Plasma-derived DNA containing-extracellular vesicles induce STING-mediated proinflammatory responses in dermatomyositis

Yubin Li, Christina Bax, Jay Patel, Thomas Vazquez, Adarsh Ravishankar, Muhammad M Bashir, Madison Grinnell, DeAnna Diaz, Victoria P Werth, Yubin Li, Christina Bax, Jay Patel, Thomas Vazquez, Adarsh Ravishankar, Muhammad M Bashir, Madison Grinnell, DeAnna Diaz, Victoria P Werth

Abstract

Objectives: Extracellular vesicles (EVs) are lipid bilayer membrane vesicles that are present in various bodily fluids and have been implicated in autoimmune disease pathogenesis. Type I interferons (IFN), specifically IFN-β, are uniquely elevated in dermatomyositis (DM). The stimulator of interferon genes (STING) works as a critical nucleic acid sensor and adaptor in type I IFN signaling with possible implications in autoimmune diseases such as DM. In the current study, we investigated whether circulating EVs contribute to proinflammatory effects in DM, whether these proinflammatory responses are mediated by the STING signaling pathway, and if so, by what mechanism STING is activated. Methods: We collected and characterized EVs from plasma of healthy controls (HC) and DM patients; analyzed their abilities to trigger proinflammatory cytokines release by ELISA, and explored STING signaling pathway activation using immunoblot and immunofluorescent staining. STING signaling pathway inhibitors and RNAi were used to further investigate whether STING was involved in EVs-triggered proinflammatory response. DNase/lipid destabilizing agent was utilized to digest EVs and their captured DNA contents to evaluate how EVs triggered STING-mediated proinflammatory response in DM. Results: EVs isolated from DM plasma triggered proinflammatory cytokines including type I IFN release with STING signaling pathway activation. The activated STING pathway was preferentially mediated by dsDNA captured by EVs. Suppression of STING or its downstream signaling proteins attenuated the EVs-mediated proinflammatory response. Conclusions: Plasma-derived, DNA containing-EVs induced STING-mediated proinflammatory effects in DM. Targeting the STING pathway may be a potential therapeutic approach for DM.

Keywords: dermatomyositis; dsDNA; extracellular vesicles; stimulator of interferon genes; type I interferon.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

© The author(s).

Figures

Figure 1
Figure 1
DM plasma-derived small extracellular vesicles are different compared to HC plasma-derived small extracellular vesicles. (A) Concentration and size distribution of small extracellular vesicles (sEVs) derived from HC plasma vs. DM plasma. (B) Total number of sEVs derived from HC plasma (4.545x1011 ± 4.019x1011 particles/mL, n = 7) and DM plasma (1.309x1012 ± 6.377x1011 particles/mL, n = 11). (C) Average size of sEVs derived from healthy plasma (83.69 ± 1.057 nm, n = 7) and DM plasma (75.33 ± 3.786 nm, n = 11). (D) Number of EVs with different surface markers CD81, CD9, and CD63 from 35 µL of HC plasma (n = 3) and DM plasma (n = 3). (E) Corresponding density of EVs with different surface makers CD81, CD9, and CD63 from HC plasma and DM plasma. (F) Representative immunoblot showing expression level of surface makers CD81, CD9, and CD63 in 0.5 µL of HC plasma (n = 3) vs DM plasma (n = 3). (G) Expression level relative intensity of CD81, CD9, and CD63 in 0.5 µL of HC plasma (n = 6) vs DM plasma (n = 6). Data in (B,C,D,G) represent mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 between groups as indicated. Comparison between two groups was analyzed by the Student t test.
Figure 2
Figure 2
DM plasma-derived extracellular vesicles triggered more pro-inflammatory cytokine release in PBMCs than HC plasma-derived extracellular vesicles. 100 µL of PBS-resuspended EVs derived from 1mL plasma of 5 healthy donors and 14 DM patients were used to stimulate PBMCs (1.5x106/mL, 2 mL/well) for 15 h. The supernatants were collected for interferon β (IFNβ), tumor necrosis factor α (TNFα), and interleukin 6 (IL-6) detection. (A) DM plasma-derived EVs triggered more IFNβ release (25.39 ± 5.700 pg/mL, n = 14) than HC plasma-derived EVs (4.285 ± 2.727 pg/mL, n = 5). (B) DM plasma-derived EVs triggered more TNFα release (1193 ± 561.8 pg/mL, n = 14) than HC plasma-derived EVs (329.0 ± 244.9 pg/mL, n = 5). (C) DM plasma-derived EVs triggered more IL6 release (600.2 ± 240.0 pg/mL, n = 14) than HC plasma-derived EVs (264.6 ± 31.30 pg/mL, n = 5). (D) DM plasma-derived EVs induced more STING phosphorylation than healthy plasma-derived EVs in PBMCs. (E) Relative intensity of phosphorylated STING in DM plasma- or HC plasma-derived EVs stimulated PBMCs (n = 3). Data in (A,B,C,E) represent mean ± SD. *P < 0.05, ***P < 0.001 between groups as indicated. Comparison between two groups was analyzed by the Student t test.
Figure 3
Figure 3
DM plasma derived extracellular vesicles induced STING phosphorylation during their triggered pro-inflammatory response in PBMCs. (A) Representative immunofluorescent staining images showing that EVs derived from DM plasma induced phosphorylation of STING in PBMCs. (Scale bar 100 µm) (B) Bar graph depicting the relative intensity of phosphorylated STING immunofluorescent staining in PBMCs with/without DM plasma-derived EVs stimulation (n = 5). (C) DM plasma-derived small EVs (sEVs) triggered more IFNβ release(30.24 ± 1.302 pg/mL, n = 4) than large EVs (lEVs) (7.22 ± 0.9899 pg/mL, n = 4) in PBMCs; DM plasma-derived sEVs (1407 ± 247.0 pg/mL, n = 4) triggered more TNFα release than lEVs (269.9 ± 62.53 pg/mL, n = 4) in PBMCs; DM plasma-derived sEVs (886.2 ± 120.4 pg/mL, n = 4) triggered more IL6 release than lEVs (468.7 ± 75.15 pg/mL, n = 4) in PBMCs. (D) Graph of DM Activity of Cutaneous Dermatomyositis Disease Area and Severity Index (CDASI) vs. DM sEVs concentration (n = 11) and lEVs concentration (n = 7). R2 = coefficient of correlation. Data in B represent median, data in C represent mean ± SD. **P < 0.01, ***P < 0.001 between groups as indicated. Comparison between two groups was analyzed by the Student t test.
Figure 4
Figure 4
Inhibition of STING impaired immunostimulatory effects of DM plasma derived small extracellular vesicles in PBMCs. (A) sEVs-stimulated PBMCs secreted less IFNβ release when STING antagonist H-151 (1μM) was present ((21.58 ± 5.45 vs. 28.34 ± 4.25) pg/mL; n = 6). (B) sEVs-stimulated PBMCs secreted less TNFα release when STING antagonist H-151 was present (434.8 ± 231.5 vs. 919.1 ± 325.7) pg/mL; n = 6). (C) sEVs-stimulated PBMCs secreted less IL6 release when STING antagonist H-151 was present ((611.5 ± 132.8 vs. 844.2 ± 180.3) pg/mL; n = 6). (D) STING antagonist H-151 suppressed DM plasma-derived sEV-induced STING phosphorylation and its downstream signaling pathway TBK1, IRF3, and NFκB phosphorylation in PBMCs. (E) Relative intensity of phosphorylated STING, phosphorylated TBK1, phosphorylated IRF3, and phosphorylated NFκB in PBMCs stimulated with/without DM derived sEVs in the presence/absence of STING antagonist H-151 (n = 3). Data in (A,B,C,E) represent mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 between groups as indicated. Comparison among three or more groups was performed using ANOVA, followed by Student-Newman-Keuls test. Comparison between two groups was analyzed by the Student t test.
