow-Derived Mesenchymal Stem Cells Toward Hepatocellular Carcinoma. Stem Cells 2016, 34, 1112–1122. [Google Scholar] [CrossRef]
  67. Yin, X.; Han, S.; Song, C.; Zou, H.; Wei, Z.; Xu, W.; Ran, J.; Tang, C.; Wang, Y.; Cai, Y.; et al. Metformin enhances gefitinib efficacy by interfering with interactions between tumor-associated macrophages and head and neck squamous cell carcinoma cells. Cell Oncol. 2019, 42, 459–475. [Google Scholar] [CrossRef]
  68. Azzaoui, I.; Yahia, S.A.; Chang, Y.; Vorng, H.; Morales, O.; Fan, Y.; Delhem, N.; Ple, C.; Tonnel, A.B.; Wallaert, B.; et al. CCL18 differentiates dendritic cells in tolerogenic cells able to prime regulatory T cells in healthy subjects. Blood 2011, 118, 3549–3558. [Google Scholar] [CrossRef]
  69. Martinez, F.O.; Gordon, S.; Locati, M.; Mantovani, A. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: New molecules and patterns of gene expression. J. Immunol. 2006, 177, 7303–7311. [Google Scholar] [CrossRef]
  70. Ben Khelil, M.; Godet, Y.; Abdeljaoued, S.; Borg, C.; Adotévi, O.; Loyon, R. Harnessing Antitumor CD4(+) T Cells for Cancer Immunotherapy. Cancers 2022, 14, 260. [Google Scholar] [CrossRef]
Figure 1. Subset of Kaplan–Meier plots based on mRNA expression of EMT-related genes which showed to be differentially expressed in relation to overall survival. Each plot lists the numbers of pts in “high” versus “low” mRNA expression cohorts based on the KaplanScan grouping algorithm. (A,B) show subset of Kaplan-Meier plots, based on gene expression levels of ADAM17 and ADER respectively. See Supplemental Figure S1 for Kaplan–Meier plots of all genes shown to be differentially expressed.
Figure 1. Subset of Kaplan–Meier plots based on mRNA expression of EMT-related genes which showed to be differentially expressed in relation to overall survival. Each plot lists the numbers of pts in “high” versus “low” mRNA expression cohorts based on the KaplanScan grouping algorithm. (A,B) show subset of Kaplan-Meier plots, based on gene expression levels of ADAM17 and ADER respectively. See Supplemental Figure S1 for Kaplan–Meier plots of all genes shown to be differentially expressed.
Genes 14 01899 g001
Figure 2. Density plot showing expression of normal versus tumor expressions for all selected genes which had both TCGA and GTEX data.
Figure 2. Density plot showing expression of normal versus tumor expressions for all selected genes which had both TCGA and GTEX data.
Genes 14 01899 g002
Figure 3. Subset of violin plot showing mRNA expression between primary tumors (Blue, n = 407) and surrounding normal tissue (Red, n = 19). For full listing, see Supplemental Figure S2. (A,B) show subset of violin plots, showing primary versus normal solid tissue violin plots for NR2F2 and TBX3 respectively.
Figure 3. Subset of violin plot showing mRNA expression between primary tumors (Blue, n = 407) and surrounding normal tissue (Red, n = 19). For full listing, see Supplemental Figure S2. (A,B) show subset of violin plots, showing primary versus normal solid tissue violin plots for NR2F2 and TBX3 respectively.
Genes 14 01899 g003
Figure 4. Bar plots showing log2 median mRNA expression levels of the EMT-survival related genes which also showed significant differences between any of the stages (Stage 1 n = 2, Stage 2 n = 130, Stage 3 n = 140, Stage 4 n = 134). (AJ) show genes for which expression levels were significantly different (stage plots of FN1, NRP2, FOXA1, NES, AGER, RUNX2, PTPN6, FBP1, TBX3, and STIM 2 respectively).
