Skip to main content
Top
Published in: Arthritis Research & Therapy 1/2020

01-12-2020 | Systemic Lupus Erythematosus | Research article

An integrative Bayesian network approach to highlight key drivers in systemic lupus erythematosus

Authors: Samaneh Maleknia, Zahra Salehi, Vahid Rezaei Tabar, Ali Sharifi-Zarchi, Kaveh Kavousi

Published in: Arthritis Research & Therapy | Issue 1/2020

Login to get access

Abstract

Background

A comprehensive intuition of the systemic lupus erythematosus (SLE), as a complex and multifactorial disease, is a biological challenge. Dealing with this challenge needs employing sophisticated bioinformatics algorithms to discover the unknown aspects. This study aimed to underscore key molecular characteristics of SLE pathogenesis, which may serve as effective targets for therapeutic intervention.

Methods

In the present study, the human peripheral blood mononuclear cell (PBMC) microarray datasets (n = 6), generated by three platforms, which included SLE patients (n = 220) and healthy control samples (n = 135) were collected. Across each platform, we integrated the datasets by cross-platform normalization (CPN). Subsequently, through BNrich method, the structures of Bayesian networks (BNs) were extracted from KEGG-indexed SLE, TCR, and BCR signaling pathways; the values of the node (gene) and edge (intergenic relationships) parameters were estimated within each integrated datasets. Parameters with the FDR < 0.05 were considered significant. Finally, a mixture model was performed to decipher the signaling pathway alterations in the SLE patients compared to healthy controls.

Results

In the SLE signaling pathway, we identified the dysregulation of several nodes involved in the (1) clearance mechanism (SSB, MACROH2A2, TRIM21, H2AX, and C1Q gene family), (2) autoantigen presentation by MHCII (HLA gene family, CD80, IL10, TNF, and CD86), and (3) end-organ damage (FCGR1A, ELANE, and FCGR2A). As a remarkable finding, we demonstrated significant perturbation in CD80 and CD86 to CD28, CD40LG to CD40, C1QA and C1R to C2, and C1S to C4A edges. Moreover, we not only replicated previous studies regarding alterations of subnetworks involved in TCR and BCR signaling pathways (PI3K/AKT, MAPK, VAV gene family, AP-1 transcription factor) but also distinguished several significant edges between genes (PPP3 to NFATC gene families). Our findings unprecedentedly showed that different parameter values assign to the same node based on the pathway topology (the PIK3CB parameter values were 1.7 in TCR vs − 0.5 in BCR signaling pathway).

