Identification of Common Hub Genes and Key Molecular Pathways between ADPKD and Renal Cell Carcinomas

Document Type : Original Article

Authors

1 Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran

2 Azad University of Medical Sciences, Tehran, Iran

3 Department of Medical Genetics, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.

4 Division of Applied Bioinformatics, German Cancer Research Center DKFZ Heidelberg

5 Department of Computer Engineering, University of Kashan, Kashan, Iran

6 Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran

Abstract

Introduction: ADPKD, a genetic ailment, leads to the emergence of tiny sacs brimming with fluid, commonly known as cysts, within the kidneys which has an association with Renal Cell Carcinoma (RCC). We explore the gene signature shared between ADPKD and RCC based on integrated network analysis.
Methods: We searched the DisGeNET database to extract the overlapped genes (until November 2023) across the dominant autosomal disorder characterized by the growth of multiple cysts in the kidneys and all types of kidney cancers. Further, Enrichr was utilized for the identification of significant Gene ontology (GO) terms in the Kyoto Encyclopedia of Genes and Genome (KEGG) pathway. Further, the highest linkage hub genes were determined across the selected disorders through the protein-protein interaction (PPI) network construction for the overlapping genes using cytoHubba.
Results: We identified 187 common genes between ADPKD and urologic disorders. We identified 5 hub genes retrieved via Enrichr and Cytohubba analysis including TNF, JAK2, TGFB1, IL6, and EPO. These genes were mostly involved in molecular pathways and the KEGG pathway. The overlapping genes were most significantly related to the regulation of cell population proliferation (GO:0042127), pathways in cancer, pancreatic cancer, and receptor-ligand activity.
Conclusion: from a total number of 187 common genes, 5 key genes were mutual across ADPKD and all types of renal carcinoma. The identified feature could be potential targets in both disorders, even to manage malignancies in polycystic kidney disease.

Graphical Abstract

Identification of Common Hub Genes and Key Molecular Pathways between ADPKD and Renal Cell Carcinomas

Highlights

  • We explore the gene signature shared between ADPKD and RCC based on integrated network analysis.
  • The identified feature could be potential targets for ADPKD patients at risk of RCC.
  • ADPKD can be developed in the kidneys and has an association with Renal Cell Carcinoma.

Keywords

Main Subjects


Introduction

Autosomal Dominant kidney disease is a condition affecting the entire body, caused by a mutation in PKD1 or PKD2 genes located on chromosome 16 or 4, respectively. These genes are responsible for encoding PC1 and PC2 proteins, known as polycystin-1 and polycystin-2 (1-3).

Malfunctions in the functions of PC1 or PC2 result in the disruption of growth regulation, G protein functions, and both canonical and non-canonical WNT pathways (4). The second hit model states that additional somatic mutations lead to increased cyst growth. In addition to the normal renal involvement with the progressive expansion of the cyst that leads to the enlargement and extensive distortion of the kidney structure and finally to the final stage of kidney disease, multiple extra-renal manifestations such as cysts in other organs, diverticulosis, abdominal and inguinal hernia, vascular malformations (5, 6). Renal cancer is a prevalent type of cancer globally, and its occurrence is on the rise. The primary histological types comprise clear cell RCC (ccRCC), chromophobe RCC, and papillary RCC (chRCC)  (7, 8). A systematic review of 14299 primary renal tumor samples and 969 metastatic samples revealed a trend toward the increases of copy number alterations (CNAs) for both losses and gains and a significant difference between primary tumors and metastatic linked to BAP1 (9, 10). A majority of patients with RCC have an inactivated von Hippel-Lindau (VHL) tumor suppressor gene (11-14). Mutations involved in RCC lead to metabolic dysregulation, intramural heterogeneity, angiogenesis and harmful tumor microenvironment (TME) interference (15).

