Journal Home > Volume 1 , Issue 3
Background

Ankylosing spondylitis (AS) is a disabling chronic inflammatory disease. Mechanisms of ferroptosis in AS remain unclear. Using bioinformatics analysis, we aimed to identify key molecules involved in ferroptosis, provide potential therapeutic targets for AS, and further explore mechanisms of ferroptosis in AS.

Methods

GSE25101 was downloaded from the Gene Expression Omnibus and intersected with a ferroptosis gene dataset. The ferroptosis‐relate differentially‐expressed genes were further subjected to functional enrichment analysis, protein interaction network analysis, and gene‐miRNA interaction network analysis, from which potential key ferroptosis genes in the pathogenesis of ankylosing spondylitis were screened.

Results

A total of 20 differentially expressed genes were screened, most of which are involved in phosphoinositide 3 kinase‐Akt or mitogen‐activated protein kinase (MAPK) signaling pathways or the endoplasmic reticulum stress response. The following target genes were identified through protein‐protein interaction network analysis and screening of key modules constructed from genes associated with PI3K‐Akt and MAPK signaling pathways: TP53, PTEN, TLR4, HSPB1, DDIT3, and XBP1. In addition, PI3K‐Akt and MAPK signaling were associated with oxidative stress, which may play a role in AS pathological ossification related to ferroptosis. Only hsa‐miR‐205‐5p was found to target at least two genes by gene‐miRNA interaction network analysis.

Conclusions

Future therapeutic drug development may intervene by modulating MAPK or PI3K‐Akt signaling pathways rather than directly affecting the interleukin 17 pathway. hsa‐miR‐205‐5p may be a potential novel biomarker for AS.


menu
Abstract
Full text
Outline
About this article

Analysis of potential key ferroptosis genes in the pathogenesis of ankylosing spondylitis by bioinformatics

Show Author's information Guoxian He1,2Zexin Chen3 ( )Jiaxiao Li1,4Lanhui Zhang1,4Suling Liu1Yang Cui1,4( )
Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
Department of Rheumatology, The People's Hospital of Gaoming District of Foshan City, Foshan, China
Department of Rheumatology and Immunology, South China Hospital of Shenzhen University, Shenzhen, China
Southern Medical University, Guangzhou, China

Abstract

Background

Ankylosing spondylitis (AS) is a disabling chronic inflammatory disease. Mechanisms of ferroptosis in AS remain unclear. Using bioinformatics analysis, we aimed to identify key molecules involved in ferroptosis, provide potential therapeutic targets for AS, and further explore mechanisms of ferroptosis in AS.

Methods

GSE25101 was downloaded from the Gene Expression Omnibus and intersected with a ferroptosis gene dataset. The ferroptosis‐relate differentially‐expressed genes were further subjected to functional enrichment analysis, protein interaction network analysis, and gene‐miRNA interaction network analysis, from which potential key ferroptosis genes in the pathogenesis of ankylosing spondylitis were screened.

Results

A total of 20 differentially expressed genes were screened, most of which are involved in phosphoinositide 3 kinase‐Akt or mitogen‐activated protein kinase (MAPK) signaling pathways or the endoplasmic reticulum stress response. The following target genes were identified through protein‐protein interaction network analysis and screening of key modules constructed from genes associated with PI3K‐Akt and MAPK signaling pathways: TP53, PTEN, TLR4, HSPB1, DDIT3, and XBP1. In addition, PI3K‐Akt and MAPK signaling were associated with oxidative stress, which may play a role in AS pathological ossification related to ferroptosis. Only hsa‐miR‐205‐5p was found to target at least two genes by gene‐miRNA interaction network analysis.

Conclusions

Future therapeutic drug development may intervene by modulating MAPK or PI3K‐Akt signaling pathways rather than directly affecting the interleukin 17 pathway. hsa‐miR‐205‐5p may be a potential novel biomarker for AS.

