Extracellular vesicles package dsDNA to aggravate Crohn's disease by activating the STING pathway

Fan Zhao, Tao Zheng, Wenbin Gong, Jie Wu, Haohao Xie, Weijie Li, Rui Zhang, Peizhao Liu, Juanhan Liu, Xiuwen Wu, Yun Zhao, Jianan Ren, Fan Zhao, Tao Zheng, Wenbin Gong, Jie Wu, Haohao Xie, Weijie Li, Rui Zhang, Peizhao Liu, Juanhan Liu, Xiuwen Wu, Yun Zhao, Jianan Ren

Abstract

Crohn's disease (CD) is an intestinal immune-dysfunctional disease. Extracellular vesicles (EVs) are membrane-enclosed particles full of functional molecules, e.g., nuclear acids. Recently, EVs have been shown to participate in the development of CD by realizing intercellular communication among intestinal cells. However, the role of EVs carrying double-strand DNA (dsDNA) shed from sites of intestinal inflammation in CD has not been investigated. Here we isolated EVs from the plasma or colon lavage of murine colitis and CD patients. The level of exosomal dsDNA, including mtDNA and nDNA, significantly increased in murine colitis and active human CD, and was positively correlated with the disease activity. Moreover, the activation of the STING pathway was verified in CD. EVs from the plasma of active human CD triggered STING activation in macrophages in vitro. EVs from LPS-damaged colon epithelial cells were also shown to raise inflammation in macrophages via activating the STING pathway, but the effect disappeared after the removal of exosomal dsDNA. These findings were further confirmed in STING-deficient mice and macrophages. STING deficiency significantly ameliorated colitis. Besides, potential therapeutic effects of GW4869, an inhibitor of EVs release were assessed. The application of GW4869 successfully ameliorated murine colitis by inhibiting STING activation. In conclusion, exosomal dsDNA was found to promote intestinal inflammation via activating the STING pathway in macrophages and act as a potential mechanistic biomarker and therapeutic target of CD.

Conflict of interest statement

The authors declare no competing interests.

© 2021. The Author(s).