Figure 4. Bar plots showing log2 median mRNA expression levels of the EMT-survival related genes which also showed significant differences between any of the stages (Stage 1 n = 2, Stage 2 n = 130, Stage 3 n = 140, Stage 4 n = 134). (AJ) show genes for which expression levels were significantly different (stage plots of FN1, NRP2, FOXA1, NES, AGER, RUNX2, PTPN6, FBP1, TBX3, and STIM 2 respectively).
Genes 14 01899 g004
Figure 5. Network analysis showing 20 of the most highlighted genes. The analysis was conducted utilizing factors such as physical and anticipated interactions, protein co-localization, and shared DNA domains, alongside various other attributes. Connections between genes based on physical interactions are highlighted in red, shared pathways in blue, shared protein domains in yellow, predicted in orange, co-expression in purple, genetic interactions in green, and co-localization in navy.
Figure 5. Network analysis showing 20 of the most highlighted genes. The analysis was conducted utilizing factors such as physical and anticipated interactions, protein co-localization, and shared DNA domains, alongside various other attributes. Connections between genes based on physical interactions are highlighted in red, shared pathways in blue, shared protein domains in yellow, predicted in orange, co-expression in purple, genetic interactions in green, and co-localization in navy.
Genes 14 01899 g005
Figure 6. Enrichment analysis of the EMT genes involved in survival (a) as well as enrichment analysis highlighted in the aforementioned network analysis (b). Labeled are statistically enriched terms which are biologic pathways selected from KEGG and other hallmark gene sets. Additionally, for both the EMT genes and network highlighted gene ((c) and (d), respectively), the representative terms were converted into a network layout with each circle representing a single biologic process, grouped into larger “themes” as labeled in the color key. The size of the circle represents the amount of analyzed genes within that term.
Figure 6. Enrichment analysis of the EMT genes involved in survival (a) as well as enrichment analysis highlighted in the aforementioned network analysis (b). Labeled are statistically enriched terms which are biologic pathways selected from KEGG and other hallmark gene sets. Additionally, for both the EMT genes and network highlighted gene ((c) and (d), respectively), the representative terms were converted into a network layout with each circle representing a single biologic process, grouped into larger “themes” as labeled in the color key. The size of the circle represents the amount of analyzed genes within that term.
Genes 14 01899 g006
Figure 7. Sample Spearman’s correlation graphs for TBX3 (full table of all EMT-related genes in Appendix A, Table A2), grouped by EMT-gene of interest. Only correlations shown to be statistically significant (p < 0.05 post multiple hypothesis correction) and deemed clinically significant |rho| > 0.4 are depicted both in the figure and included in the summary below. Subset shown, with (A) showing TBX3 expression correlation with CXCL16 expression and (B) showing correlation between TBX3 and CXCL13 expression; for other genes NRP2, FN1, FOXA1, FBP1, ANXA1, LAMC2, HOOK1, NES, RUNX2, and PTPN6 see Supplemental Figure S3.
Figure 7. Sample Spearman’s correlation graphs for TBX3 (full table of all EMT-related genes in Appendix A, Table A2), grouped by EMT-gene of interest. Only correlations shown to be statistically significant (p < 0.05 post multiple hypothesis correction) and deemed clinically significant |rho| > 0.4 are depicted both in the figure and included in the summary below. Subset shown, with (A) showing TBX3 expression correlation with CXCL16 expression and (B) showing correlation between TBX3 and CXCL13 expression; for other genes NRP2, FN1, FOXA1, FBP1, ANXA1, LAMC2, HOOK1, NES, RUNX2, and PTPN6 see Supplemental Figure S3.