Keywords: bioinformatics, ferroptosis, TLR4, ankylosing spondylitis, miR‐205‐5p, TP53

References(54)

[1]

Mauro D, Thomas R, Guggino G, Lories R, Brown MA, Ciccia F. Ankylosing spondylitis: an autoimmune or autoinflammatory disease? Nat Rev Rheumatol. 2021;17(7):387–404. https://doi.org/10.1038/s41584-021-00625-y

[2]

J Li, S Liu, Y Cui, Ray SD. Oxidative and antioxidative stress linked biomarkers in ankylosing spondylitis: a systematic review and meta‐analysis. Oxid Med Cell Longev. 2020;2020:47594511. https://doi.org/10.1155/2020/4759451

[3]

Saha A, Bagchi A, Chatterjee S, Dutta S, Misra S, Bhattacharjee D, et al. Phenotypic characterization of circulating endothelial cells induced by inflammation and oxidative stress in ankylosing spondylitis. Free Radic Res. 2021;55(5):520–32. https://doi.org/10.1080/10715762.2020.1870113

[4]

Solmaz D, Kozaci D, Sari I, Taylan A, Onen F, Akkoc N, et al. Oxidative stress and related factors in patients with ankylosing spondylitis. Eur J Rheumatol. 2016;3(1):20–4. https://doi.org/10.5152/eurjrheum.2015.0031

[5]

Biasi D, Carletto A, Caramaschi P, Bellavite P, Andrioli G, Caraffi M, et al. Neutrophil functions, spondylarthropathies and HLA‐B27: a study of 43 patients. Clin Exp Rheumatol. 1995;13(5):623–7.

[6]

Liu K, Wu L, Shi X, Wu F. Protective effect of naringin against ankylosing spondylitis via ossification, inflammation and oxidative stress in mice. Exp Ther Med. 2016;12(2):1153–8. https://doi.org/10.3892/etm.2016.3410

[7]

Ye G, Xie Z, Zeng H, Wang P, Li J, Zheng G, et al. Oxidative stress‐mediated mitochondrial dysfunction facilitates mesenchymal stem cell senescence in ankylosing spondylitis. Cell Death Dis. 2020;11(9):775. https://doi.org/10.1038/s41419-020-02993-x

[8]

Karakoc M, Altindag O, Keles H, Soran N, Selek S. Serum oxidative‐antioxidative status in patients with ankylosing spondilitis. Rheumatol Int. 2007;27(12):1131–4. https://doi.org/10.1007/s00296-007-0352-3

[9]

Feijoo M, Tunez I, Ruiz A, Tasset I, Munoz E, Collantes E. [Oxidative stress biomarkers as indicator of chronic inflammatory joint diseases stage]. Reumatol Clin. 2010;6(2):91–4. https://doi.org/10.1016/j.reuma.2008.12.016

[10]

Galli C, Passeri G, Macaluso GM. FoxOs, Wnts and oxidative stress‐induced bone loss: new players in the periodontitis arena? J Periodontal Res. 2011;46(4):397–406. https://doi.org/10.1111/j.1600-0765.2011.01354.x

[11]

Almeida M, Han L, Martin‐Millan M, O'Brien CA, Manolagas SC. Oxidative stress antagonizes Wnt signaling in osteoblast precursors by diverting beta‐catenin from T cell factor‐ to forkhead box O‐mediated transcription. J Biol Chem. 2007;282(37):27298–305. https://doi.org/10.1074/jbc.M702811200

[12]

Braem K, Luyten FP, Lories RJ. Blocking p38 signalling inhibits chondrogenesis in vitro but not ankylosis in a model of ankylosing spondylitis in vivo. Ann Rheum Dis. 2012;71(5):722–8. https://doi.org/10.1136/annrheumdis-2011-200377

[13]

Henrotin YE, Bruckner P, Pujol JP. The role of reactive oxygen species in homeostasis and degradation of cartilage. Osteoarthritis Cartilage. 2003;11(10):747–55. https://doi.org/10.1016/s1063-4584(03)00150-x

[14]

Wegner AM, Haudenschild DR. NADPH oxidases in bone and cartilage homeostasis and disease: a promising therapeutic target. J Orthop Res. 2020;38(10):2104–12. https://doi.org/10.1002/jor.24693

[15]

Ozgocmen S, Sogut S, Ardicoglu O, Fadillioglu E, Pekkutucu I, Akyol O. Serum nitric oxide, catalase, superoxide dismutase, and malondialdehyde status in patients with ankylosing spondylitis. Rheumatol Int. 2004;24(2):80–3. https://doi.org/10.1007/s00296-003-0335-y