Figure 5
Figure 5
Silencing of STING suppressed immunostimulatory effects of DM plasma derived small extracellular vesicles in PBMCs. (A) Representative immunoblot images showed that total STING level was decreased in siSTING transfected PBMCs, and DM plasma-derived sEVs induced more STING phosphorylation in siCTRL transfected PBMCs than siSTING transfected PBMCs. (B) DM plasma-derived sEVs induced more STING phosphorylation in siCTRL transfected PBMCs than siSTING transfected PBMCs (n = 3). (C) Total STING level was decreased in siSTING transfected PBMCs (n = 3). (D) DM plasma-derived sEVs induced more IFNβ release in siCTRL transfected PBMCs (29.32 ± 4.299 pg/mL; n = 5) than siSTING transfected PBMCs (17.18 ± 6.531 pg/mL; n = 5). Data in (B,C,D) represent mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 between groups as indicated. Comparison among three or more groups was performed using ANOVA, followed by Student-Newman-Keuls test.
Figure 6
Figure 6
Inhibition of TBK1 decreased DM plasma derived small extracellular vesicles' immunostimulatory effects in PBMCs. (A) Representative immunofluorescent staining images showing that sEVs derived from DM plasma induced phosphorylation of TBK1 in PBMCs. (Scale bar 100 µm) (B) Bar graphic depicting the relative intensity of phosphorylated TBK1 immunofluorescent staining in PBMCs with/without DM plasma-derived sEVs stimulation (n = 5). (C) TBK1 inhibitors suppressed DM plasma-derived sEVs induced TBK1 and IRF3 phosphorylation in PBMCs. (D) DM plasma-derived sEVs induced IFNβ release in PBMCs (11.40 ± 4.669 pg/mL, n = 5) when compared with untreated PBMCs (2.000 ± 0.7674 pg/mL, n = 5). 2.5 µM of Amlexanox (TBK1 inhibitor) pretreatment impaired sEVs-triggered IFNβ release in PBMCs (3.933 ± 2.002 pg/mL, n = 5); 2.5 µM of MRT67307 (TBK1 inhibitor) pretreatment impaired DM sEVs-triggered IFNβ release in PBMCs (4.067 ± 1.511 pg/mL, n = 5). Data in B represent median. Data in D represent mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 between groups as indicated. Comparison between two groups was analyzed by the Student t test. Comparison among three or more groups was performed using ANOVA, followed by Student-Newman-Keuls test.
Figure 7
Figure 7
Digestion of DM plasma-derived small extracellular vesicles-captured DNA impaired their triggered pro-inflammatory response in PBMCs. (A) Pre-treatment with Triton X-100 and DNase (DNase I or dsDNase) attenuated sEVs ability to trigger IFNβ release in PBMCs (n = 3-6). (B) Pre-treatment with Triton X-100 and DNase (DNase I or dsDNase) attenuated sEVs ability to trigger TNFα release in PBMCs (n = 3-6). (C) Pre-treatment with Triton X-100 and DNase (DNase I or dsDNase) attenuated sEVs ability to trigger IL6 release in PBMCs (n = 3-6). (D) Genomic/mitochondrial DNA captured by EVs in the presence/absence of 0.075% Triton X-100 with/without DNase I was assessed by PCR using relative specified primers. (E) Genomic/mitochondrial DNA captured by EVs in the presence/absence of 0.075% Triton X-100 with/without dsDNase was assessed by PCR using relative specified primers. Data in (A,B,C) represent mean ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 between groups as indicated. Comparison among three or more groups was performed using ANOVA, followed by Student-Newman-Keuls test.
Figure 8
Figure 8
Digestion of DM plasma-derived small extracellular vesicles-captured DNA impaired their triggered STING signaling pathway activation in PBMCs. (A) The effects of DM plasma derived sEVs pretreated in the presence/absence of 0.075% Triton X-100 with/without DNase I on STING phosphorylation and its downstream signaling pathway TBK1, and IRF3 phosphorylation in PBMCs. (B) The effects of DM plasma derived sEVs pretreated in the presence/absence of 0.075% Triton X-100 with/without dsDNase on STING phosphorylation and its downstream signaling pathway TBK1, and IRF3 phosphorylation in PBMCs. Relative intensity of phosphorylated STING (C), phosphorylated TBK1(D), and phosphorylated IRF3 (E) in PBMCs stimulated by DM plasma derived sEVs pretreated in the presence/absence of 0.075% Triton X-100 with/without DNase I (n = 3). Relative intensity of phosphorylated STING (F), phosphorylated TBK1(G), and phosphorylated IRF3 (H) in PBMCs stimulated by DM plasma derived sEVs pretreated in the presence/absence of 0.075% Triton X-100 with/without dsDNase (n = 3). Data were represent mean ± SD. *P < 0.05,**P < 0.01 between groups as indicated. Comparison among three or more groups was performed using ANOVA, followed by Student-Newman-Keuls test.