Genes 14 01899 g007
Figure 8. Correlations of various gene expression levels to tumor purity and immune infiltration. Only correlations to immune infiltration shown to be statistically significant (p < 0.05 post multiple hypothesis correction) and deemed clinically significant |rho| > 0.3 are depicted both in the figure and included in the summary below. Subset shown, for other genes ANXA1, ARMC8, FBP1, FN1, FOXA1, LAMC2, MAP2K1, NRP2, PTPN6, RUNX2, STIM2, TBX3 see Supplemental Figure S2. (A) shows correlation between ADAM17 gene expression and tumor purity (amount of non-cancerous cells in tumor sample) and (B) shows correlation between ADAM17 expression level and calculated macrophage infiltration.
Figure 8. Correlations of various gene expression levels to tumor purity and immune infiltration. Only correlations to immune infiltration shown to be statistically significant (p < 0.05 post multiple hypothesis correction) and deemed clinically significant |rho| > 0.3 are depicted both in the figure and included in the summary below. Subset shown, for other genes ANXA1, ARMC8, FBP1, FN1, FOXA1, LAMC2, MAP2K1, NRP2, PTPN6, RUNX2, STIM2, TBX3 see Supplemental Figure S2. (A) shows correlation between ADAM17 gene expression and tumor purity (amount of non-cancerous cells in tumor sample) and (B) shows correlation between ADAM17 expression level and calculated macrophage infiltration.
Genes 14 01899 g008
Table 1. Tabulated data from Kaplan–Meier plots based on mRNA expression of EMT-related genes which showed to be differentially expressed in regard to overall survival.
Table 1. Tabulated data from Kaplan–Meier plots based on mRNA expression of EMT-related genes which showed to be differentially expressed in regard to overall survival.
Genep-ValueExpression in Worse Prognosis
ADAM178.65 × 10−6low
AGER4.13 × 10−7high
ANXA11.80 × 10−6low
ARMC83.99 × 10−8low
ART11.36 × 10−3high
BBC35.28 × 10−6high
CEMIP1.83 × 10−5low
ELSPBP11.65 × 10−3high
FBP13.43 × 10−5high
FN11.11 × 10−5low
FOXA18.51 × 10−5high
HOOK14.71 × 10−12high
HTN11.02 × 10−3high
IL224.18 × 10−4high
INPP4B2.37 × 10−5high
LAMC21.98 × 10−9low
LYPD33.33 × 10−5low
MAP2K13.73 × 10−7low
NES2.91 × 10−9low
NR2F21.34 × 10−6low
NRP25.85 × 10−5low
PDCD6IP1.31 × 10−5low
PEBP42.16 × 10−4high
PRKCI2.65 × 10−4low
PTPN62.03 × 10−7high
RUNX27.19 × 10−8low
SCEL2.27 × 10−7low
SLC9A3R13.78 × 10−4low
SOX33.92 × 10−4high
SPRR2A1.17 × 10−5low
STIM22.11 × 10−11high
TBX32.50 × 10−4high
Table 2. List of genes and p-values for which high mRNA expression is correlated with worse prognosis.
Table 2. List of genes and p-values for which high mRNA expression is correlated with worse prognosis.
Genep-Value
ADAM170.0182
ANXA10.0433
ARMC80.0009
CEMIP0.0300
FN10.0443
LAMC20.0050
LYPD30.0461
MAP2K10.0419
NES0.0413
NR2F20.0495
NRP20.0481
PDCD6IP0.0311
PRKCI0.0457
RUNX20.0007
SCEL0.0018
SLC9A3R10.0467
SPRR2A0.0342
Table 3. List of genes and p-values for which low mRNA expression is correlated with worse prognosis.
Table 3. List of genes and p-values for which low mRNA expression is correlated with worse prognosis.
Genep-Value
AGER0.0441
ART10.0446
BBC30.0317
ELSPBP10.0486
FBP10.0359
FOXA10.0356
HOOK10.0011
HTN10.0478
IL220.0389
INPP4B0.0314
PEBP40.0322
PTPN60.0137
SOX30.0324
STIM20.0500
TBX30.0482
Table 4. Violin plot genes and p-values.
Table 4. Violin plot genes and p-values.
