Psoriasis Associated Hub Genes Revealed by Weighted Gene Co-Expression Network Analysis

Document Type : Original Article

Authors

1 Department of Biostatistics and Epidemiology, School of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran

2 Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran

3 Department of Biostatistics, School of Public Health and Modeling of Non-communicable Disease Research Center, Hamadan University of Medical Sciences, Hamadan, Iran

Abstract

Objective: Psoriasis, an immune-mediated disorder, is a multifactorial disease of unidentified cause. This study aims to discover the possible biomarkers of this papulosquamous skin disease.

Materials and Methods: The gene chip GSE55201, resulted from an experimental study, including 44 psoriasis patients and 30 healthy controls was downloaded from GEO and weighted gene co-expression network analysis was utilized to identify the hub genes. The key modules were determined using the module eigenvalues. We used biological functions, cellular components, and molecular functions in the Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes enrichment analysis in the gene metabolic pathway were used for enrichment analysis.

Results: Adjacency matrix was built by using power adjacency function and the power to turn the correlation to adjacency matrix was 4 with a topology fit index of 0.92. Using the weighted gene co-expression network analysis, 11 modules were identified. The green-yellow module eigenvalues were significantly associated with psoriasis (Pearson correlation=0.53, p<0.001). Candidate hub genes were determined by their higher connectivity and relationship with module eigenvalue. The genes including SIGLEC8, IL5RA, CCR3, RNASE2, CPA3, GATA2, c-KIT, and PRSS33 were recorded as the hub genes.

Conclusion: In summary we can conclude that SIGLEC8, IL5RA, CCR3, RNASE2, CPA3, GATA2, c-KIT, and PRSS33 have an important role in the immune response regulation and could be considered as a potential diagnostic biomarker and therapeutic target for Psoriasis.

Keywords


  1. Rendon A, Schäkel K. Psoriasis pathogenesis and treatment. Int J Mol Sci. 2019; 20(6): 1475.
  2. Stern RS, Nijsten T, Feldman SR, Margolis DJ, Rolstad T. Psoriasis is common, carries a substantial burden even when not extensive, and is associated with widespread treatment dissatisfaction. J Investig Dermatol Symp Proc. 2004; 9(2): 136-139.
  3. Javitz HS, Ward MM, Farber E, Nail L, Vallow SG. The direct cost of care for psoriasis and psoriatic arthritis in the United States. J Am Acad Dermatol. 2002; 46(6): 850-860.
  4. Tapak L, Afshar S, Afrasiabi M, Ghasemi MK, Alirezaei P. Application of genetic algorithm-based support vector machine in identification of gene expression signatures for psoriasis classification: a hybrid model. Biomed Res Int. 2021; 2021: 5520710.
  5. Parisi R, Iskandar IYK, Kontopantelis E, Augustin M, Griffiths CEM, Ashcroft DM, et al. National, regional, and worldwide epidemiology of psoriasis: systematic analysis and modelling study. BMJ. 2020; 369: m1590.
  6. Parisi R, Symmons DP, Griffiths CE, Ashcroft DM; Identification and Management of Psoriasis and Associated ComorbidiTy (IMPACT) project team. Global epidemiology of psoriasis: a systematic review of incidence and prevalence. J Invest Dermatol. 2013; 133(2): 377- 385.
  7. Armstrong AW, Read C. Pathophysiology, clinical presentation, and treatment of psoriasis: a review. JAMA. 2020; 323(19): 1945-1960.
  8. Xie J, Li H, Chen L, Cao Y, Hu Y, Zhu Z, et al. A novel pyroptosisrelated lncRNA signature for predicting the prognosis of skin cutaneous melanoma. Int J Gen Med. 2021; 14: 6517-6527.
  9. Johnson-Huang LM, Lowes MA, Krueger JG. Putting together the psoriasis puzzle: an update on developing targeted therapies. Dis Model Mech. 2012; 5(4): 423-433.
  10. Shi K, Bing ZT, Cao GQ, Guo L, Cao YN, Jiang HO, et al. Identify the signature genes for diagnose of uveal melanoma by weight gene co-expression network analysis. Int J Ophthalmol. 2015; 8(2): 269-274.
  11. Liu H, Liu M, You H, Li X, Li X. Oncogenic network and Hub genes for natural killer/T-cell lymphoma utilizing WGCNA. Front Oncol. 2020; 10: 223.
  12. Zhao W, Langfelder P, Fuller T, Dong J, Li A, Hovarth S. Weighted gene coexpression network analysis: state of the art. J Biopharm Stat. 2010; 20(2): 281-300.
  13. Wang CQF, Suárez-Fariñas M, Nograles KE, Mimoso CA, Shrom D, Dow ER, et al. IL-17 induces inflammation-associated gene products in blood monocytes, and treatment with ixekizumab reduces their expression in psoriasis patient blood. J Invest Dermatol. 2014; 134(12): 2990-2993.
  14. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008; 9: 55.
  15. 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: S11.
  16. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009; 4(1): 44-57
  17. Bigler J, Rand HA, Kerkof K, Timour M, Russell CB. Cross-study homogeneity of psoriasis gene expression in skin across a large expression range. PLoS One. 2013; 8(1): e52242.
  18. Choudhary S, Pradhan D, Khan NS, Singh H, Thomas G, Jain AK. Decoding psoriasis: integrated bioinformatics approach to understand Hub genes and involved pathways. Curr Pharm Des. 2020; 26(29): 3619-3630
  19. Gao LJ, Shen J, Ren YN, Shi JY, Wang DP, Cao JM. Discovering novel hub genes and pathways associated with the pathogenesis of psoriasis. Dermatol Ther. 2020; 33(6): e13993.
  20. Zhang YJ, Sun YZ, Gao XH, Qi RQ. Integrated bioinformatic analysis of differentially expressed genes and signaling pathways in plaque psoriasis. Mol Med Rep. 2019; 20(1): 225-235.
  21. Xing L, Wu T, Yu L, Zhou N, Zhang Z, Pu Y, et al. Exploration of biomarkers of psoriasis through combined multiomics analysis. Mediators Inflamm. 2022; 2022: 7731082.
  22. Su W, Wei Y, Huang B, Ji J. Identification of Hub genes and immune infiltration in psoriasis by bioinformatics method. Front Genet. 2021; 12: 606065.
  23. Yue Q, Li Z, Zhang Q, Jin Q, Zhang X, Jin G. Identification of novel Hub genes associated with psoriasis using integrated bioinformatics analysis. Int J Mol Sci. 2022; 23(23): 15286.
  24. Nickles MA, Huang K, Chang YS, Tsoukas MM, Sweiss NJ, Perkins DL, et al. Gene co-expression networks identifies common Hub genes between cutaneous sarcoidosis and discoid lupus erythematosus. Front Med (Lausanne). 2020; 7: 606461.
  25. Schanin J, Gebremeskel S, Korver W, Falahati R, Butuci M, Haw TJ, et al. A monoclonal antibody to Siglec-8 suppresses non-allergic airway inflammation and inhibits IgE-independent mast cell activation. Mucosal Immunol. 2021; 14(2): 366-376.
  26. Youngblood BA, Leung J, Falahati R, Williams J, Schanin J, Brock EC, et al. Discovery, function, and therapeutic targeting of siglec-8. Cells. 2020; 10(1): 19.
  27. Wang W, Sun Y, Fu Y, Yu X, Li M. Interaction of C1GALT1-IL5RA on the susceptibility to IgA nephropathy in Southern Han Chinese. J Hum Genet. 2013; 58(1): 40-46.
  28. Ellinghaus E, Stuart PE, Ellinghaus D, Nair RP, Debrus S, Raelson JV, et al. Genome-wide meta-analysis of psoriatic arthritis identifies susceptibility locus at REL. J Invest Dermatol. 2012; 132(4): 1133-1140.
  29. Zayed M, Iohara K, Watanabe H, Nakashima M. CCR3 antagonist protects against induced cellular senescence and promotes rejuvenation in periodontal ligament cells for stimulating pulp regeneration in the aged dog. Sci Rep. 2020; 10(1): 8631.
  30. Gerber BO, Zanni MP, Uguccioni M, Loetscher M, Mackay CR, Pichler WJ, et al. Functional expression of the eotaxin receptor CCR3 in T lymphocytes co-localizing with eosinophils. Curr Biol. 1997; 7(11): 836-843.
  31. Geng R, Liu H, Wang W. Differential expression of six Rnase2 and three Rnase3 paralogs identified in blunt snout bream in response to aeromonas hydrophila infection. Genes (Basel). 2018; 9(2): 95.
  32. Sun D, Han C, Sheng J. The role of human ribonuclease A family in health and diseases: a systematic review. iScience. 2022; 25(11): 105284.
  33. Bing PF, Xia W, Wang L, Zhang YH, Lei SF, Deng FY. Common marker genes identified from various sample types for systemic lupus erythematosus. PLoS One. 2016; 11(6): e0156234.
  34. Akula S, Hellman L, Avilés FX, Wernersson S. Analysis of the mast cell expressed carboxypeptidase A3 and its structural and evolutionary relationship to other vertebrate carboxypeptidases. Dev Comp Immunol. 2022; 127: 104273.
  35. Aleksiejczuk M, Gromotowicz-Poplawska A, Marcinczyk N, Przylipiak A, Chabielska E. The expression of the renin-angiotensin-aldosterone system in the skin and its effects on skin physiology and pathophysiology. J Physiol Pharmacol. 2019; 70(3): 325-336.
  36. Vicente C, Conchillo A, García-Sánchez MA, Odero MD. The role of the GATA2 transcription factor in normal and malignant hematopoiesis. Crit Rev Oncol Hematol. 2012; 82(1): 1-17.
  37. Sahoo SS, Kozyra EJ, Wlodarski MW. Germline predisposition in myeloid neoplasms: Unique genetic and clinical features of GATA2 deficiency and SAMD9/SAMD9L syndromes. Best Pract Res Clin Haematol. 2020; 33(3): 101197.
  38. Amarnani AA, Poladian KR, Marciano BE, Daub JR, Williams SG, Livinski AA, et al. A panoply of rheumatological manifestations in patients with GATA2 deficiency. Sci Rep. 2020; 10(1): 8305.
  39. Chen C, Darrow AL, Qi JS, D’Andrea MR, Andrade-Gordon P. A novel serine protease predominately expressed in macrophages. Biochem J. 2003; 374(Pt 1): 97-107.
  40. Toyama S, Okada N, Matsuda A, Morita H, Saito H, Fujisawa T, et al. Human eosinophils constitutively express a unique serine protease, PRSS33. Allergol Int. 2017; 66(3): 463-471.