Figures

Fig. 1. Abnormally elevated exosomal dsDNA was…
Fig. 1. Abnormally elevated exosomal dsDNA was positively correlated with the disease activity in murine colitis and active human CD.
AC Classification of EVs in colon lavage of murine colitis. A Western blot of specific EVs markers: The simultaneous expression of three classic positive markers of EVs including Alix, CD63, and CD81, along with the absence of Calnexin, together identified EVs. The whole-cell lysate of CT26, a murine cell line, was used as a positive control. B Nanoparticle-tracking analysis of diluted EVs fractions was determined by ZetaView PMX 110 (Particle Metrix, Meerbusch, Germany). The distribution of EVs particle concentration (y axis) by size (x axis) was shown and 126.2-nm particles accounted for the highest proportion (98.5%). C Representative transmission electron microscope images of EVs fractions. The yellow arrow pointed to examples of EVs, which were cup-shape membrane-enclosed particles with diameters of 30–150 nm. D Absolute quantification of nDNA and mtDNA within EVs extracted from plasma and colon lavage of murine colitis. Hist1h3F gene and mtCOI gene were applied to represent nDNA and mtDNA respectively. Exosomal mtDNA and nDNA were significantly higher in the plasma and colon lavage of murine colitis. Murine colitis treated with GW4869 were also examined. n = 5–10/group. In the illustrations, healthy wild-type model, murine colitis, and murine colitis treated with GW4869 were abbreviated as WT, WT + DSS, and WT + DSS (GW4869 IP) respectively. Colon lavage was abbreviated as CL. E Classification of EVs isolated from plasma of active human CD, including western blot, nanoparticle-tracking analysis, and representative transmission electron microscope images. In the nanoparticle-tracking analysis, 117.7-nm particles accounted for the highest proportion (98.6%). In the representative transmission electron microscope image, the yellow arrow pointed to EVs. F Levels of exosomal nDNA and mtDNA from the plasma of CD patients were measured (n = 5/group). Both were significantly higher in patients with active CD. H3 clustered histone 7 gene and mtCOI gene were applied to represent nDNA and mtDNA respectively. G Correlations between the disease activity and levels of exosomal dsDNA in plasma in murine colitis and CD patients (Spearman’s rank correlation analysis). Disease activity index, Crohn’s Disease Activity Index, exosomal nDNA, and exosomal mtDNA were abbreviated as DAI, CDAI, exo-nDNA, and exo-mtDNA in the illustrations respectively. Data were displayed as mean values ± SD at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 2. EVs from damaged intestinal epithelial…
Fig. 2. EVs from damaged intestinal epithelial cells were internalized by macrophages in vitro.
A EVs presence in colons of human CD, exhibited by immunohistochemistry of CD63 (brown particles pointed by black arrows in the illustration), a transmembrane protein of EVs. The nuclei (blue) at the bottom of intestinal epithelial cells were pointed by black arrowheads. More EVs were stained in active CD and gathered in the intestinal epithelium. Colonoscopy images of corresponding patients were shown. B Transmission electron microscopy images of mitochondrial damages in colonic epithelia of murine colitis. Mitochondrial vesicular swelling and peripheral lucent zones were observed in the murine colitis model (abbreviated as WT + DSS in the illustration), compared to the wild-type healthy control. C Classification of EVs from LPS-damaged CT26 cells, including western blot, transmission electron microscopy, and nanoparticle-tracking analysis. The whole-cell lysate of CT26 was set as a positive control in the western blot experiment. In the nanoparticle-tracking analysis, 104.1-nm particles accounted for the highest proportion (98.5%). In the representative transmission electron microscope image, the yellow arrow pointed to EVs. D Labeled EVs were shown to be internalized by bone marrow-derived macrophages (BMDMs) after 15 h incubation. EVs from LPS-damaged CT26 cells were isolated and labeled with PKH26 (red). EVs from normal CT26 cells were used as control. BMDMs were treated with labeled EVs for 15 h and then observed under fluorescence confocal microscopy. F-actin (green) and nuclei (blue) of BMDMs were stained before the observation. EVs from damaged colon epithelial cells are abbreviated as c-EV in the illustration. All results were representative of at least three independent experiments.
Fig. 3. EVs activated the STING pathway…
Fig. 3. EVs activated the STING pathway and triggered macrophages to be pro-inflammatory in a dsDNA-dependent manner in CD.
A Trauma patients with no history of CD and no gastrointestinal symptoms were used as controls. Expressions of p-STING, STING, p-IRF3, IRF3, p-p65, and p65 in colon tissues of active CD patients and controls were determined by western blot. The activation of the STING pathway was shown in active CD. B Immunofluorescence co-staining of CD68, a macrophage marker, and phosphorylated STING (p-STING) was performed in the colonic mucosa of active human CD. Nuclei were counterstained with DAPI. Trauma patients with no history of CD and no gastrointestinal symptoms were used as controls. Scale bar, 20 μm. CE EVs from LPS-damaged CT26 cells were incubated with wild type (WT) or STING−/− BMDMs. EVs were shown to trigger inflammation in macrophages by activating STING pathway. C Activation of STING pathway in BMDMs was determined by western blot. D, E mRNA levels of inflammatory cytokines, Arg1 and IL12p40 in BMDMs. The increased mRNA levels of IL-6, TNF-α, IFN-β, IL12p40 and decreased mRNA level of Arg1 showed that inflammation in macrophages was triggered by EVs from LPS-damaged CT26 cells. EVs from LPS-damaged CT26 cells are abbreviated as c-EV in the illustrations. F, G EVs from the equal number of damaged CT26 cells were assigned to two groups. One was lysed by sonication and digested with dsDNase so that exosomal dsDNA was digested thoroughly. The other group was directly treated with dsDNase to digest free dsDNA outside EVs, therefore exosomal dsDNA stayed intact. WT and STING−/− BMDMs were treat with the two groups of EVs. Exosomal dsDNA was abbreviated as exoDNA in the illustrations. F Activation of STING pathway in BMDMs stimulated by the two groups of EVs. The EVs treated with dsDNase without sonication still activated the STING pathway in BMDMs while EVs treated with dsDNase plus sonication failed to. G, H mRNA levels of inflammatory cytokines, Arg1 and IL12p40 in BMDMs. EVs treated with dsDNase plus sonication failed to trigger inflammation in BMDMs. Data were displayed as mean values ± SD at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 4. Blockade of EVs release by…
Fig. 4. Blockade of EVs release by GW4869 inhibited intestinal inflammatory responses via inhibiting the activation of the STING pathway.
AD The expressions of p-STING, STING, p-IRF3, IRF3, p-p65, and p65 in colon tissues from Wild type (WT), STING−/−, WT murine colitis, WT murine colitis treated with GW4869, and STING−/− murine colitis models were determined by western blot. WT murine colitis was abbreviated as WT + DSS in the illustrations. WT murine colitis treated with GW4869 was abbreviated as WT + DSS (GW4869 IP). STING−/− murine colitis was abbreviated as WT + DSS. The activation of the STING pathway was observed in the WT murine colitis model and was inhibited in WT murine colitis treated with GW4869. E, F The proportion of TUNEL-positive cells was quantified to estimate intestinal apoptotic level in WT, WT murine colitis, WT murine colitis treated with GW4869, and STING−/− murine colitis models. The level of apoptosis was higher in the WT murine colitis model and lower in WT murine colitis treated with GW4869, and STING−/− murine colitis models. Representative images were shown. Scale bar = 50 µm. G The expression of inflammatory cytokines including IL-6, TNF-α, and IFN-β in the homogenates of murine colon tissues was measured by ELISA (n = 5–9/group). All results were representative of at least three independent experiments. Data were displayed as mean values ± SD. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 5. Either GW4869 administration to inhibit…
Fig. 5. Either GW4869 administration to inhibit EVs release or STING deficiency improved the disease prognosis of murine colitis.
AE Comparison of disease prognosis among six experimental murine models (n = 11–20/group). WT (GW4869 IP): to examine possible side effects of GW4869, GW4869 was administrated in normal wild-type mice. WT + DSS (GW4869 IP): Murine colitis models treated with GW4869. STING−/− + DSS: STING knockout (STING−/−) mice were employed and induced by DSS to establish colitis. Indicators of disease activity and severity were examined including A Disease activity index (DAI), B percent changes in body weight, C survival rate, D lengths and representative images of colons, E histological score of colons and representative colon H&E images. Data were displayed as mean values ± SD at least three independent experiments. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig. 6. Model of exosomal dsDNA-mediated intestinal…
Fig. 6. Model of exosomal dsDNA-mediated intestinal inflammation via activation of STING pathway in CD.
sEVs full of dsDNA, which could be secreted by damaged intestinal epithelial cells, were taken up by macrophages to activate the STING pathway and trigger inflammation. Inflammatory cytokines released by macrophages, such as IL-6, TNF-α and IFN-β, further damaged intestinal epithelial cells.