Genep-ValueHigher Expression
ADAM178.31 × 10−4Primary tumor
AGER2.77 × 10−2Primary tumor
ANXA11.41 × 10−2Normal tissue
ARMC82.35 × 10−4Normal tissue
ART11.73 × 10−1Normal tissue
BBC35.39 × 10−5Primary tumor
CEMIP/KIAA11991.93 × 10−3Primary tumor
ELSPBP12.52 × 10−1Normal tissue
FBP17.76 × 10−1Normal tissue
FN13.88 × 10−1Primary tumor
FOXA11.64 × 10−1Primary tumor
HOOK15.13 × 10−3Primary tumor
IL229.30 × 10−2Normal tissue
INPP4B2.06 × 10−2Primary tumor
LAMC21.05 × 10−4Primary tumor
LYPD33.03 × 10−1Primary tumor
MAP2K12.88 × 10−3Primary tumor
NES8.36 × 10−8Normal tissue
NR2F23.04 × 10−1Normal tissue
NRP21.24 × 10−4Normal tissue
PDCD6IP3.67 × 10−1Primary tumor
PEBP42.17 × 10−4Normal tissue
PRKCI4.31 × 10−3Primary tumor
PTPN63.14 × 10−3Primary tumor
RUNX21.90 × 10−2Primary tumor
SCEL2.05 × 10−1Primary tumor
SLC9A3R18.95 × 10−3Primary tumor
SOX33.35 × 10−2Normal tissue
SPRR2A2.77 × 10−1Primary tumor
STIM28.06 × 10−1Normal tissue
TBX37.13 × 10−1Primary tumor
Table 5. List of genes differentially expressed regarding overall survival when comparing normal versus tumor samples with p-values.
Table 5. List of genes differentially expressed regarding overall survival when comparing normal versus tumor samples with p-values.
Genep-Value
ADAM170.0008
ARMC80.0002
BBC30.0001
CEMIP0.0019
LAMC20.0001
MAP2K10.0029
NES<0.0001
NRP20.0001
PEBP40.0002
PTPN60.0031
Table 6. List of genes that approach significance after FDR correction with p-values (cutoff p value is 0.003143).
Table 6. List of genes that approach significance after FDR correction with p-values (cutoff p value is 0.003143).
Genep-Value
HOOK10.0051
HTN10.0069
SLC9A3R10.0090
Table 7. List of stages that showed significantly different expression of the relevant gene based off TCGA datasets.
Table 7. List of stages that showed significantly different expression of the relevant gene based off TCGA datasets.
GeneDifferent Stages
FN12 vs. 3 (p = 1.22 × 10−8), 2 vs. 4 (4.20 × 10−12)
NRP22 vs. 4 (p = 2.10 × 10−8)
FOXA11 vs. 3 (p = 5.08 × 10−3)
NES2 vs. 3 (p = 1.71 × 10−5)
AGER2 vs. 3 (p = 2.00 × 10−3), 2 vs. 4 (p = 2.39 × 10−5)
RUNX22 vs. 4 (p = 2.99 × 10−4)
PTPN62 vs. 4 (p = 7.24 × 10−3)
FBP12 vs. 3 (p = 5.65 × 10−3)
TBX32 vs. 3 (p = 3.56 × 10−3)
STIM22 vs. 4 (p = 3.97 × 10−5)
Table 8. Summary of Spearman’s correlation data between EMT-related gene and various immunomodulators, grouped by EMT-gene of interest.
Table 8. Summary of Spearman’s correlation data between EMT-related gene and various immunomodulators, grouped by EMT-gene of interest.