[16]

Karkucak M, Capkin E, Alver A, Akyuz A, Kiris A, Ak E, et al. The effect of anti‐TNF agent on oxidation status in patients with ankylosing spondylitis. Clin Rheumatol. 2010;29(3):303–7. https://doi.org/10.1007/s10067-009-1325-6

[17]

Naziroglu M, Akkus S, Celik H. Levels of lipid peroxidation and antioxidant vitamins in plasma and erythrocytes of patients with ankylosing spondylitis. Clin Biochem. 2011;44(17‐18):1412–5. https://doi.org/10.1016/j.clinbiochem.2011.09.005

[18]

Ozgocmen S, Sogut S, Fadillioglu E, Ardicoglu A, Ardicoglu O. Antioxidant status and lipid peroxidation in seminal plasma and spermatozoa of patients with ankylosing spondylitis. Rheumatology. 2003;42(6):805–7. https://doi.org/10.1093/rheumatology/keg194

[19]

Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 2021;22(4):266–82. https://doi.org/10.1038/s41580-020-00324-8

[20]

Zhao T, Yang Q, Xi Y, Xie Z, Shen J, Li Z, et al. Ferroptosis in rheumatoid arthritis: a potential therapeutic strategy. Front Immunol. 2022;13:779585. https://doi.org/10.3389/fimmu.2022.779585

[21]

Xie Z, Hou H, Luo D, An R, Zhao Y, Qiu C. ROS‐dependent lipid peroxidation and reliant antioxidant ferroptosis‐suppressor‐protein 1 in rheumatoid arthritis: a covert clue for potential therapy. Inflammation. 2021;44(1):35–47. https://doi.org/10.1007/s10753-020-01338-2

[22]

Luo H, Zhang R. Icariin enhances cell survival in lipopolysaccharide‐induced synoviocytes by suppressing ferroptosis via the Xc‐/GPX4 axis. Exp Ther Med. 2021;21(1):72. https://doi.org/10.3892/etm.2020.9504

[23]

Wu J, Feng Z, Chen L, Li Y, Bian H, Geng J, et al. TNF antagonist sensitizes synovial fibroblasts to ferroptotic cell death in collagen‐induced arthritis mouse models. Nat Commun. 2022;13(1):676. https://doi.org/10.1038/s41467-021-27948-4

[24]

Jing X, Du T, Chen K, Guo J, Xiang W, Yao X, et al. Icariin protects against iron overload‐induced bone loss via suppressing oxidative stress. J Cell Physiol. 2019;234(7):10123–37. https://doi.org/10.1002/jcp.27678

[25]

Cen WJ, Feng Y, Li SS, Huang LW, Zhang T, Zhang W, et al. Iron overload induces G1 phase arrest and autophagy in murine preosteoblast cells. J Cell Physiol. 2018;233(9):6779–89. https://doi.org/10.1002/jcp.26405

[26]

Xiao W, Beibei F, Guangsi S, Yu J, Wen Z, Xi H, et al. Iron overload increases osteoclastogenesis and aggravates the effects of ovariectomy on bone mass. J Endocrinol. 2015;226(3):121–34. https://doi.org/10.1530/JOE-14-0657

[27]

Samakova A, Gazova A, Sabova N, Valaskova S, Jurikova M, Kyselovic J. The PI3k/Akt pathway is associated with angiogenesis, oxidative stress and survival of mesenchymal stem cells in pathophysiologic condition in ischemia. Physiol Res. 2019;68(Suppl 2):S131–8. https://doi.org/10.33549/physiolres.934345

[28]

Nguyen THP, Mahalakshmi B, Velmurugan BK. Functional role of ferroptosis on cancers, activation and deactivation by various therapeutic candidates‐an update. Chem Biol Interact. 2020;317:108930. https://doi.org/10.1016/j.cbi.2019.108930

[29]

Li J, Chen Z, Liao H, Zhong Y, Hua J, Su M, et al. Anti‐osteogenic effect of Danshensu in ankylosing spondylitis: an in vitro study based on integrated network pharmacology. Front Pharmacol. 2021;12:772190. https://doi.org/10.3389/fphar.2021.772190

[30]