Conclusions

Applying the BNrich as a hybridized network construction method, we highlight under-appreciated systemic alterations of SLE, TCR, and BCR signaling pathways in SLE. Consequently, having such a systems biology approach opens new insights into the context of multifactorial disorders.
Appendix
Available only for authorised users
Literature
1.
go back to reference Yang F, Zhai Z, Luo X, Luo G, Zhuang L, Zhang Y, Li Y, Sun E, He Y. Bioinformatics identification of key candidate genes and pathways associated with systemic lupus erythematosus. Clin Rheumatol. 2019;39:425–34. Yang F, Zhai Z, Luo X, Luo G, Zhuang L, Zhang Y, Li Y, Sun E, He Y. Bioinformatics identification of key candidate genes and pathways associated with systemic lupus erythematosus. Clin Rheumatol. 2019;39:425–34.
2.
go back to reference Javinani A, Ashraf-Ganjouei A, Aslani S, Jamshidi A, Mahmoudi M. Exploring the etiopathogenesis of systemic lupus erythematosus: a genetic perspective. Immunogenetics. 2019;71:283–97.CrossRef Javinani A, Ashraf-Ganjouei A, Aslani S, Jamshidi A, Mahmoudi M. Exploring the etiopathogenesis of systemic lupus erythematosus: a genetic perspective. Immunogenetics. 2019;71:283–97.CrossRef
3.
go back to reference Helmick CG, Felson DT, Lawrence RC, Gabriel S, Hirsch R, Kwoh CK, Liang MH, Kremers HM, Mayes MD, Merkel PA, et al. Estimates of the prevalence of arthritis and other rheumatic conditions in the United States. Part I. Arthritis Rheum. 2008;58:15–25.CrossRef Helmick CG, Felson DT, Lawrence RC, Gabriel S, Hirsch R, Kwoh CK, Liang MH, Kremers HM, Mayes MD, Merkel PA, et al. Estimates of the prevalence of arthritis and other rheumatic conditions in the United States. Part I. Arthritis Rheum. 2008;58:15–25.CrossRef
4.
go back to reference Garris C, Shah M, Farrelly E. The prevalence and burden of systemic lupus erythematosus in a medicare population: retrospective analysis of medicare claims. Cost Eff Resour Alloc. 2015;13. Garris C, Shah M, Farrelly E. The prevalence and burden of systemic lupus erythematosus in a medicare population: retrospective analysis of medicare claims. Cost Eff Resour Alloc. 2015;13.
5.
go back to reference Meas R, Burak MJ, Sweasy JB. DNA repair and systemic lupus erythematosus. DNA Repair (Amst). 2017;56:174–82.CrossRef Meas R, Burak MJ, Sweasy JB. DNA repair and systemic lupus erythematosus. DNA Repair (Amst). 2017;56:174–82.CrossRef
6.
go back to reference Zharkova O, Celhar T, Cravens PD, Satterthwaite AB, Fairhurst AM, Davis LS. Pathways leading to an immunological disease: systemic lupus erythematosus. Rheumatology (Oxford). 2017;56:i55–66.CrossRef Zharkova O, Celhar T, Cravens PD, Satterthwaite AB, Fairhurst AM, Davis LS. Pathways leading to an immunological disease: systemic lupus erythematosus. Rheumatology (Oxford). 2017;56:i55–66.CrossRef
7.
go back to reference Tsokos GC, Lo MS, Reis PC, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol. 2016;12:716–30.CrossRef Tsokos GC, Lo MS, Reis PC, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol. 2016;12:716–30.CrossRef
8.
go back to reference Wardemann H, Yurasov S, Schaefer A, Young JW, Meffre E, Nussenzweig MC. Predominant autoantibody production by early human B cell precursors. Science (80-. ). 2003;301:1374–7.CrossRef Wardemann H, Yurasov S, Schaefer A, Young JW, Meffre E, Nussenzweig MC. Predominant autoantibody production by early human B cell precursors. Science (80-. ). 2003;301:1374–7.CrossRef
9.
go back to reference Swanson CL, Wilson TJ, Strauch P, Colonna M, Pelanda R, Torres RM. Type I IFN enhances follicular B cell contribution to the T cell-independent antibody response. J Exp Med. 2010;207:1485–500.CrossRef Swanson CL, Wilson TJ, Strauch P, Colonna M, Pelanda R, Torres RM. Type I IFN enhances follicular B cell contribution to the T cell-independent antibody response. J Exp Med. 2010;207:1485–500.CrossRef
10.
go back to reference Xu HC, Grusdat M, Pandyra AA, Polz R, Huang J, Sharma P, Deenen R, Köhrer K, Rahbar R, Diefenbach A, et al. Type I interferon protects antiviral CD8+ T cells from NK cell cytotoxicity. Immunity. 2014;40:949–60.CrossRef Xu HC, Grusdat M, Pandyra AA, Polz R, Huang J, Sharma P, Deenen R, Köhrer K, Rahbar R, Diefenbach A, et al. Type I interferon protects antiviral CD8+ T cells from NK cell cytotoxicity. Immunity. 2014;40:949–60.CrossRef
11.
go back to reference Katsuyama T, Tsokos GC, Moulton VR. Aberrant T cell signaling and subsets in systemic lupus erythematosus. Front Immunol. 2018;9:1–15. Katsuyama T, Tsokos GC, Moulton VR. Aberrant T cell signaling and subsets in systemic lupus erythematosus. Front Immunol. 2018;9:1–15.
12.
go back to reference Li Y, Higgs RE, Hoffman RW, Dow ER, Liu X, Petri M, Wallace DJ, Dörner T, Eastwood BJ, Miller BB, et al. A Bayesian gene network reveals insight into the JAK-STAT pathway in systemic lupus erythematosus. PLoS One. 2019;14:1-19. Li Y, Higgs RE, Hoffman RW, Dow ER, Liu X, Petri M, Wallace DJ, Dörner T, Eastwood BJ, Miller BB, et al. A Bayesian gene network reveals insight into the JAK-STAT pathway in systemic lupus erythematosus. PLoS One. 2019;14:1-19.
13.
go back to reference Yu J, Smith VA, Wang PP, Hartemink AJ, Jarvis ED. Advances to Bayesian network inference for generating causal networks from observational biological data. Bioinformatics. 2004;20:3594–603.CrossRef Yu J, Smith VA, Wang PP, Hartemink AJ, Jarvis ED. Advances to Bayesian network inference for generating causal networks from observational biological data. Bioinformatics. 2004;20:3594–603.CrossRef
14.
go back to reference Gendelman R, Xing H, Mirzoeva OK, Sarde P, Curtis C, Feiler HS, McDonagh P, Gray JW, Khalil I, Korn WM. Bayesian network inference modeling identifies TRIB1 as a novel regulator of cell-cycle progression and survival in cancer cells. Cancer Res. 2017;77:1575–85.CrossRef Gendelman R, Xing H, Mirzoeva OK, Sarde P, Curtis C, Feiler HS, McDonagh P, Gray JW, Khalil I, Korn WM. Bayesian network inference modeling identifies TRIB1 as a novel regulator of cell-cycle progression and survival in cancer cells. Cancer Res. 2017;77:1575–85.CrossRef
15.
go back to reference Luo, Y.; El Naqa, I.; McShan, D.L.; Ray, D.; Lohse, I.; Matuszak, M.M.; Owen, D.; Jolly, S.; Lawrence, T.S.; Kong, F.-M. (Spring); et al. Unraveling biophysical interactions of radiation pneumonitis in non-small-cell lung cancer via Bayesian network analysis. Radiother.Oncol. 2017, 123, 85–92. Luo, Y.; El Naqa, I.; McShan, D.L.; Ray, D.; Lohse, I.; Matuszak, M.M.; Owen, D.; Jolly, S.; Lawrence, T.S.; Kong, F.-M. (Spring); et al. Unraveling biophysical interactions of radiation pneumonitis in non-small-cell lung cancer via Bayesian network analysis. Radiother.Oncol. 2017, 123, 85–92.
16.
go back to reference Agrahari R, Foroushani A, Docking TR, Chang L, Duns G, Hudoba M, Karsan A, Zare H. Applications of Bayesian network models in predicting types of hematological malignancies. Sci Rep. 2018;8:6951.CrossRef Agrahari R, Foroushani A, Docking TR, Chang L, Duns G, Hudoba M, Karsan A, Zare H. Applications of Bayesian network models in predicting types of hematological malignancies. Sci Rep. 2018;8:6951.CrossRef
17.
go back to reference Ramos J, Das J, Felty Q, Yoo C, Poppiti R, Murrell D, Foster PJ, Roy D. NRF1 motif sequence-enriched genes involved in ER/PR −ve HER2 +ve breast cancer signaling pathways. Breast Cancer Res Treat. 2018;172:469–85.CrossRef Ramos J, Das J, Felty Q, Yoo C, Poppiti R, Murrell D, Foster PJ, Roy D. NRF1 motif sequence-enriched genes involved in ER/PR −ve HER2 +ve breast cancer signaling pathways. Breast Cancer Res Treat. 2018;172:469–85.CrossRef
18.
go back to reference Maleknia, S.; Sharifi-Zarchi, A.; Tabar, V.R.; Namazi, M.; Kavousi, K. BNrich: a Bayesian network approach to the pathway enrichment analysis. bioRxiv 2020, 2020.01.13.905448. Maleknia, S.; Sharifi-Zarchi, A.; Tabar, V.R.; Namazi, M.; Kavousi, K. BNrich: a Bayesian network approach to the pathway enrichment analysis. bioRxiv 2020, 2020.01.13.905448.
19.
go back to reference Hamid JS, Hu P, Roslin NM, Ling V, Greenwood CMT, Beyene J. Data integration in genetics and genomics: methods and challenges. Hum Genomics Proteomics. 2009;1:1-13. Hamid JS, Hu P, Roslin NM, Ling V, Greenwood CMT, Beyene J. Data integration in genetics and genomics: methods and challenges. Hum Genomics Proteomics. 2009;1:1-13.
20.
go back to reference Larsen MJ, Thomassen M, Tan Q, Sørensen KP, Kruse TA. Microarray-based RNA profiling of breast cancer: batch effect removal improves cross-platform consistency. Biomed Res Int. 2014;2014:1-11. Larsen MJ, Thomassen M, Tan Q, Sørensen KP, Kruse TA. Microarray-based RNA profiling of breast cancer: batch effect removal improves cross-platform consistency. Biomed Res Int. 2014;2014:1-11.
21.
go back to reference Irigoyen A, Jimenez-Luna C, Benavides M, Caba O, Gallego J, Ortuño FM, Guillen-Ponce C, Rojas I, Aranda E, Torres C, et al. Integrative multi-platform meta-analysis of gene expression profiles in pancreatic ductal adenocarcinoma patients for identifying novel diagnostic biomarkers. PLoS One. 2018;13:1-16. Irigoyen A, Jimenez-Luna C, Benavides M, Caba O, Gallego J, Ortuño FM, Guillen-Ponce C, Rojas I, Aranda E, Torres C, et al. Integrative multi-platform meta-analysis of gene expression profiles in pancreatic ductal adenocarcinoma patients for identifying novel diagnostic biomarkers. PLoS One. 2018;13:1-16.
22.
go back to reference Taminau J, Lazar C, Meganck S, Nowé A. Comparison of merging and meta-analysis as alternative approaches for integrative gene expression analysis. ISRN Bioinforma. 2014;2014:1–7.CrossRef Taminau J, Lazar C, Meganck S, Nowé A. Comparison of merging and meta-analysis as alternative approaches for integrative gene expression analysis. ISRN Bioinforma. 2014;2014:1–7.CrossRef
23.
go back to reference Walsh C, Hu P, Batt J, Santos C. Microarray meta-analysis and cross-platform normalization: integrative genomics for robust biomarker discovery. Microarrays. 2015;4:389–406.CrossRef Walsh C, Hu P, Batt J, Santos C. Microarray meta-analysis and cross-platform normalization: integrative genomics for robust biomarker discovery. Microarrays. 2015;4:389–406.CrossRef
24.
go back to reference Zhang X, Yu D, Zou G, Liang H. Optimal model averaging estimation for generalized linear models and generalized linear mixed-effects models. J Am Stat Assoc. 2016;111:1775–90.CrossRef Zhang X, Yu D, Zou G, Liang H. Optimal model averaging estimation for generalized linear models and generalized linear mixed-effects models. J Am Stat Assoc. 2016;111:1775–90.CrossRef
25.
go back to reference Lee HM, Sugino H, Aoki C, Nishimoto N. Underexpression of mitochondrial-DNA encoded ATP synthesis-related genes and DNA repair genes in systemic lupus erythematosus. Arthritis Res. Ther. 2011;13:R63.CrossRef Lee HM, Sugino H, Aoki C, Nishimoto N. Underexpression of mitochondrial-DNA encoded ATP synthesis-related genes and DNA repair genes in systemic lupus erythematosus. Arthritis Res. Ther. 2011;13:R63.CrossRef
26.
go back to reference Lee HM, Mima T, Sugino H, Aoki C, Adachi Y, Yoshio-Hoshino N, Matsubara K, Nishimoto N. Interactions among type I and type II interferon, tumor necrosis factor, and β-estradiol in the regulation of immune response-related gene expressions in systemic lupus erythematosus. Arthritis Res Ther. 2009;11:R1.CrossRef Lee HM, Mima T, Sugino H, Aoki C, Adachi Y, Yoshio-Hoshino N, Matsubara K, Nishimoto N. Interactions among type I and type II interferon, tumor necrosis factor, and β-estradiol in the regulation of immune response-related gene expressions in systemic lupus erythematosus. Arthritis Res Ther. 2009;11:R1.CrossRef
27.
go back to reference Kennedy WP, Maciuca R, Wolslegel K, Tew W, Abbas AR, Chaivorapol C, Morimoto A, McBride JM, Brunetta P, Richardson BC, et al. Association of the interferon signature metric with serological disease manifestations but not global activity scores in multiple cohorts of patients with SLE. Lupus Sci Med. 2015;2:1-11. Kennedy WP, Maciuca R, Wolslegel K, Tew W, Abbas AR, Chaivorapol C, Morimoto A, McBride JM, Brunetta P, Richardson BC, et al. Association of the interferon signature metric with serological disease manifestations but not global activity scores in multiple cohorts of patients with SLE. Lupus Sci Med. 2015;2:1-11.
29.
go back to reference Zollars E, Courtney SM, Wolf BJ, Allaire N, Ranger A, Hardiman G, Petri M. Clinical application of a modular genomics technique in systemic lupus erythematosus: Progress towards precision medicine. Int J Genomics. 2016;2016:1-7. Zollars E, Courtney SM, Wolf BJ, Allaire N, Ranger A, Hardiman G, Petri M. Clinical application of a modular genomics technique in systemic lupus erythematosus: Progress towards precision medicine. Int J Genomics. 2016;2016:1-7.
30.
go back to reference Toro-Domínguez D, Martorell-Marugán J, Goldman D, Petri M, Carmona-Sáez P, Alarcón-Riquelme ME. Stratification of systemic lupus erythematosus patients into three groups of disease activity progression according to longitudinal gene expression. Arthritis Rheumatol. 2018;70:2025–35.CrossRef Toro-Domínguez D, Martorell-Marugán J, Goldman D, Petri M, Carmona-Sáez P, Alarcón-Riquelme ME. Stratification of systemic lupus erythematosus patients into three groups of disease activity progression according to longitudinal gene expression. Arthritis Rheumatol. 2018;70:2025–35.CrossRef
31.
go back to reference Petri M, Fu W, Ranger A, Allaire N, Cullen P, Magder LS, Zhang Y. Association between changes in gene signatures expression and disease activity among patients with systemic lupus erythematosus. BMC Med Genet. 2019;12:1-19. Petri M, Fu W, Ranger A, Allaire N, Cullen P, Magder LS, Zhang Y. Association between changes in gene signatures expression and disease activity among patients with systemic lupus erythematosus. BMC Med Genet. 2019;12:1-19.
32.
go back to reference Jiang SH, Athanasopoulos V, Ellyard JI, Chuah A, Cappello J, Cook A, Prabhu SB, Cardenas J, Gu J, Stanley M, et al. Functional rare and low frequency variants in BLK and BANK1 contribute to human lupus. Nat Commun. 2019;10:1-12. Jiang SH, Athanasopoulos V, Ellyard JI, Chuah A, Cappello J, Cook A, Prabhu SB, Cardenas J, Gu J, Stanley M, et al. Functional rare and low frequency variants in BLK and BANK1 contribute to human lupus. Nat Commun. 2019;10:1-12.
33.
go back to reference Ritchie ME, Phipson B, Wu DI, Hu Y, Law CW, Shi W, Smyth GK. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.CrossRef Ritchie ME, Phipson B, Wu DI, Hu Y, Law CW, Shi W, Smyth GK. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.CrossRef
34.
go back to reference Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The SVA package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28:882–3.CrossRef Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The SVA package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28:882–3.CrossRef
35.
go back to reference Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45:D353–61.CrossRef Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45:D353–61.CrossRef
36.
go back to reference Andrade JM, Estévez-Pérez MG. Statistical comparison of the slopes of two regression lines: a tutorial. Anal Chim Acta. 2014;838:1–12.CrossRef Andrade JM, Estévez-Pérez MG. Statistical comparison of the slopes of two regression lines: a tutorial. Anal Chim Acta. 2014;838:1–12.CrossRef
37.
go back to reference Wasserman, L. All of statistics: a concise course in statistical inference. In The American Statistician; 2004; Vol. 59, pp. 161–163&216–217 ISBN 0387402721. Wasserman, L. All of statistics: a concise course in statistical inference. In The American Statistician; 2004; Vol. 59, pp. 161–163&216–217 ISBN 0387402721.
38.
go back to reference Wang J, Taaffe MR. Multivariate mixtures of normal distributions: properties, random vector generation, fitting, and as models of market daily changes. INFORMS J Comput. 2015;27:193–203.CrossRef Wang J, Taaffe MR. Multivariate mixtures of normal distributions: properties, random vector generation, fitting, and as models of market daily changes. INFORMS J Comput. 2015;27:193–203.CrossRef
39.
go back to reference Asadipour M, Hassan-Zadeh V, Aryaeian N, Shahram F, Mahmoudi M. Histone variants expression in peripheral blood mononuclear cells of patients with rheumatoid arthritis. Int J Rheum Dis. 2018;21:1831–7.CrossRef Asadipour M, Hassan-Zadeh V, Aryaeian N, Shahram F, Mahmoudi M. Histone variants expression in peripheral blood mononuclear cells of patients with rheumatoid arthritis. Int J Rheum Dis. 2018;21:1831–7.CrossRef
40.
go back to reference Hung T, Pratt GA, Sundararaman B, Townsend MJ, Chaivorapol C, Bhangale T, Graham RR, Ortmann W, Criswell LA, Yeo GW, et al. The Ro60 autoantigen binds endogenous retroelements and regulates inflammatory gene expression. Science (80-. ). 2015;350:455–9.CrossRef Hung T, Pratt GA, Sundararaman B, Townsend MJ, Chaivorapol C, Bhangale T, Graham RR, Ortmann W, Criswell LA, Yeo GW, et al. The Ro60 autoantigen binds endogenous retroelements and regulates inflammatory gene expression. Science (80-. ). 2015;350:455–9.CrossRef
41.
go back to reference Kamiyama R, Yoshimi R, Takeno M, Iribe Y, Tsukahara T, Kishimoto D, Kunishita Y, Sugiyama Y, Tsuchida N, Nakano H, et al. Dysfunction of TRIM21 in interferon signature of systemic lupus erythematosus. Mod Rheumatol. 2018;28:993–1003.CrossRef Kamiyama R, Yoshimi R, Takeno M, Iribe Y, Tsukahara T, Kishimoto D, Kunishita Y, Sugiyama Y, Tsuchida N, Nakano H, et al. Dysfunction of TRIM21 in interferon signature of systemic lupus erythematosus. Mod Rheumatol. 2018;28:993–1003.CrossRef
42.
go back to reference Espinosa A, Dardalhon V, Brauner S, Ambrosi A, Higgs R, Quintana FJ, Sjöstrand M, Eloranta ML, Gabhann JN, Winqvist O, et al. Loss of the lupus autoantigen Ro52/Trim21 induces tissue inflammation and systemic autoimmunity by disregulating the IL-23-Th17 pathway. J Exp Med. 2009;206:1661–71.CrossRef Espinosa A, Dardalhon V, Brauner S, Ambrosi A, Higgs R, Quintana FJ, Sjöstrand M, Eloranta ML, Gabhann JN, Winqvist O, et al. Loss of the lupus autoantigen Ro52/Trim21 induces tissue inflammation and systemic autoimmunity by disregulating the IL-23-Th17 pathway. J Exp Med. 2009;206:1661–71.CrossRef
43.
go back to reference Shimada-Sugimoto M, Otowa T, Miyagawa T, Khor SS, Kashiwase K, Sugaya N, Kawamura Y, Umekage T, Kojima H, Saji H, et al. Immune-related pathways including HLA-DRB1*13:02 are associated with panic disorder. Brain Behav Immun. 2015;46:96–103.CrossRef Shimada-Sugimoto M, Otowa T, Miyagawa T, Khor SS, Kashiwase K, Sugaya N, Kawamura Y, Umekage T, Kojima H, Saji H, et al. Immune-related pathways including HLA-DRB1*13:02 are associated with panic disorder. Brain Behav Immun. 2015;46:96–103.CrossRef
44.
go back to reference Kunishita Y, Yoshimi R, Kamiyama R, Kishimoto D, Yoshida K, Hashimoto E, Komiya T, Sakurai N, Sugiyama Y, Kirino Y, et al. TRIM21 dysfunction enhances aberrant B-cell differentiation in autoimmune pathogenesis. Front Immunol. 2020;11. Kunishita Y, Yoshimi R, Kamiyama R, Kishimoto D, Yoshida K, Hashimoto E, Komiya T, Sakurai N, Sugiyama Y, Kirino Y, et al. TRIM21 dysfunction enhances aberrant B-cell differentiation in autoimmune pathogenesis. Front Immunol. 2020;11.
45.
go back to reference Hachicha, H.; Kammoun, A.; Mahfoudh, N.; Marzouk, S.; Feki, S.; Fakhfakh, R.; Fourati, H.; Haddouk, S.; Frikha, F.; Gaddour, L.; et al. Human leukocyte antigens-DRB1*03 is associated with systemic lupus erythematosus and anti-SSB production in South Tunisia. Int. J. Health Sci. (Qassim). 12, 21–27. Hachicha, H.; Kammoun, A.; Mahfoudh, N.; Marzouk, S.; Feki, S.; Fakhfakh, R.; Fourati, H.; Haddouk, S.; Frikha, F.; Gaddour, L.; et al. Human leukocyte antigens-DRB1*03 is associated with systemic lupus erythematosus and anti-SSB production in South Tunisia. Int. J. Health Sci. (Qassim). 12, 21–27.
46.
go back to reference De Leeuw, K.; Kallenberg, C.G.M. Antibodies against c1q; 2018; ISBN 9780323479271. De Leeuw, K.; Kallenberg, C.G.M. Antibodies against c1q; 2018; ISBN 9780323479271.
47.
go back to reference Quiroz EN, Chavez-Estrada V, Macias-Ochoa K, Ayala-Navarro MF, Flores-Aguilar AS, Morales-Navarrete F, Lopez F, de la C, Escorcia LG, Musso CG, Martinez GA, et al. Epigenetic mechanisms and posttranslational modifications in systemic lupus erythematosus. Int. J Mol Sci. 2019;20:1-20. Quiroz EN, Chavez-Estrada V, Macias-Ochoa K, Ayala-Navarro MF, Flores-Aguilar AS, Morales-Navarrete F, Lopez F, de la C, Escorcia LG, Musso CG, Martinez GA, et al. Epigenetic mechanisms and posttranslational modifications in systemic lupus erythematosus. Int. J Mol Sci. 2019;20:1-20.
48.
go back to reference Webber D, Cao J, Dominguez D, Gladman DD, Levy DM, Ng L, Paterson AD, Touma Z, Urowitz MB, Wither JE, et al. Association of systemic lupus erythematosus (SLE) genetic susceptibility loci with lupus nephritis in childhood-onset and adult-onset SLE. Rheumatol. (United Kingdom). 2020;59:90–8. Webber D, Cao J, Dominguez D, Gladman DD, Levy DM, Ng L, Paterson AD, Touma Z, Urowitz MB, Wither JE, et al. Association of systemic lupus erythematosus (SLE) genetic susceptibility loci with lupus nephritis in childhood-onset and adult-onset SLE. Rheumatol. (United Kingdom). 2020;59:90–8.
49.
go back to reference Jacob N, Stohl W. Cytokine disturbances in systemic lupus erythematosus. Arthritis Res. Ther. 2011;13. Jacob N, Stohl W. Cytokine disturbances in systemic lupus erythematosus. Arthritis Res. Ther. 2011;13.
50.
go back to reference Karnell JL, Rieder SA, Ettinger R, Kolbeck R. Targeting the CD40-CD40L pathway in autoimmune diseases: Humoral immunity and beyond. Adv Drug Deliv Rev. 2019;141:92–103.CrossRef Karnell JL, Rieder SA, Ettinger R, Kolbeck R. Targeting the CD40-CD40L pathway in autoimmune diseases: Humoral immunity and beyond. Adv Drug Deliv Rev. 2019;141:92–103.CrossRef
51.
go back to reference Macedo ACL, Isaac L. Systemic lupus erythematosus and deficiencies of early components of the complement classical pathway. Front Immunol. 2016;7:1-7. Macedo ACL, Isaac L. Systemic lupus erythematosus and deficiencies of early components of the complement classical pathway. Front Immunol. 2016;7:1-7.
52.
go back to reference Haynes WA, Haddon DJ, Diep VK, Khatri A, Bongen E, Yiu G, Balboni I, Bolen CR, Mao R, Utz PJ, et al. Integrated, multicohort analysis reveals unified signature of systemic lupus erythematosus. JCI Insight. 2020;5(4):1-20. Haynes WA, Haddon DJ, Diep VK, Khatri A, Bongen E, Yiu G, Balboni I, Bolen CR, Mao R, Utz PJ, et al. Integrated, multicohort analysis reveals unified signature of systemic lupus erythematosus. JCI Insight. 2020;5(4):1-20.
53.
go back to reference Jakez-Ocampo J, Paulín-Vera CM, Gómez-Martín D, Lima G, Vargas-Rojas MI, Llorente L, Rivadeneyra-Espinoza L, Pérez-Romano B, Calva-Cevenini G, Ruiz-Argüelles A, et al. Vß T cell receptor (TCR) genes in circulating cells of patients with systemic lupus erythematosus and their healthy relatives. Gac Med Mex. 2018;154:74–9.PubMed Jakez-Ocampo J, Paulín-Vera CM, Gómez-Martín D, Lima G, Vargas-Rojas MI, Llorente L, Rivadeneyra-Espinoza L, Pérez-Romano B, Calva-Cevenini G, Ruiz-Argüelles A, et al. Vß T cell receptor (TCR) genes in circulating cells of patients with systemic lupus erythematosus and their healthy relatives. Gac Med Mex. 2018;154:74–9.PubMed
54.