References

    1. Okogbaa J, Batiste L. Dermatomyositis: An acute flare and current treatments. Clin Med Insights Case Rep. 2019;12:1179547619855370.
    1. DeWane ME, Waldman R, Lu J. Dermatomyositis: Clinical features and pathogenesis. J Am Acad Dermatol. 2020;82:267–81.
    1. Marvi U, Chung L, Fiorentino DF. Clinical presentation and evaluation of dermatomyositis. Indian J Dermatol. 2012;57:375–81.
    1. Baechler EC, Bauer JW, Slattery CA, Ortmann WA, Espe KJ, Novitzke J. et al. An interferon signature in the peripheral blood of dermatomyositis patients is associated with disease activity. Mol Med. 2007;13:59–68.
    1. Baechler EC, Bilgic H, Reed AM. Type i interferon pathway in adult and juvenile dermatomyositis. Arthritis Res Ther. 2011;13:249.
    1. Wong D, Kea B, Pesich R, Higgs BW, Zhu W, Brown P. et al. Interferon and biologic signatures in dermatomyositis skin: Specificity and heterogeneity across diseases. PLoS One. 2012;7:e29161.
    1. Huard C, Gulla SV, Bennett DV, Coyle AJ, Vleugels RA, Greenberg SA. Correlation of cutaneous disease activity with type 1 interferon gene signature and interferon beta in dermatomyositis. Br J Dermatol. 2017;176:1224–30.
    1. Greenberg SA. Dermatomyositis and type 1 interferons. Curr Rheumatol Rep. 2010;12:198–203.
    1. Gurtler C, Bowie AG. Innate immune detection of microbial nucleic acids. Trends Microbiol. 2013;21:413–20.
    1. Barber GN. Sting: Infection, inflammation and cancer. Nat Rev Immunol. 2015;15:760–70.
    1. Chen Q, Sun L, Chen ZJ. Regulation and function of the cgas-sting pathway of cytosolic DNA sensing. Nat Immunol. 2016;17:1142–9.
    1. Motwani M, Pesiridis S, Fitzgerald KA. DNA sensing by the cgas-sting pathway in health and disease. Nat Rev Genet. 2019;20:657–74.
    1. Liu S, Cai X, Wu J, Cong Q, Chen X, Li T. et al. Phosphorylation of innate immune adaptor proteins mavs, sting, and trif induces irf3 activation. Science. 2015;347:aaa2630.
    1. Tanaka Y, Chen ZJ. Sting specifies irf3 phosphorylation by tbk1 in the cytosolic DNA signaling pathway. Sci Signal. 2012;5:ra20.
    1. Abe T, Barber GN. Cytosolic-DNA-mediated, sting-dependent proinflammatory gene induction necessitates canonical nf-kappab activation through tbk1. J Virol. 2014;88:5328–41.
    1. Devhare PB, Ray RB. Extracellular vesicles: Novel mediator for cell to cell communications in liver pathogenesis. Mol Aspects Med. 2018;60:115–22.
    1. Lane RE, Korbie D, Hill MM, Trau M. Extracellular vesicles as circulating cancer biomarkers: Opportunities and challenges. Clin Transl Med. 2018;7:14.
    1. Turpin D, Truchetet ME, Faustin B, Augusto JF, Contin-Bordes C, Brisson A. et al. Role of extracellular vesicles in autoimmune diseases. Autoimmun Rev. 2016;15:174–83.
    1. Jiang K, Karasawa R, Hu Z, Chen Y, Holmes L, O'Neil KM. et al. Plasma exosomes from children with juvenile dermatomyositis are taken up by human aortic endothelial cells and are associated with altered gene expression in those cells. Pediatr Rheumatol Online J. 2019;17:41.
    1. Baka Z, Senolt L, Vencovsky J, Mann H, Simon PS, Kittel A. et al. Increased serum concentration of immune cell derived microparticles in polymyositis/dermatomyositis. Immunol Lett. 2010;128:124–30.
    1. Brown M, Davies DH, Skinner MA, Bowen G, Hollingsworth SJ, Mufti GJ. et al. Antigen gene transfer to cultured human dendritic cells using recombinant avipoxvirus vectors. Cancer Gene Ther. 1999;6:238–45.
    1. Sauer JD, Sotelo-Troha K, von Moltke J, Monroe KM, Rae CS, Brubaker SW. et al. The n-ethyl-n-nitrosourea-induced goldenticket mouse mutant reveals an essential function of sting in the in vivo interferon response to listeria monocytogenes and cyclic dinucleotides. Infect Immun. 2011;79:688–94.
    1. Saito S, Nakano M. Nitric oxide production by peritoneal macrophages of mycobacterium bovis bcg-infected or non-infected mice: Regulatory role of t lymphocytes and cytokines. J Leukoc Biol. 1996;59:908–15.
    1. Durrieu L, Bharadwaj A, Waisman DM. Analysis of the thrombotic and fibrinolytic activities of tumor cell-derived extracellular vesicles. Blood Adv. 2018;2:1054–65.
    1. Chen G, Huang AC, Zhang W, Zhang G, Wu M, Xu W. et al. Exosomal pd-l1 contributes to immunosuppression and is associated with anti-pd-1 response. Nature. 2018;560:382–6.
    1. Torralba D, Baixauli F, Villarroya-Beltri C, Fernandez-Delgado I, Latorre-Pellicer A, Acin-Perez R. et al. Priming of dendritic cells by DNA-containing extracellular vesicles from activated t cells through antigen-driven contacts. Nat Commun. 2018;9:2658.
    1. Harkonen K, Oikari S, Kyykallio H, Capra J, Hakkola S, Ketola K, Cd44s assembles hyaluronan coat on filopodia and extracellular vesicles and induces tumorigenicity of mkn74 gastric carcinoma cells. Cells. 2019. 8.
    1. Li Y, Zhu H, Wang S, Qian X, Fan J, Wang Z. et al. Interplay of oxidative stress and autophagy in pamam dendrimers-induced neuronal cell death. Theranostics. 2015;5:1363–77.
    1. Kahraman T, Gucluler G, Simsek I, Yagci FC, Yildirim M, Ozen C. et al. Circulating ll37 targets plasma extracellular vesicles to immune cells and intensifies behcet's disease severity. J Extracell Vesicles. 2017;6:1284449.
    1. Li Y, Li M, Weigel B, Mall M, Werth VP, Liu ML. Nuclear envelope rupture and net formation is driven by pkcalpha-mediated lamin b disassembly. EMBO Rep. 2020;21:e48779.
    1. Dong SXM, Caballero R, Ali H, Roy DLF, Cassol E, Kumar A. Transfection of hard-to-transfect primary human macrophages with bax sirna to reverse resveratrol-induced apoptosis. RNA Biol. 2020;17:755–64.
    1. Zhang Y, Jin X, Liang J, Guo Y, Sun G, Zeng X. et al. Extracellular vesicles derived from odn-stimulated macrophages transfer and activate cdc42 in recipient cells and thereby increase cellular permissiveness to ev uptake. Sci Adv. 2019;5:eaav1564.
    1. Anyanwu CO, Fiorentino DF, Chung L, Dzuong C, Wang Y, Okawa J. et al. Validation of the cutaneous dermatomyositis disease area and severity index: Characterizing disease severity and assessing responsiveness to clinical change. Br J Dermatol. 2015;173:969–74.
    1. Haag SM, Gulen MF, Reymond L, Gibelin A, Abrami L, Decout A. et al. Targeting sting with covalent small-molecule inhibitors. Nature. 2018;559:269–73.
    1. Lee JM, Ghonime MG, Cassady KA. Sting restricts ohsv replication and spread in resistant mpnsts but is dispensable for basal ifn-stimulated gene upregulation. Mol Ther Oncolytics. 2019;15:91–100.
    1. Chen KL, Zeidi M, Werth VP. Recent advances in pharmacological treatments of adult dermatomyositis. Curr Rheumatol Rep. 2019;21:53.
    1. Somani AK, Swick AR, Cooper KD, McCormick TS. Severe dermatomyositis triggered by interferon beta-1a therapy and associated with enhanced type i interferon signaling. Arch Dermatol. 2008;144:1341–9.
    1. Katsiougiannis S. Extracellular vesicles: Evolving contributors in autoimmunity. For Immunopathol Dis Therap. 2015;6:163–70.
    1. Chen KL, Patel J, Zeidi M, Wysocka M, Bashir MM, Patel B, Myeloid dendritic cells are major producers of interferon-beta in dermatomyositis and may contribute to hydroxychloroquine refractoriness. J Invest Dermatol. 2021.
    1. Sarhan J, Liu BC, Muendlein HI, Weindel CG, Smirnova I, Tang AY. et al. Constitutive interferon signaling maintains critical threshold of mlkl expression to license necroptosis. Cell death and differentiation. 2019;26:332–47.
    1. Burdette DL, Vance RE. Sting and the innate immune response to nucleic acids in the cytosol. Nat Immunol. 2013;14:19–26.
    1. Kato Y, Park J, Takamatsu H, Konaka H, Aoki W, Aburaya S. 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;77:1507–15.
    1. Jiang J, Zhao M, Chang C, Wu H, Lu Q. Type i interferons in the pathogenesis and treatment of autoimmune diseases. Clin Rev Allergy Immunol. 2020;59:248–72.
    1. Patel J, Maddukuri S, Li Y, Bax C, Werth VP. Highly multiplexed mass cytometry identifies the immunophenotype in the skin of dermatomyositis. J Invest Dermatol. 2021.
    1. Deschamps T, Kalamvoki M. Extracellular vesicles released by herpes simplex virus 1-infected cells block virus replication in recipient cells in a sting-dependent manner. J Virol. 2018. 92.
    1. Doyle LM, Wang MZ. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 2019. 8.
    1. Cassius C, Le Buanec H, Bouaziz JD, Amode R. Biomarkers in adult dermatomyositis: Tools to help the diagnosis and predict the clinical outcome. Journal of immunology research. 2019;2019:9141420.
    1. Shu X, Peng Q, Lu X, Wang G. Hmgb1 may be a biomarker for predicting the outcome in patients with polymyositis /dermatomyositis with interstitial lung disease. PLoS One. 2016;11:e0161436.
    1. Thim-Uam A, Prabakaran T, Tansakul M, Makjaroen J, Wongkongkathep P, Chantaravisoot N. et al. Sting mediates lupus via the activation of conventional dendritic cell maturation and plasmacytoid dendritic cell differentiation. iScience. 2020;23:101530.
    1. Chen H, Sun H, You F, Sun W, Zhou X, Chen L. et al. Activation of stat6 by sting is critical for antiviral innate immunity. Cell. 2011;147:436–46.
    1. Takahashi A, Okada R, Nagao K, Kawamata Y, Hanyu A, Yoshimoto S. et al. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat Commun. 2017;8:15287.
    1. Yamamoto S, Niida S, Azuma E, Yanagibashi T, Muramatsu M, Huang TT. et al. Inflammation-induced endothelial cell-derived extracellular vesicles modulate the cellular status of pericytes. Sci Rep. 2015;5:8505.
    1. He WR, Cao LB, Yang YL, Hua D, Hu MM, Shu HB. Vrk2 is involved in the innate antiviral response by promoting mitostress-induced mtdna release. Cell Mol Immunol. 2021.
    1. Lood C, Blanco LP, Purmalek MM, Carmona-Rivera C, De Ravin SS, Smith CK. et al. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nature medicine. 2016;22:146–53.
    1. Kahlenberg JM, Kaplan MJ. Little peptide, big effects: The role of ll-37 in inflammation and autoimmune disease. Journal of immunology. 2013;191:4895–901.
    1. Chamilos G, Gregorio J, Meller S, Lande R, Kontoyiannis DP, Modlin RL. et al. Cytosolic sensing of extracellular self-DNA transported into monocytes by the antimicrobial peptide ll37. Blood. 2012;120:3699–707.
    1. Apel F, Andreeva L, Knackstedt LS, Streeck R, Frese CK, Goosmann C, The cytosolic DNA sensor cgas recognizes neutrophil extracellular traps. Sci Signal. 2021. 14.

Source: PubMed

Подписаться