Zhang J, Wang X, Vikash V, Ye Q, Wu D, Liu Y, et al. ROS and ROS‐mediated cellular signaling. Oxid Med Cell Longev. 2016;2016:4350965. https://doi.org/10.1155/2016/4350965

[31]

Huang M, Xu S, Liu L, Zhang M, Guo J, Yuan Y, et al. m6A methylation regulates osteoblastic differentiation and bone remodeling. Front Cell Dev Biol. 2021;9:783322. https://doi.org/10.3389/fcell.2021.783322

[32]

Kang R, Kroemer G, Tang D. The tumor suppressor protein p53 and the ferroptosis network. Free Radic Biol Med. 2019;133:162–8. https://doi.org/10.1016/j.freeradbiomed.2018.05.074

[33]

Taghadosi M, Adib M, Jamshidi A, Mahmoudi M, Farhadi E. The p53 status in rheumatoid arthritis with focus on fibroblast‐like synoviocytes. Immunol Res. 2021;69(3):225–38. https://doi.org/10.1007/s12026-021-09202-7

[34]

Li W, Feng G, Gauthier JM, Lokshina I, Higashikubo R, Evans S, et al. Ferroptotic cell death and TLR4/Trif signaling initiate neutrophil recruitment after heart transplantation. J Clin Invest. 2019;129(6):2293–304. https://doi.org/10.1172/JCI126428

[35]

Zhu K, Zhu X, Sun S, Yang W, Liu S, Tang Z, et al. Inhibition of TLR4 prevents hippocampal hypoxic‐ischemic injury by regulating ferroptosis in neonatal rats. Exp Neurol. 2021;345:113828. https://doi.org/10.1016/j.expneurol.2021.113828

[36]

Gan T, Yang Y, Hu F, Chen X, Zhou J, Li Y, et al. TLR3 regulated poly I:C‐induced neutrophil extracellular traps and acute lung injury partly through p38 MAP kinase. Front Microbiol. 2018;9:3174. https://doi.org/10.3389/fmicb.2018.03174

[37]

He W, Qu T, Yu Q, Wang Z, Lv H, Zhang J, et al. LPS induces IL‐8 expression through TLR4, MyD88, NF‐kappaB and MAPK pathways in human dental pulp stem cells. Int Endod J. 2013;46(2):128–36. https://doi.org/10.1111/j.1365-2591.2012.02096.x

[38]

Li Y, Huang L, Cai Z, Deng W, Wang P, Su H, et al. A study of the immunoregulatory function of TLR3 and TLR4 on mesenchymal stem cells in ankylosing spondylitis. Stem Cell Dev. 2019;28(20):1398–412. https://doi.org/10.1089/scd.2019.0039

[39]

Zhang J, Xu R, Wu L, Jiang J. Expression and function of Toll‐like receptors in peripheral blood mononuclear cells in patients with ankylosing spondylitis. Mol Med Rep. 2019;20(4):3565–72. https://doi.org/10.3892/mmr.2019.10631

[40]

Zhang L, Wang F, Li D, Yan Y, Wang H. Transferrin receptor‐mediated reactive oxygen species promotes ferroptosis of KGN cells via regulating NADPH oxidase 1/PTEN induced kinase 1/acyl‐CoA synthetase long chain family member 4 signaling. Bioengineered. 2021;12(1):4983–94. https://doi.org/10.1080/21655979.2021.1956403

[41]

Li XF, Chen X, Bao J, Xu L, Zhang L, Huang C, et al. PTEN negatively regulates the expression of pro‐inflammatory cytokines and chemokines of fibroblast‐like synoviocytes in adjuvant‐induced arthritis. Artif Cells, Nanomed Biotechnol. 2019;47(1):3687–96. https://doi.org/10.1080/21691401.2019.1661849

[42]

Wang CR, Shiau AL, Chen SY, Lin LL, Tai MH, Shieh GS, et al. Amelioration of collagen‐induced arthritis in rats by adenovirus‐mediated PTEN gene transfer. Arthritis Rheum. 2008;58(6):1650–6. https://doi.org/10.1002/art.23517

[43]

Sun X, Ou Z, Xie M, Kang R, Fan Y, Niu X, et al. HSPB1 as a novel regulator of ferroptotic cancer cell death. Oncogene. 2015;34(45):5617–25. https://doi.org/10.1038/onc.2015.32

[44]

Zauner D, Quehenberger F, Hermann J, Dejaco C, Stradner MH, Stojakovic T, et al. Whole body hyperthermia treatment increases interleukin 10 and toll‐like receptor 4 expression in patients with ankylosing spondylitis: a pilot study. Int J Hyperther. 2014;30(6):393–401. https://doi.org/10.3109/02656736.2014.956810

[45]

Huang R, Hui Z, Wei S, Li D, Li W, Daping W, et al. IRE1 signaling regulates chondrocyte apoptosis and death fate in the osteoarthritis. J Cell Physiol. 2022;237(1):118–27. https://doi.org/10.1002/jcp.30537

[46]

Dickie LJ, Aziz AM, Savic S, Lucherini OM, Cantarini L, Geiler J, et al. Involvement of X‐box binding protein 1 and reactive oxygen species pathways in the pathogenesis of tumour necrosis factor receptor‐associated periodic syndrome. Ann Rheum Dis. 2012;71(12):2035–43. https://doi.org/10.1136/annrheumdis-2011-201197

[47]

Hu F, Tang Y, Wang P, Bai M, Li X, Song J, et al. Endoplasmic reticulum stress perpetuated toll‐like receptor signalling‐mediated inflammation in rheumatoid arthritis via X‐box‐binding protein‐1. Clin Exp Rheumatol. 2021;39(4):859–67. https://doi.org/10.55563/clinexprheumatol/a7ehz7

[48]

Song AF, Kang L, Wang YF, Wang M. MiR‐34a‐5p inhibits fibroblast‐like synoviocytes proliferation via XBP1. Eur Rev Med Pharmacol Sci. 2020;24(22):11675–82. https://doi.org/10.26355/eurrev_202011_23812

[49]

Yu X, Xu X, Dong W, Yang C, Luo Y, He Y, et al. DDIT3/CHOP mediates the inhibitory effect of ER stress on chondrocyte differentiation by AMPKalpha‐SIRT1 pathway. Biochim Biophys Acta Mol Cell Res. 2022;1869(8):119265. https://doi.org/10.1016/j.bbamcr.2022.119265

[50]

Muratsu‐Ikeda S, Nangaku M, Ikeda Y, Tanaka T, Wada T, Inagi R. Downregulation of miR‐205 modulates cell susceptibility to oxidative and endoplasmic reticulum stresses in renal tubular cells. PLoS One. 2012;7(7):e41462. https://doi.org/10.1371/journal.pone.0041462

[51]

Magenta A, Dellambra E, Ciarapica R, Capogrossi MC. Oxidative stress, microRNAs and cytosolic calcium homeostasis. Cell Calcium. 2016;60(3):207–17. https://doi.org/10.1016/j.ceca.2016.04.002

[52]

Chen S, Paveley R, Kraal L, Sritharan L, Stevens E, Dedi N, et al. Selective targeting of PI3Kdelta suppresses human IL‐17‐producing T cells and innate‐like lymphocytes and may be therapeutic for IL‐17‐mediated diseases. J Autoimmun. 2020;111:102435. https://doi.org/10.1016/j.jaut.2020.102435

[53]

Lories RJ, Derese I, Luyten FP, de Vlam K. Activation of nuclear factor kappa B and mitogen activated protein kinases in psoriatic arthritis before and after etanercept treatment. Clin Exp Rheumatol. 2008;26(1):96–102.

[54]

Fiechter RH, de Jong HM, van Mens LJJ, Fluri IA, Tas SW, Baeten DLP, et al. IL‐12p40/IL‐23p40 blockade with ustekinumab decreases the synovial inflammatory infiltrate through modulation of multiple signaling pathways including MAPK‐ERK and Wnt. Front Immunol. 2021;12:611656. https://doi.org/10.3389/fimmu.2021.611656

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 26 March 2023
Accepted: 23 July 2023
Published: 14 September 2023
Issue date: September 2023

Copyright

© 2023 The Authors. Tsinghua University Press.

Acknowledgements

ACKNOWLEDGMENTS

We thank reviewers for the comments on the content of the article, and we would like to thank all participants who participated in the study for their time and involvement.

Rights and permissions

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

Return