The Cancer Genome Atlas  Research Network reported 19 genes that were significantly mutated in ccRCC (16). It is noteworthy that the enhanced comprehension of the pathologic and signaling anomalies that are distinctive of ADPKD has unveiled distinct parallels with solid tumors.(17). The similarities between ADPKD and cancer are intriguing, particularly given the promising outcomes of recent studies - both preclinical and clinical - that have employed drugs initially designed for treating malignancies (18). ADPKD exhibits several cancer symptoms described by Hanahan and Weinberg in the literature. However, there exist crucial differences between ADPKD and cancer, such as the obligatory preservation of cell polarity, which is a fundamental characteristic of ADPKD, unlike cancer traits such as heightened proliferation or genomic instability. Nonetheless, ADPKD presents some similar defects to those observed in tumors (18). Therefore, the objection of this study is to explore the gene signature shared between ADPKD and RCC based on integrated network analysis.

 

Methods

The genes associated with ADPKD and renal cancers were downloaded independently. We managed to exclude the overlapped genes. Consequently, the function of common genes was defined and hub genes were detected. The flow chart of this study is shown in Figure 1.

 

 

Figure 1. Study workflow. GDA, Gene Disease Association; GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; DPI, disease probability index.

 

Data collection

A list of diseases related to autosomal polycystic disease and renal carcinoma was determined. Selected search items were as for ADPKD: “polycystic”,  “polycystic kidney”, “autosomal polycystic” and “polycystic kidney disease”; for RCC: “nephron”, “Renal carcinoma”, “renal malignancy”, “clear cell renal carcinoma”, “papillary renal cell carcinoma”, “chromophobe renal cell carcinoma”, which were searched in DisGenNET (19), one of the largest available platform with collected data based on human diseases genetics, (https://www.disgenet.org), then the codes related to that disease were extracted. Moreover, the commonly shared genes were identified via drawing a Venn diagram (20). Of 2797genes related to renal carcinomas of all types and 295 genes related to autosomal dominant polycystic disease, a shared group of 187 genes was identified in common between the two diseases as shown in Figure 2.

 

 

Figure 2. Ven diagram of shared genes between ADPKD and renal carcinoma.

 

Gene set enrichment analysis

To analyze the overlapped genes, we utilized the Enrichr web-based tool (https://maayanlab.cloud/Enrichr) for Gene Ontology assessments in three categories, namely Molecular Function (MF), Biological Process (BP), and Cellular Compartment (CC), and also for identifying Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways (21, 22). Significantly enriched Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, with adjusted P-values lower than 0.05, were identified. Further analysis was conducted on the overlapped genes within the pathways to determine the overrepresented genes within a set of pathways.

Constructing PPI Network to detect hub genes

To create protein interaction (PPI) networks of the overlapping genes, we utilized the STRING (Search Tool for the Retrieval of Interacting Genes/Proteins) database (version 11.5) (https://string-db.org), considering a confidence score greater than 0.4. We then employed the cytoHubba plugin (version 0.1) in the Cytoscape software (version 3.8.2) to identify potential hub genes (23, 24).

                                                       

Results

Genetic Correlation across ADPKD and RCC

The selected ADPKD and RCC were searched in DisGeNET and the codes and genes related to them were determined. Following removing duplicates, the pairwise-based analysis identified genetic overlap across the 13 selected disorders (Table 1).

Among 2797 ADPKD-associated genes, 187 genes were shared between ADPKD and RCC. When the genes with DisGeNET score > 0.1 were filtered, 187 genes were detected as common genes among the selected disorders. Remarkably, TGFB1, SNIA2, STAT6, CSF1, and IL6 were the common genes with the highest DisGeNET score.

 

Table 1. Shared genes of renal cell carcinoma with ADPKD

Renal Cell Carcinoma

Shared genes with ADPKD, n (%)

Clear cell type

98(52%)

Papillary

39(20%)

Chromophobe

50(26%)

 

GO and KEGG enrichment analysis

To prospect the potential biological function of 2797 common genes, GO and KEGG pathways enrichment analyses were conducted using the Enrichr, and the component with adjusted P-value ≤ 0.01 were selected. The genes were mainly enriched in the regulation of cell population pathway (GO:0042127) (BP), cytokine-mediated signaling pathway (GO:0019221) (BP), and receptor-ligand activity (GO:0048018) (MF). The 10 top enriched GO terms of overlapped genes between MS and UDs are shown in Figures 3-5. The GO results were consistent with those of the KEGG pathway analysis. For instance, the results revealed that overlapping genes were related to pathways in cancer, the AGE-RAGE signaling pathway in diabetic complications, the PI3k -Akt signaling pathway, and microRNAs in malignancies (Figures 3-5).