GeneImmunomodulator
(Positive Correlation)
Immunomodulator
(Negative Correlation)
TBX3 CXCL16, CXCL13, CXCL11, CXCL10, CXCL9, CXCL5, CXCL3, CXCL2, CXCL1, CCL26, CCL23, CCL18, CCL13, CCL8, CCL7, CCL5, CCL4, CCL3, TIGIT, TGFBR1, PDCD1LG2, PDCD1, LAG3, IL10, IDO1, HAVCR2, CTLA4, CSF1R, CD274
NRP2CXCL13, CXCL12, CXCL11, CXCL10, CXCL9, CXCL2, CCL26, CCL23, CCL21, CCL19, CCL18, CCL13, CCL11, CCL8, CCL7, CCL5, CCL4, CCL3, CCL2, LAG3, TIGIT, TGFBR1, PDCD1LG2, PDCD1, IL10, HAVCR2, CTLA4, CSF1R, BTLA, ADORA2A
FN1TGFBR1, TGFB1, PDCD1LG2, LAG3, IL10, HAVCR2, CSF1R, CD274, CXCL13, CXCL12, CXCL11, CXCL10, CXCL9, CXCL5, CXCL2, CCL26, CCL23, CCL21, CCL18, CCL13, CCL11, CCL7, CCL5, CCL4, CCL3, CCL2
FOXA1CCL15CXCL12, CXCL11, CXCL10, CXCL9, CXCL5, CXCL3, CXCL2, CCL26, CCL23, CCL21, CCL18, CCL13, CCL8, CCL7, CCL5, CCL4, CCL3, CCL2, TGFBR1, TGFB1, PDCD1LG2, LAG3, IL10, HAVCR2, CTLA4, CSF1R, CD274
FBP1CCL15CCL4, TGFBR1, PDCD1LG2, CD274
ANXA1PDCD1LG2, CD274, CCL7
SPRR2ACXCL8, TGFB1
LAMC2CXCL8, CXCL1, TGFB1
HOOK1TGFB1, CSF1R, CCL23
NESCXCL12, KDR
PTPN6LGAGLS9
RUNX2PDCD1LG2
Table 9. Summary of Spearman’s correlation data between EMT-related gene and various immune cell types, grouped by EMT-gene of interest.
Table 9. Summary of Spearman’s correlation data between EMT-related gene and various immune cell types, grouped by EMT-gene of interest.
GeneImmune Infiltrates
(Positive Correlation)
Immune Infiltrates
(Negative Correlation)
ADAM17Macrophage, Neutrophil
AGER T cell CD8+
ANX1T cell CD8+, Neutrophil, Myeloid dendritic cell
ARMC8Macrophage
FBP1 T cell CD8+, Neutrophil, Myeloid dendritic cell
FN1T cell CD4+, T cell CD8+, Macrophage, Myeloid dendritic cell
FOXA1T cell CD4+T cell CD8+, Myeloid dendritic cell
LAMC2T cell CD8+, Neutrophil, Myeloid dendritic cell
MAP2K1T cell CD8+, Neutrophil
NRP2T cell CD8+, Macrophage, Myeloid dendritic cell
PTPN6 B cell
RUNX2T cell CD8+, Myeloid dendritic cell
STIM2T cell CD4+
TBX3 T cell CD8+, T cell CD4+, Neutrophil, Myeloid dendritic cell
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ali, W.; Xiao, W.; Jacobs, D.; Kajdacsy-Balla, A. Survival and Enrichment Analysis of Epithelial–Mesenchymal Transition Genes in Bladder Urothelial Carcinoma. Genes 2023, 14, 1899. https://doi.org/10.3390/genes14101899

AMA Style

Ali W, Xiao W, Jacobs D, Kajdacsy-Balla A. Survival and Enrichment Analysis of Epithelial–Mesenchymal Transition Genes in Bladder Urothelial Carcinoma. Genes. 2023; 14(10):1899. https://doi.org/10.3390/genes14101899

Chicago/Turabian Style

Ali, Waleed, Weirui Xiao, Daniel Jacobs, and Andre Kajdacsy-Balla. 2023. "Survival and Enrichment Analysis of Epithelial–Mesenchymal Transition Genes in Bladder Urothelial Carcinoma" Genes 14, no. 10: 1899. https://doi.org/10.3390/genes14101899

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop