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

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

Increased MLKL mRNA level in the PBMCs is correlated with autoantibody production, renal involvement, and SLE disease activity

Authors: Mingjiao Zhang, Hongyu Jie, Yong Wu, Xinai Han, Xing Li, Yi He, Xingliang Shi, Yuwei Luo, Ying Sun, Jinlong Yang, Jing Yang, Shulv Quan, Xiaobin Lao, Liping Tan, Erwei Sun

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

Login to get access

Abstract

Background

Necroptosis is a form of regulated necrosis that is involved in various autoimmune diseases. Mixed lineage kinase domain-like pseudokinase (MLKL) has been identified as a key executor of necroptosis; however, the significance of MLKL in peripheral blood mononuclear cells (PBMCs) of systemic lupus erythematosus (SLE) has not been investigated. In this study, we aimed to determine the mRNA level of MLKL in PBMCs and examine its relationship with clinical features and serological parameters in SLE.

Methods

Real-time transcription-polymerase chain reaction (RT-PCR) analysis was used to determine the expression of MLKL mRNA in PBMCs from 59 patients with SLE, 25 patients with rheumatoid arthritis (RA), and 30 age- and sex-matched healthy controls (HC). Spearman’s correlation test was performed to assess the correlation of MLKL mRNA with clinical variables. The receiver operating characteristic (ROC) curve was created to evaluate the diagnostic value.

Results

Our results showed MLKL mRNA in PBMCs was upregulated in SLE patients compared to that in RA and HC individuals. SLE patients positive for antinuclear antibodies had significantly higher MLKL mRNA than antibody-negative patients. In SLE patients, MLKL mRNA was found to be upregulated in patients with lupus nephritis (LN) as compared with patients without LN, and also higher in active patients than in stable patients. MLKL mRNA level was significantly and positively correlated with c-reaction protein (CRP) (r = 0.3577, p = 0.0237), erythrocyte sedimentation rate (ESR) (r = 0.4091, p = 0.0043), serum immunoglobulin G (IgG) concentration (r = 0.3546, p = 0.0289), and the numbers of positive antinuclear antibodies (ANAs) (r = 0.3945, p = 0.0432). ROC analysis showed that MLKL mRNA in PBMCs had an area under the curve of 0.9277 (95% CI 0.8779–0.9775, p < 0.001) to discriminate SLE from controls.

Conclusions

These results suggest that increased MLKL mRNA level in the PBMCs of SLE patients is correlated with renal involvement and disease activity, identifying a subgroup of patients with SLE or LN who may benefit from early diagnosis and therapies targeting MLKL.
Literature
1.
go back to reference Yeoh S, Dias SS, Isenberg DA. Advances in systemic lupus erythematosus. Medicine. 2018;46(2):84–92. Yeoh S, Dias SS, Isenberg DA. Advances in systemic lupus erythematosus. Medicine. 2018;46(2):84–92.
2.
go back to reference Olsen NJ, Karp DR. Autoantibodies and SLE—the threshold for disease. Nat Rev Rheumatol. 2014;10(3):181–6.PubMed Olsen NJ, Karp DR. Autoantibodies and SLE—the threshold for disease. Nat Rev Rheumatol. 2014;10(3):181–6.PubMed
3.
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(12):716–30.PubMed Tsokos GC, Lo MS, Reis PC, Sullivan KE. New insights into the immunopathogenesis of systemic lupus erythematosus. Nat Rev Rheumatol. 2016;12(12):716–30.PubMed
4.
go back to reference Visan I. Necroptosis regulator. Nat Immunol. 2015;16(5):447. Visan I. Necroptosis regulator. Nat Immunol. 2015;16(5):447.
5.
go back to reference He S, Liang Y, Shao F, Wang X. Toll-like receptors activate programmed necrosis in macrophages through a receptor-interacting kinase-3-mediated pathway. Proc Natl Acad Sci U S A. 2011;108(50):20054–9.PubMedPubMedCentral He S, Liang Y, Shao F, Wang X. Toll-like receptors activate programmed necrosis in macrophages through a receptor-interacting kinase-3-mediated pathway. Proc Natl Acad Sci U S A. 2011;108(50):20054–9.PubMedPubMedCentral
6.
go back to reference Sarhan J, Liu BC, Muendlein HI, Weindel CG, Smirnova I, Tang AY, Ilyukha V, Sorokin M, Buzdin A, Fitzgerald KA, et al. Constitutive interferon signaling maintains critical threshold of MLKL expression to license necroptosis. Cell Death Differ. 2019;26(2):332–47.PubMed Sarhan J, Liu BC, Muendlein HI, Weindel CG, Smirnova I, Tang AY, Ilyukha V, Sorokin M, Buzdin A, Fitzgerald KA, et al. Constitutive interferon signaling maintains critical threshold of MLKL expression to license necroptosis. Cell Death Differ. 2019;26(2):332–47.PubMed
7.
go back to reference Maeda A, Fadeel B. Mitochondria released by cells undergoing TNF-α-induced necroptosis act as danger signals. Cell Death Dis. 2014;5(7):e1312.PubMedPubMedCentral Maeda A, Fadeel B. Mitochondria released by cells undergoing TNF-α-induced necroptosis act as danger signals. Cell Death Dis. 2014;5(7):e1312.PubMedPubMedCentral
8.
go back to reference Someda M, Kuroki S, Miyachi H, Tachibana M, Yonehara S. Caspase-8, receptor-interacting protein kinase 1 (RIPK1), and RIPK3 regulate retinoic acid-induced cell differentiation and necroptosis. Cell Death Differ. 2020;27(5):1539-53. Someda M, Kuroki S, Miyachi H, Tachibana M, Yonehara S. Caspase-8, receptor-interacting protein kinase 1 (RIPK1), and RIPK3 regulate retinoic acid-induced cell differentiation and necroptosis. Cell Death Differ. 2020;27(5):1539-53.
9.
go back to reference Weber K, Roelandt R, Bruggeman I, Estornes Y, Vandenabeele P. Nuclear RIPK3 and MLKL contribute to cytosolic necrosome formation and necroptosis. Commun Biol. 2018;1(1):6.PubMedPubMedCentral Weber K, Roelandt R, Bruggeman I, Estornes Y, Vandenabeele P. Nuclear RIPK3 and MLKL contribute to cytosolic necrosome formation and necroptosis. Commun Biol. 2018;1(1):6.PubMedPubMedCentral
10.
go back to reference Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature. 2015;517(7534):311–20.PubMed Pasparakis M, Vandenabeele P. Necroptosis and its role in inflammation. Nature. 2015;517(7534):311–20.PubMed
11.
go back to reference Guo C, Fu R, Zhou M, Wang S, Huang Y, Hu H, Zhao J, Gaskin F, Yang N, Fu SM. Pathogenesis of lupus nephritis: RIP3 dependent necroptosis and NLRP3 inflammasome activation. J Autoimmun. 2019;103:102286.PubMedPubMedCentral Guo C, Fu R, Zhou M, Wang S, Huang Y, Hu H, Zhao J, Gaskin F, Yang N, Fu SM. Pathogenesis of lupus nephritis: RIP3 dependent necroptosis and NLRP3 inflammasome activation. J Autoimmun. 2019;103:102286.PubMedPubMedCentral
12.
go back to reference Franklyn K, Hoi A, Nikpour M, Morand EF. The need to define treatment goals for systemic lupus erythematosus. Nat Rev Rheumatol. 2014;10(9):567–71.PubMed Franklyn K, Hoi A, Nikpour M, Morand EF. The need to define treatment goals for systemic lupus erythematosus. Nat Rev Rheumatol. 2014;10(9):567–71.PubMed
13.
go back to reference Salem D, Subang R, Kuwana M, Levine JS, Rauch J. T cells from induced and spontaneous models of SLE recognize a common T cell epitope on β2-glycoprotein I. Cell Mol Immunol. 2019;16(8):685–93.PubMed Salem D, Subang R, Kuwana M, Levine JS, Rauch J. T cells from induced and spontaneous models of SLE recognize a common T cell epitope on β2-glycoprotein I. Cell Mol Immunol. 2019;16(8):685–93.PubMed
14.
go back to reference Morel L. Immunometabolism in systemic lupus erythematosus. Nat Rev Rheumatol. 2017;13(5):280–90.PubMed Morel L. Immunometabolism in systemic lupus erythematosus. Nat Rev Rheumatol. 2017;13(5):280–90.PubMed
15.
go back to reference Catalina MD, Bachali P, Geraci NS, Grammer AC, Lipsky PE. Gene expression analysis delineates the potential roles of multiple interferons in systemic lupus erythematosus. Commun Biol. 2019;2(1):140.PubMedPubMedCentralCrossRef Catalina MD, Bachali P, Geraci NS, Grammer AC, Lipsky PE. Gene expression analysis delineates the potential roles of multiple interferons in systemic lupus erythematosus. Commun Biol. 2019;2(1):140.PubMedPubMedCentralCrossRef
16.
go back to reference Fan H, Liu F, Dong G, Ren D, Xu Y, Dou J, Wang T, Sun L, Hou Y. Activation-induced necroptosis contributes to B-cell lymphopenia in active systemic lupus erythematosus. Cell Death Dis. 2014;5(9):e1416.PubMedPubMedCentralCrossRef Fan H, Liu F, Dong G, Ren D, Xu Y, Dou J, Wang T, Sun L, Hou Y. Activation-induced necroptosis contributes to B-cell lymphopenia in active systemic lupus erythematosus. Cell Death Dis. 2014;5(9):e1416.PubMedPubMedCentralCrossRef
17.
go back to reference Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40(9):1725.CrossRef Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40(9):1725.CrossRef
18.
go back to reference Bombardier C, Gladman DD, Urowitz MB, Caron D, Chang CH. Derivation of the SLEDAI. A disease activity index for lupus patients. The Committee on Prognosis Studies in SLE. Arthritis Rheum. 1992;35(6):630–40.PubMedCrossRef Bombardier C, Gladman DD, Urowitz MB, Caron D, Chang CH. Derivation of the SLEDAI. A disease activity index for lupus patients. The Committee on Prognosis Studies in SLE. Arthritis Rheum. 1992;35(6):630–40.PubMedCrossRef
19.
go back to reference Paludan SR, Reinert LS, Hornung V. DNA-stimulated cell death: implications for host defence, inflammatory diseases and cancer. Nat Rev Immunol. 2019;19(3):141–53.PubMedPubMedCentralCrossRef Paludan SR, Reinert LS, Hornung V. DNA-stimulated cell death: implications for host defence, inflammatory diseases and cancer. Nat Rev Immunol. 2019;19(3):141–53.PubMedPubMedCentralCrossRef
21.
go back to reference Kato Y, Park J, Takamatsu H, Konaka H, Aoki W, Aburaya S, Ueda M, Nishide M, Koyama S, Hayama Y, et al. Apoptosis-derived membrane vesicles drive the cGAS-STING pathway and enhance type I IFN production in systemic lupus erythematosus. Ann Rheum Dis. 2018;77(10):1507–15.PubMedPubMedCentral Kato Y, Park J, Takamatsu H, Konaka H, Aoki W, Aburaya S, Ueda M, Nishide M, Koyama S, Hayama Y, et al. Apoptosis-derived membrane vesicles drive the cGAS-STING pathway and enhance type I IFN production in systemic lupus erythematosus. Ann Rheum Dis. 2018;77(10):1507–15.PubMedPubMedCentral
22.
go back to reference Liu M, Guo Q, Wu C, Sterlin D, Goswami S, Zhang Y, Li T, Bao C, Shen N, Fu Q, et al. Type I interferons promote the survival and proinflammatory properties of transitional B cells in systemic lupus erythematosus patients. Cell Mol Immunol. 2019;16(4):367–79.PubMed Liu M, Guo Q, Wu C, Sterlin D, Goswami S, Zhang Y, Li T, Bao C, Shen N, Fu Q, et al. Type I interferons promote the survival and proinflammatory properties of transitional B cells in systemic lupus erythematosus patients. Cell Mol Immunol. 2019;16(4):367–79.PubMed
23.
go back to reference Niewold TB, Hua J, Lehman TJA, Harley JB, Crow MK. High serum IFN-α activity is a heritable risk factor for systemic lupus erythematosus. Genes Immun. 2007;8(6):492–502.PubMedPubMedCentral Niewold TB, Hua J, Lehman TJA, Harley JB, Crow MK. High serum IFN-α activity is a heritable risk factor for systemic lupus erythematosus. Genes Immun. 2007;8(6):492–502.PubMedPubMedCentral
24.
go back to reference Almaani S, Meara A, Rovin BH. Update on lupus nephritis. Clin J Am Soc Nephrol. 2017;12(5):825–35.PubMed Almaani S, Meara A, Rovin BH. Update on lupus nephritis. Clin J Am Soc Nephrol. 2017;12(5):825–35.PubMed
25.
go back to reference Nnodum B, Dudley L. Class IV lupus nephritis in the setting of serologically quiescent disease and normal urine sediment in a patient with late-onset systemic lupus erythematosus. Case Rep Rheumatol. 2019;2019:1219529.PubMedPubMedCentral Nnodum B, Dudley L. Class IV lupus nephritis in the setting of serologically quiescent disease and normal urine sediment in a patient with late-onset systemic lupus erythematosus. Case Rep Rheumatol. 2019;2019:1219529.PubMedPubMedCentral
26.
go back to reference Scharer CD, Blalock EL, Mi T, Barwick BG, Jenks SA, Deguchi T, Cashman KS, Neary BE, Patterson DG, Hicks SL, et al. Epigenetic programming underpins B cell dysfunction in human SLE. Nat Immunol. 2019;20(8):1071–82.PubMedPubMedCentral Scharer CD, Blalock EL, Mi T, Barwick BG, Jenks SA, Deguchi T, Cashman KS, Neary BE, Patterson DG, Hicks SL, et al. Epigenetic programming underpins B cell dysfunction in human SLE. Nat Immunol. 2019;20(8):1071–82.PubMedPubMedCentral
27.
go back to reference Vasquez-Canizares N, Wahezi D, Putterman C. Diagnostic and prognostic tests in systemic lupus erythematosus. Best Pract Res Clin Rheumatol. 2017;31(3):351–63.PubMedPubMedCentral Vasquez-Canizares N, Wahezi D, Putterman C. Diagnostic and prognostic tests in systemic lupus erythematosus. Best Pract Res Clin Rheumatol. 2017;31(3):351–63.PubMedPubMedCentral
Metadata
Title
Increased MLKL mRNA level in the PBMCs is correlated with autoantibody production, renal involvement, and SLE disease activity
Authors
Mingjiao Zhang
Hongyu Jie
Yong Wu
Xinai Han
Xing Li
Yi He
Xingliang Shi
Yuwei Luo
Ying Sun
Jinlong Yang
Jing Yang
Shulv Quan
Xiaobin Lao
Liping Tan
Erwei Sun
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/s13075-020-02332-7

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.