go back to reference Ma K, Du W, Wang X, Yuan S, Cai X, Liu D, Li J, Lu L. Multiple functions of B cells in the pathogenesis of systemic lupus erythematosus. Int J. Mol. Sci. 2019;20:1-19. Ma K, Du W, Wang X, Yuan S, Cai X, Liu D, Li J, Lu L. Multiple functions of B cells in the pathogenesis of systemic lupus erythematosus. Int J. Mol. Sci. 2019;20:1-19.
55.
go back to reference Sharabi A, Tsokos GC. T cell metabolism: new insights in systemic lupus erythematosus pathogenesis and therapy. Nat Rev Rheumatol. 2020;16:100–12.CrossRef Sharabi A, Tsokos GC. T cell metabolism: new insights in systemic lupus erythematosus pathogenesis and therapy. Nat Rev Rheumatol. 2020;16:100–12.CrossRef
56.
go back to reference Matsuo T, Hashimoto M, Sakaguchi S, Sakaguchi N, Ito Y, Hikida M, Tsuruyama T, Sakai K, Yokoi H, Shirakashi M, et al. Strain-specific manifestation of lupus-like systemic autoimmunity caused by Zap70 mutation. J Immunol. 2019;202:3161–72.CrossRef Matsuo T, Hashimoto M, Sakaguchi S, Sakaguchi N, Ito Y, Hikida M, Tsuruyama T, Sakai K, Yokoi H, Shirakashi M, et al. Strain-specific manifestation of lupus-like systemic autoimmunity caused by Zap70 mutation. J Immunol. 2019;202:3161–72.CrossRef
57.
go back to reference Julià A, López-Longo FJ, Pérez Venegas JJ, Bonàs-Guarch S, Olivé À, Andreu JL, Aguirre-Zamorano MÁ, Vela P, Nolla JM, de la Fuente JLM, et al. Genome-wide association study meta-analysis identifies five new loci for systemic lupus erythematosus. Arthritis Res. Ther. 2018;20:1-10. Julià A, López-Longo FJ, Pérez Venegas JJ, Bonàs-Guarch S, Olivé À, Andreu JL, Aguirre-Zamorano MÁ, Vela P, Nolla JM, de la Fuente JLM, et al. Genome-wide association study meta-analysis identifies five new loci for systemic lupus erythematosus. Arthritis Res. Ther. 2018;20:1-10.
58.
go back to reference He J, Ma J, Ren B, Liu A. Advances in systemic lupus erythematosus pathogenesis via mTOR signaling pathway. Semin Arthritis Rheum. He J, Ma J, Ren B, Liu A. Advances in systemic lupus erythematosus pathogenesis via mTOR signaling pathway. Semin Arthritis Rheum.
59.
go back to reference Beşliu AN, Pistol G, Marica CM, Bǎnicǎ LM, Chiţonu C, Ionescu R, Tǎnǎseanu C, Tamsulea I, Matache C, Stefǎnescu M. PI3K/Akt signaling in peripheral T lymphocytes from systemic lupus erythematosus patients. Roum Arch Microbiol Immunol. 2009;68:69–79.PubMed Beşliu AN, Pistol G, Marica CM, Bǎnicǎ LM, Chiţonu C, Ionescu R, Tǎnǎseanu C, Tamsulea I, Matache C, Stefǎnescu M. PI3K/Akt signaling in peripheral T lymphocytes from systemic lupus erythematosus patients. Roum Arch Microbiol Immunol. 2009;68:69–79.PubMed
60.
go back to reference Wilbe M, Kozyrev SV, Farias FHG, Bremer HD, Hedlund A, Pielberg GR, Seppälä EH, Gustafson U, Lohi H, Carlborg Ö, et al. Multiple changes of gene expression and function reveal genomic and phenotypic complexity in SLE-like disease. PLoS Genet. 2015;11:1-27. Wilbe M, Kozyrev SV, Farias FHG, Bremer HD, Hedlund A, Pielberg GR, Seppälä EH, Gustafson U, Lohi H, Carlborg Ö, et al. Multiple changes of gene expression and function reveal genomic and phenotypic complexity in SLE-like disease. PLoS Genet. 2015;11:1-27.
61.
go back to reference Sui W, Hou X, Che W, Yang M, Dai Y. The applied basic research of systemic lupus erythematosus based on the biological omics. Genes Immun. 2013;14:133–46. Sui W, Hou X, Che W, Yang M, Dai Y. The applied basic research of systemic lupus erythematosus based on the biological omics. Genes Immun. 2013;14:133–46.
Metadata
Title
An integrative Bayesian network approach to highlight key drivers in systemic lupus erythematosus
Authors
Samaneh Maleknia
Zahra Salehi
Vahid Rezaei Tabar
Ali Sharifi-Zarchi
Kaveh Kavousi
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Arthritis Research & Therapy / Issue 1/2020
Electronic ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-020-02239-3

Other articles of this Issue 1/2020

Arthritis Research & Therapy 1/2020 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.