 

 

Figure 3. GO biological pathway

 

Figure 4. GO molecular function

 

Figure 5. GO cellular function

                                    

To prioritize candidate common genes, we further categorized them by the rank of disease-associated pathways that they are involved. Then, we ranked the 20 top genes for each GO term and pathway and performed a pairwise analysis (Table 1). According to our analysis, TNF, IL6, TGFB1, and, TGFA, EPO overlapped among biological process and molecular function terms, and KEGG pathway.  

 

Table 2. Go enrichment analysis of overlapped genes between ADPKD and RCC

GO_Biological_Process

Go_Molecular_Function

GO_Cellular_Component

KEGG-Pathway

CXCL9

TNF

SPARC

PIK3CB

CSF1

CSF1

CSF1          

PIK3CD

EPO

EPO

COL14A1

EPO

MYC

CXCL9

HP

EDNRA

TGFB1

TGFB1

TGFB1       

TGFB1

TIMP1

EDN1

ETFA

TNF

JUNB

PTH

CSF1

IL6

JAK2

EGF

JAK2          

JAK2

EDN1

IL18

EGFR

EDN1

TGFA

TGFA

GLS

TGFA

ADAM10

MIF

CST3

TP53

TSC2

TNF

MUC1

EGFR

TSC1

AGT

ERBB4

ERBB2

RUNX1

VEGFA

MAPK1

MAPK1

SFRP4

LGALS3

APOE

EDNRB

IL6

GH1

IL6             

TGFB2

AGTR1

CXCL10

PTGS2

STAT5B

TP53

IL6

MIF

SMAD2

TNF

SST

FGF23

HMOX1

AVPR2

IL1B

TNF

IL13

 

Identification of hub genes via PPI network

The PPI network of overlapped genes was constructed with 985 nodes using STRING. The degree was calculated using the cytoHubba plugin in Cytoscape software and the top 20 ranking genes, including AXIN1, MYCN, TWIST1, MMP11, AGTR, CASP1, STAT6, TNF, IL6, CSF, SNAI1, TGFB1, JAK2 were identified as hub genes (Figure 6).

 

 

Figure 6. PPI network and common hub genes

 

The hub genes were also searched among the genes which were determined as key genes with the highest over presentations in disease-associated pathways (Figure 7). According to our analysis, TNF, TGFB1 and JAK2, EPO, CSF1, and TGFA were detected as the overrepresented genes in biological process and molecular function terms as well as KEGG.


 

 

Figure 7. The hub genes among the genes, which were determined as keygens with the highest over presentations in disease-associated pathways

 

Discussion

We identified five hub genes between ADPKD and RCC based on integrated network analysis including TGFB1, SNIA2, STAT6, CSF1, and IL6. Numerous studies have confirmed that a wide variety of pathological pathways are involved in ADPKD. An integrative bioinformatics study on molecular pathway of CKD  by Zhou LT, and colleagues indicated to numerous key genes (e.g. OAS1, JUN, and FOS) can play critical roles in the progression of CKD (25). Transcriptomic profiles of different types of CKD indicated several previously known key CKD genes (e.g., TGFBI, COL4A1, and FCN1 (25). TGF-beta (transforming growth factor-beta) is one of our five-candidate genes between ADPKD and RCC. Systematically, TGF-β signaling, together with histone deacetylase 7 (HDAC7), suppresses tricarboxylic acid cycle, (TCA cycle), also called Krebs cycle expression. (26). 

