Browse > Article
http://dx.doi.org/10.3345/cep.2020.00619

Expression profiling of cultured podocytes exposed to nephrotic plasma reveals intrinsic molecular signatures of nephrotic syndrome  

Panigrahi, Stuti (Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College)
Pardeshi, Varsha Chhotusing (Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College)
Chandrasekaran, Karthikeyan (Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College)
Neelakandan, Karthik (Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College)
PS, Hari (Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College)
Vasudevan, Anil (Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College)
Publication Information
Clinical and Experimental Pediatrics / v.64, no.7, 2021 , pp. 355-363 More about this Journal
Abstract
Background: Nephrotic syndrome (NS) is a common renal disorder in children attributed to podocyte injury. However, children with the same diagnosis have markedly variable treatment responses, clinical courses, and outcomes, suggesting molecular heterogeneity. Purpose: This study aimed to explore the molecular responses of podocytes to nephrotic plasma to identify specific genes and signaling pathways differentiating various clinical NS groups as well as biological processes that drive injury in normal podocytes. Methods: Transcriptome profiles from immortalized human podocyte cell line exposed to the plasma of 8 subjects (steroid-sensitive nephrotic syndrome [SSNS], n=4; steroid-resistant nephrotic syndrome [SRNS], n=2; and healthy adult individuals [control], n=2) were generated using microarray analysis. Results: Unsupervised hierarchical clustering of global gene expression data was broadly correlated with the clinical classification of NS. Differential gene expression (DGE) analysis of diseased groups (SSNS or SRNS) versus healthy controls identified 105 genes (58 up-regulated, 47 down-regulated) in SSNS and 139 genes (78 up-regulated, 61 down-regulated) in SRNS with 55 common to SSNS and SRNS, while the rest were unique (50 in SSNS, 84 genes in SRNS). Pathway analysis of the significant (P≤0.05, -1≤ log2 FC ≥1) differentially expressed genes identified the transforming growth factor-β and Janus kinase-signal transducer and activator of transcription pathways to be involved in both SSNS and SRNS. DGE analysis of SSNS versus SRNS identified 2,350 genes with values of P≤0.05, and a heatmap of corresponding expression values of these genes in each subject showed clear differences in SSNS and SRNS. Conclusion: Our study observations indicate that, although podocyte injury follows similar pathways in different clinical subgroups, the pathways are modulated differently as evidenced by the heatmap. Such transcriptome profiling with a larger cohort can stratify patients into intrinsic subtypes and provide insight into the molecular mechanisms of podocyte injury.
Keywords
Nephrotic syndrome; Child; Podocytes; Microarray;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Larkins N, Kim S, Craig J, Hodson E. Steroid-sensitive nephrotic syndrome: An evidence-based update of immunosuppressive treatment in children. Arch Dis Child 2016;101:404-8.   DOI
2 Ramanathan ASK, Vijayan M, Rajagopal S, Rajendiran P, Senguttuvan P. WT1 and NPHS2 gene mutation analysis and clinical management of steroid-resistant nephrotic syndrome. Mol Cell Biochem 2017;426:177-81.   DOI
3 Kim SH, Park SJ, Han KH, Kronbichler A, Saleem MA, Oh J, et al. Pathogenesis of minimal change nephrotic syndrome: an immunological concept. Korean J Pediatr 2016;59:205-11.   DOI
4 Peterson KS, Huang JF, Zhu J, D'Agati V, Liu X, Miller N, et al. Characterization of heterogeneity in the molecular pathogenesis of lupus nephritis from transcriptional profiles of laser-captured glomeruli. J Clin Invest 2004;113:1722-33.   DOI
5 Wang Z, Wang Z, Zhou Z, Ren Y. Crucial genes associated with diabetic nephropathy explored by microarray analysis. BMC Nephrol 2016;17:128.   DOI
6 Otalora L, Chavez E, Watford D, Tueros L, Correa M, Nair V, et al. Identification of glomerular and podocyte-specific genes and pathways activated by sera of patients with focal segmental glomerulosclerosis. PLoS One 2019;14:e0222948.   DOI
7 Jain S, De Petris L, Hoshi M, Akilesh S, Chatterjee R, Liapis H. Expression profiles of podocytes exposed to high glucose reveal new insights into early diabetic glomerulopathy. Lab Invest 2011;91:488-98.   DOI
8 Saleem MA, O'Hare MJ, Reiser J, Coward RJ, Inward CD, Farren T, et al. A conditionally immortalized human podocyte cell line demonstrating nephrin and podocin expression. J Am Soc Nephrol 2002;13:630-8.   DOI
9 Smyth GK, Ritchie M, Thorne N, Wettenhall J, Shi W, Hu Y. limma: linear models for microarray and RNA-seq data user's guide. Melbourne (Australia): Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, 2016.
10 Gavala ML, Hill LM, Lenertz LY, Karta MR, Bertics PJ. Activation of the transcription factor FosB/activating protein-1 (AP-1) is a prominent downstream signal of the extracellular nucleotide receptor P2RX7 in monocytic and osteoblastic cells. J Biol Chem 2010;285:34288-98.   DOI
11 Mi H, Muruganujan A, Casagrande JT, Thomas PD. Large-scale gene function analysis with the PANTHER classification system. Nat Protoc 2013;8:1551-66.   DOI
12 Glaab E, Baudot A, Krasnogor N, Schneider R, Valencia A. EnrichNet: network-based gene set enrichment analysis. Bioinformatics 2012;28:i451-7.   DOI
13 Loeffler I, Wolf G. Transforming growth factor-β and the progression of renal disease. Nephrol Dial Transplant 2014;29:i37-45.   DOI
14 Kaimal V, Bardes EE, Tabar SC, Jegga AG, Aronow BJ. ToppCluster: a multiple gene list feature analyzer for comparative enrichment clustering and network-based dissection of biological systems. Nucleic Acids Res 2010 J;38:W96-102.   DOI
15 Untergasser A, Cutcutache I, Koressaar T, Ye J, Faircloth BC, Remm M, et al. Primer3-new capabilities and interfaces. Nucleic Acids Res 2012;40:e115.   DOI
16 Webb H, McKeever S. 002 Influence of a renal biopsy result on treatment and management of steroid-resistant, idiopathic nephrotic syndrome in children. Arch Dis Child 2018;103(Suppl 2):A1.
17 Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci 2004;117(Pt 8):1281-3.   DOI
18 Galvan DL, Green NH, Danesh FR. The hallmarks of mitochondrial dysfunction in chronic kidney disease. Kidney Int 2017;92:1051-7.   DOI
19 Zhang H, Nair V, Saha J, Atkins KB, Hodgin JB, Saunders TL, et al. Podocyte-specific JAK2 overexpression worsens diabetic kidney disease in mice. Kidney Int 2017;92:909-21.   DOI
20 Ghayur A, Margetts PJ. Transforming growth factor-beta and the glomerular filtration barrier. Kidney Res Clin Pract 2013;32:3-10   DOI
21 Zhu L, Chen S, Chen Y. Unraveling the biological functions of Smad7 with mouse models. Cell Biosci 2011;1:44.   DOI
22 Lee D, Shenoy S, Nigatu Y, Plotkin M. Id proteins regulate capillary repair and perivascular cell proliferation following ischemia-reperfusion injury. PLoS One 2014;9:e88417.   DOI
23 Walton KL, Johnson KE, Harrison CA. Targeting TGF-β mediated SMAD signaling for the prevention of fibrosis. Front Pharmacol 2017;8:461.   DOI
24 Ostalska-Nowicka D, Smiech M, Jaroniec M, Zaorska K, Zawierucha P, Szaflarski W, et al. SOCS3 and SOCS5 mRNA expressions may predict initial steroid response in nephrotic syndrome children. Folia Histochem Cytobiol 2011;49:719-28.   DOI
25 Ortiz-Munoz G, Lopez-Parra V, Lopez-Franco O, Fernandez-Vizarra P, Mallavia B, Flores C, et al. Suppressors of cytokine signaling abrogate diabetic nephropathy. J Am Soc Nephrol 2010;21:763-72.   DOI
26 Holm Nielsen S, Guldager Kring Rasmussen D, Brix S, Fenton A, Jesky M, Ferro CJ, et al. A novel biomarker of laminin turnover is associated with disease progression and mortality in chronic kidney disease. PLoS One 2018;13:e0204239.   DOI
27 Qi H, Casalena G, Shi S, Yu L, Ebefors K, Sun Y, et al. Glomerular endothelial mitochondrial dysfunction is essential and characteristic of diabetic kidney disease susceptibility. Diabetes 2017;66:763-78.   DOI
28 R Development Core Team R. R: A Language and Environment for Statistical Computing. Team RDC, editor. R Foundation for Statistical Computing. R Foundation for Statistical Computing; 2011. p. 409. (R Foundation for Statistical Computing; vol. 1).
29 Rao X, Huang X, Zhou Z, Lin X. An improvement of the 2ˆ(-delta delta CT) method for quantitative real-time polymerase chain reaction data analysis. Biostat Bioinforma Biomath 2013:71-85.
30 Sinha A, Hari P, Sharma PK, Gulati A, Kalaivani M, Mantan M, et al. Disease course in steroid sensitive nephrotic syndrome. Indian Pediatr 2012;49:881-7.   DOI
31 Zhang B, Kirov S, Snoddy J. WebGestalt: an integrated system for exploring gene sets in various biological contexts. Nucleic Acids Res 2005;33:W741-8.   DOI
32 Lefebvre-Legendre L, Salin B, Schaeffer J, Brethes D, Dautant A, Ackerman SH, et al. Failure to assemble the α 3 β 3 subcomplex of the ATP synthase leads to accumulation of the α and β subunits within inclusion bodies and the loss of mitochondrial cristae in saccharomyces cerevisiae. J Biol Chem 2005;280:18386-92.   DOI
33 Warnes G, Bolker B, Bonebakker L, Gentleman R, Huber W, Liaw A, et al. Package 'gplots': Various R programming tools for plotting data. R package version. 2. 2016.
34 Hodgin JB, Borczuk AC, Nasr SH, Markowitz GS, Nair V, Martini S, et al. A molecular profile of focal segmental glomerulosclerosis from formalin-fixed, paraffin-embedded tissue. Am J Pathol 2010;177:1674-86.   DOI
35 Smoot ME, Ono K, Ruscheinski J, Wang PL, Ideker T. Cytoscape 2.8: new features for data integration and network visualization. Bioinformatics 2011;27:431-2.   DOI