References

    1. Feuerstein JD, Cheifetz AS. Crohn disease: epidemiology, diagnosis, and management. Mayo Clin Proc. 2017;92:1088–103.
    1. Torres J, Mehandru S, Colombel JF, Peyrin-Biroulet L. Crohn’s disease. Lancet. 2017;389:1741–55.
    1. Wang G, Yuan J, Cai X, Xu Z, Wang J, Ocansey DKW, et al. HucMSC-exosomes carrying miR-326 inhibit neddylation to relieve inflammatory bowel disease in mice. Clin Transl Med. 2020;10:e113.
    1. Na YR, Stakenborg M, Seok SH, Matteoli G. Macrophages in intestinal inflammation and resolution: a potential therapeutic target in IBD. Nat Rev Gastroenterol Hepatol. 2019;16:531–43.
    1. Boyapati RK, Tamborska A, Dorward DA, Ho GT. Advances in the understanding of mitochondrial DNA as a pathogenic factor in inflammatory diseases. F1000Res. 2017;6:169.
    1. Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7:1535750.
    1. van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19:213–28.
    1. Shah R, Patel T, Freedman JE. Circulating extracellular vesicles in human disease. N. Engl J Med. 2018;379:958–66.
    1. Zempleni J, Sukreet S, Zhou F, Wu D, Mutai E. Milk-derived exosomes and metabolic regulation. Annu Rev Anim Biosci. 2019;7:245–62.
    1. Fu H, Hu D, Zhang L, Tang P. Role of extracellular vesicles in rheumatoid arthritis. Mol Immunol. 2018;93:125–32.
    1. Karpman D, Ståhl AL, Arvidsson I. Extracellular vesicles in renal disease. Nat Rev Nephrol. 2017;13:545–62.
    1. Kubo H. Extracellular vesicles in lung disease. Chest. 2018;153:210–6.
    1. Becker A, Thakur BK, Weiss JM, Kim HS, Peinado H, Lyden D. Extracellular vesicles in cancer: cell-to-cell mediators of metastasis. Cancer Cell. 2016;30:836–48.
    1. Budnik V, Ruiz-Cañada C, Wendler F. Extracellular vesicles round off communication in the nervous system. Nat Rev Neurosci. 2016;17:160–72.
    1. Bebelman MP, Smit MJ, Pegtel DM, Baglio SR. Biogenesis and function of extracellular vesicles in cancer. Pharm Ther. 2018;188:1–11.
    1. Chang W, Wang J. Exosomes and their noncoding RNA cargo are emerging as new modulators for diabetes mellitus. Cells. 2019;8:853.
    1. Tang D, Xu X, Ying J, Xie T, Cao G. Transfer of metastatic traits via miR-200c in extracellular vesicles derived from colorectal cancer stem cells is inhibited by atractylenolide I. Clin Transl Med. 2020;10:e139.
    1. Valter M, Verstockt S, Finalet Ferreiro JA, Cleynen I. Extracellular vesicles in inflammatory bowel disease: small particles, big players. J Crohns Colitis. 2021;15:499–510.
    1. Yang X, Meng S, Jiang H, Chen T, Wu W. Exosomes derived from interleukin-10-treated dendritic cells can inhibit trinitrobenzene sulfonic acid-induced rat colitis. Scand J Gastroenterol. 2010;45:1168–77.
    1. Carrière J, Bretin A, Darfeuille-Michaud A, Barnich N, Nguyen HT. Exosomes released from cells infected with Crohn’s disease-associated adherent-invasive Escherichia coli activate host innate immune responses and enhance bacterial intracellular replication. Inflamm Bowel Dis. 2016;22:516–28.
    1. Liu R, Tang A, Wang X, Chen X, Zhao L, Xiao Z, et al. Inhibition of lncRNA NEAT1 suppresses the inflammatory response in IBD by modulating the intestinal epithelial barrier and by exosome-mediated polarization of macrophages. Int J Mol Med. 2018;42:2903–13.
    1. Zheng X, Chen F, Zhang Q, Liu Y, You P, Sun S, et al. Salivary exosomal PSMA7: a promising biomarker of inflammatory bowel disease. Protein Cell. 2017;8:686–95.
    1. Lou H, Pickering MC. Extracellular DNA and autoimmune diseases. Cell Mol Immunol. 2018;15:746–55.
    1. Lian Q, Xu J, Yan S, Huang M, Ding H, Sun X, et al. Chemotherapy-induced intestinal inflammatory responses are mediated by exosome secretion of double-strand DNA via AIM2 inflammasome activation. Cell Res. 2017;27:784–800.
    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. Sokolova O, Naumann M. Crosstalk between DNA damage and inflammation in the multiple steps of gastric carcinogenesis. Curr Top Microbiol Immunol. 2019;421:107–37.
    1. Jeon H, Lee J, Lee S, Kang SK, Park SJ, Yoo SM, et al. Extracellular vesicles from KSHV-infected cells stimulate antiviral immune response through mitochondrial DNA. Front Immunol. 2019;10:876.
    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. Canesso MCC, Lemos L, Neves TC, Marim FM, Castro TBR, Veloso É S, et al. The cytosolic sensor STING is required for intestinal homeostasis and control of inflammation. Mucosal Immunol. 2018;11:820–34.
    1. Boyapati RK, Dorward DA, Tamborska A, Kalla R, Ventham NT, Doherty MK, et al. Mitochondrial DNA is a pro-inflammatory damage-associated molecular pattern released during active IBD. Inflamm Bowel Dis. 2018;24:2113–22.
    1. Wirtz S, Popp V, Kindermann M, Gerlach K, Weigmann B, Fichtner-Feigl S, et al. Chemically induced mouse models of acute and chronic intestinal inflammation. Nat Protoc. 2017;12:1295–309.
    1. Cooper HS, Murthy SN, Shah RS, Sedergran DJ. Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest. 1993;69:238–49.
    1. Erben U, Loddenkemper C, Doerfel K, Spieckermann S, Haller D, Heimesaat MM, et al. A guide to histomorphological evaluation of intestinal inflammation in mouse models. Int J Clin Exp Pathol. 2014;7:4557–76.
    1. Wu X, Ren J, Chen G, Wu L, Song X, Li G, et al. Systemic blockade of P2X7 receptor protects against sepsis-induced intestinal barrier disruption. Sci Rep. 2017;7:4364.
    1. Wei R, Zhao L, Kong G, Liu X, Zhu S, Zhang S, et al. Combination of size-exclusion chromatography and ultracentrifugation improves the proteomic profiling of plasma-derived small extracellular vesicles. Biol Proced Online. 2020;22:12.
    1. Boyapati RK, Rossi AG, Satsangi J, Ho GT. Gut mucosal DAMPs in IBD: from mechanisms to therapeutic implications. Mucosal Immunol. 2016;9:567–82.
    1. Wei M, Gao X, Liu L, Li Z, Wan Z, Dong Y, et al. Visceral adipose tissue derived exosomes exacerbate colitis severity via pro-inflammatory MiRNAs in high fat diet fed mice. ACS Nano. 2020;14:5099–110.
    1. Sharma A, Johnson A. Exosome DNA: critical regulator of tumor immunity and a diagnostic biomarker. J Cell Physiol. 2020;235:1921–32.
    1. Sansone P, Savini C, Kurelac I, Chang Q, Amato LB, Strillacci A, et al. Packaging and transfer of mitochondrial DNA via exosomes regulate escape from dormancy in hormonal therapy-resistant breast cancer. Proc Natl Acad Sci USA. 2017;114:E9066–e9075.
    1. Allenson K, Castillo J, San Lucas FA, Scelo G, Kim DU, Bernard V, et al. High prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Ann Oncol. 2017;28:741–7.
    1. Ishikawa H, Barber GN. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature. 2008;455:674–8.
    1. Burdette DL, Monroe KM, Sotelo-Troha K, Iwig JS, Eckert B, Hyodo M, et al. STING is a direct innate immune sensor of cyclic di-GMP. Nature. 2011;478:515–8.
    1. Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013;339:786–91.
    1. Martin GR, Blomquist CM, Henare KL, Jirik FR. Stimulator of interferon genes (STING) activation exacerbates experimental colitis in mice. Sci Rep. 2019;9:14281.
    1. Ahn J, Son S, Oliveira SC, Barber GN. STING-dependent signaling underlies IL-10 controlled inflammatory colitis. Cell Rep. 2017;21:3873–84.

Source: PubMed

3
Se inscrever