The PPI network encoded by the common DEGs showed ten hub proteins (EPHB2, PRKAR2B, CAV1, ARHGEF12, HSP90B1, ITGA2B, BCL2L1, E2F1, TUBB1, and C3) identified common gene signature and pathways in COVID-19 patients with CKD comorbidity (27). A bioinformatics analysis and machine learning on CKD and non-alcoholic fatty liver disease (NAFLD) suggested four NAFLD-related genes (DUSP1, NR4A1, FOSB, ZFP36) as diagnostic markers in CKD patients with NAFLD.  Several studies indicated different genes in  IgA nephropathy (IgAN) (28, 29).  IL10, IGLL5 and POU2AF1 genes were selected as key genes and their targets were recognized as MAPK1 and PPKCA in the biomarkers associated with RCC microenvironment with therapeutic and prognostic value. A positive correlation between IL/POU2AF1 expression and abundance of six immune cells was observed (30).

The study by Grimaldi AM., as a comparative bioinformatic analysis on two major subtypes of RCC showed the importance of the CBX gene family.  CBX1, CBX6, and CBX7 were also expressively related to the tumor stage. Additionally, decreased expression levels of CBX1, CBX5, CBX6, CBX7, and increased expression of CBX8 were associated with poor prognosis (31). High mRNA expression of cyclin D1 (CCND1), fms-related tyrosine kinase 1 (FLT1), plasminogen (PLG), and von Willebrand factor (VWF)  are important in recurrent free survival of RCC patients (32).  Also, Using bioinformatics analysis exposed that Fibroblast Growth Factor 1 (FGF1) expression was abnormally lost in ccRCC which statistically suggestively connected to the patient's overall survival (OS) (33). 

 

Conclusion

In 187 common genes between ADPKD and RCC, the final 5 key genes TGFB1, SNIA2, STAT6, CSF1, and IL6 were mutual across. The identified feature could be potential targets for ADPKD patients at risk of RCC.

 

Acknowledgments

Special thanks to Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran.

 

Competing interests     

The authors declare no Conflict of Interest for this article.

 

Founding

There is no funding

 

Ethical statement

Not Applicable

 

Data Availability

All necessary data are included in the manuscript and no additional data are included.

 

Abbreviations

ADPKD            Autosomal dominant polycystic kidney disease 

BP                     Biological process

CC                    Cellular compartment

CKD                 Chronic Kidney Disease

CNAs                Copy number alterations

MF                    Molecular function

NAFLD             Non-alcoholic fatty liver disease

TME                  Tumor microenvironmental

VHL                  Von Hippel-Lindau

VWF                  Von Willebrand factor

  1. The polycystic kidney disease 1 gene encodes a 14 kb transcript and lies within a duplicated region on chromosome 16. The European Polycystic Kidney Disease Consortium. Cell. 1994;77(6):881-94. doi: 10.1016/0092-8674(94)90137-6. PubMed PMID: 8004675.
  2. Hughes J, Ward CJ, Peral B, Aspinwall R, Clark K, San Millán JL, et al. The polycystic kidney disease 1 (PKD1) gene encodes a novel protein with multiple cell recognition domains. Nat Genet. 1995;10(2):151-60. doi: 10.1038/ng0695-151. PubMed PMID: 7663510.
  3. Mochizuki T, Wu G, Hayashi T, Xenophontos SL, Veldhuisen B, Saris JJ, et al. PKD2, a gene for polycystic kidney disease that encodes an integral membrane protein. Science. 1996;272(5266):1339-42. doi: 10.1126/science.272.5266.1339. PubMed PMID: 8650545.
  4. Kim DY, Park JH. Genetic Mechanisms of ADPKD. Adv Exp Med Biol. 2016;933:13-22. doi: 10.1007/978-981-10-2041-4_2. PubMed PMID: 27730431.
  5. Cornec-Le Gall E, Alam A, Perrone RD. Autosomal dominant polycystic kidney disease. Lancet. 2019;393(10174):919-35. Epub 20190225. doi: 10.1016/s0140-6736(18)32782-x. PubMed PMID: 30819518.
  6. Chapman AB, Devuyst O, Eckardt KU, Gansevoort RT, Harris T, Horie S, et al. Autosomal-dominant polycystic kidney disease (ADPKD): executive summary from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2015;88(1):17-27. Epub 20150318. doi: 10.1038/ki.2015.59. PubMed PMID: 25786098; PubMed Central PMCID: PMCPMC4913350.
  7. Linehan WM, Ricketts CJ. The Cancer Genome Atlas of renal cell carcinoma: findings and clinical implications. Nat Rev Urol. 2019;16(9):539-52. Epub 20190705. doi: 10.1038/s41585-019-0211-5. PubMed PMID: 31278395.
  8. Aghamir SMK, Nasir Shirazi M, Khatami F. A Systematic Review of Circulating Tumor Cells in Renal Cell Carcinoma. Translational Research in Urology. 2021;3(1):10-8. doi: 10.22034/tru.2020.257587.1050.
  9. Bui TO, Dao VT, Nguyen VT, Feugeas JP, Pamoukdjian F, Bousquet G. Genomics of Clear-cell Renal Cell Carcinoma: A Systematic Review and Meta-analysis. Eur Urol. 2022;81(4):349-61. Epub 20220103. doi: 10.1016/j.eururo.2021.12.010. PubMed PMID: 34991918.
  10. Guitynavard F, Mousavibahar SH, Dadkhah Tehrani F, Eftekhar Javadi A, zia H. An Unusual Extrarenal Renal Cell Carcinoma: Case Report. Translational Research in Urology. 2019;1(1):23-6. doi: 10.22034/au.2020.227372.1015.
  11. Gnarra JR, Tory K, Weng Y, Schmidt L, Wei MH, Li H, et al. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat Genet. 1994;7(1):85-90. doi: 10.1038/ng0594-85. PubMed PMID: 7915601.
  12. Shuin T, Kondo K, Torigoe S, Kishida T, Kubota Y, Hosaka M, et al. Frequent somatic mutations and loss of heterozygosity of the von Hippel-Lindau tumor suppressor gene in primary human renal cell carcinomas. Cancer Res. 1994;54(11):2852-5. PubMed PMID: 8187067.
  13. Aghamir SMK, Heshmat R, Ebrahimi M, Ketabchi SE, Dizaji SP, Khatami F. The impact of succinate dehydrogenase gene (SDH) mutations in renal cell carcinoma (RCC): A systematic review. OncoTargets and therapy. 2019;12:7929.
  14. Clifford SC, Prowse AH, Affara NA, Buys CH, Maher ER. Inactivation of the von Hippel-Lindau (VHL) tumour suppressor gene and allelic losses at chromosome arm 3p in primary renal cell carcinoma: evidence for a VHL-independent pathway in clear cell renal tumourigenesis. Genes, chromosomes & cancer. 1998;22(3):200-9. Epub 1998/06/13. doi: 10.1002/(sici)1098-2264(199807)22:3<200::aid-gcc5>3.0.co;2-#. PubMed PMID: 9624531.
  15. Wolf MM, Kimryn Rathmell W, Beckermann KE. Modeling clear cell renal cell carcinoma and therapeutic implications. Oncogene. 2020;39(17):3413-26. Epub 20200302. doi: 10.1038/s41388-020-1234-3. PubMed PMID: 32123314; PubMed Central PMCID: PMCPMC7194123.
  16. Fenner A. Genetics: a molecular atlas of clear cell renal cell carcinoma. Nat Rev Clin Oncol. 2013;10(9):485. Epub 20130709. doi: 10.1038/nrclinonc.2013.122. PubMed PMID: 23836316.
  17. Seeger-Nukpezah T, Geynisman DM, Nikonova AS, Benzing T, Golemis EA. The hallmarks of cancer: relevance to the pathogenesis of polycystic kidney disease. Nature reviews Nephrology. 2015;11(9):515-34. Epub 2015/04/15. doi: 10.1038/nrneph.2015.46. PubMed PMID: 25870008; PubMed Central PMCID: PMCPMC5902186.
  18. Seeger-Nukpezah T, Geynisman DM, Nikonova AS, Benzing T, Golemis EA. The hallmarks of cancer: relevance to the pathogenesis of polycystic kidney disease. Nature Reviews Nephrology. 2015;11(9):515-34.
  19. Baxevanis AD. The importance of biological databases in biological discovery. Curr Protoc Bioinformatics. 2009;Chapter 1:Unit 1. doi: 10.1002/0471250953.bi0101s27. PubMed PMID: 19728285.
  20. Jia A, Xu L, Wang Y. Venn diagrams in bioinformatics. Brief Bioinform. 2021;22(5). doi: 10.1093/bib/bbab108. PubMed PMID: 33839742.
  21. Piñero J, Ramírez-Anguita JM, Saüch-Pitarch J, Ronzano F, Centeno E, Sanz F, et al. The DisGeNET knowledge platform for disease genomics: 2019 update. Nucleic Acids Res. 2020;48(D1):D845-d55. doi: 10.1093/nar/gkz1021. PubMed PMID: 31680165; PubMed Central PMCID: PMCPMC7145631.
  22. Xie Z, Bailey A, Kuleshov MV, Clarke DJB, Evangelista JE, Jenkins SL, et al. Gene Set Knowledge Discovery with Enrichr. Curr Protoc. 2021;1(3):e90. doi: 10.1002/cpz1.90. PubMed PMID: 33780170; PubMed Central PMCID: PMCPMC8152575.
  23. Chin CH, Chen SH, Wu HH, Ho CW, Ko MT, Lin CY. cytoHubba: identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 2014;8 Suppl 4(Suppl 4):S11. Epub 20141208. doi: 10.1186/1752-0509-8-s4-s11. PubMed PMID: 25521941; PubMed Central PMCID: PMCPMC4290687.
  24. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498-504. doi: 10.1101/gr.1239303. PubMed PMID: 14597658; PubMed Central PMCID: PMCPMC403769.
  25. Zhou L-T, Qiu S, Lv L-L, Li Z-L, Liu H, Tang R-N, et al. Integrative bioinformatics analysis provides insight into the molecular mechanisms of chronic kidney disease. Kidney and Blood Pressure Research. 2018;43(2):568-81.
  26. Nam H, Kundu A, Karki S, Brinkley G, Chandrashekar DS, Kirkman RL, et al. TGF-β signaling suppresses TCA cycle metabolism in renal cancer. bioRxiv. 2021:2021.02. 19.429599.
  27. Auwul MR, Zhang C, Rahman MR, Shahjaman M, Alyami SA, Moni MA. Network-based transcriptomic analysis identifies the genetic effect of COVID-19 to chronic kidney disease patients: A bioinformatics approach. Saudi Journal of Biological Sciences. 2021;28(10):5647-56.
  28. Chen X, Sun M. Identification of key genes, pathways and potential therapeutic agents for IgA nephropathy using an integrated bioinformatics analysis. Journal of the Renin-Angiotensin-Aldosterone System. 2020;21(2):1470320320919635.
  29. Qian W, Xiaoyi W, Zi Y. Screening and bioinformatics analysis of IgA nephropathy gene based on GEO databases. BioMed research international. 2019;2019.
  30. Zeng Q, Zhang W, Li X, Lai J, Li Z. Bioinformatic identification of renal cell carcinoma microenvironment-associated biomarkers with therapeutic and prognostic value. Life sciences. 2020;243:117273.
  31. Grimaldi AM, Affinito O, Salvatore M, Franzese M. CBX Family Members in Two Major Subtypes of Renal Cell Carcinoma: A Comparative Bioinformatic Analysis. Diagnostics. 2022;12(10):2452.
  32. Luo T, Chen X, Zeng S, Guan B, Hu B, Meng Y, et al. Bioinformatic identification of key genes and analysis of prognostic values in clear cell renal cell carcinoma. Oncology letters. 2018;16(2):1747-57.
  33. Zhang X, Wang Z, Zeng Z, Shen N, Wang B, Zhang Y, et al. Bioinformatic analysis identifying FGF1 gene as a new prognostic indicator in clear cell Renal Cell Carcinoma. Cancer Cell International. 2021;21:1-16.