The NLRP3 inflammasome pathway contributes to chronic inflammation in experimental autoimmune uveitis

Avik Shome , Ilva D. Rupenthal , Rachael L. Niederer , Odunayo O. Mugisho

Animal Models and Experimental Medicine ›› 2025, Vol. 8 ›› Issue (6) : 1080 -1094.

PDF
Animal Models and Experimental Medicine ›› 2025, Vol. 8 ›› Issue (6) : 1080 -1094. DOI: 10.1002/ame2.70011
ORIGINAL ARTICLE

The NLRP3 inflammasome pathway contributes to chronic inflammation in experimental autoimmune uveitis

Author information +
History +
PDF

Abstract

Background: Noninfectious uveitis, a chronic ocular inflammatory disease, is characterized by the activation of immune cells in the eye, with most studies focusing on the role of the adaptive immune system in the disease. However, limited data exist on the potential contribution of the innate immune system, specifically the nucleotide-binding oligomerization domain and leucine-rich repeat receptor-3 (NLRP3) inflammasome pathway. This pathway has previously been identified as a driver of inflammation in several low-grade, progressive inflammatory eye diseases such as diabetic retinopathy. The aim of this study was to determine whether the NLRP3 inflammasome pathway plays a role in the pathogenesis and chronicity of experimental autoimmune uveitis (EAU).

Methods: EAU was induced in C57BL/6J mice via intraperitoneal pertussis toxin and subcutaneous interphotoreceptor retinoid-binding protein injections. After 12 weeks, eyes were enucleated, and whole eye sections were assessed for inflammasome, macrophage, and microglial markers in the retina, ciliary body, and cornea using immunohistochemistry.

Results: Our study confirmed higher NLRP3 inflammasome activation (increased expression of NLRP3 and cleaved caspase 1 labeling) in EAU mouse retinas compared to controls. This correlated with increased astrogliosis and microglial activation throughout the eye. Migratory innate and adaptive peripheral immune cells (macrophages and leukocytes) were also found within the retina and ciliary body of EAU mice. Connexin43 proteins, which form hexameric hemichannels that can release adenosine triphosphate (ATP), an upstream inflammasome priming signal, were also found upregulated in the retina and cornea of EAU mice.

Conclusion: Overall, our findings support the idea that in the EAU model there is active inflammation, even 12 weeks post induction, and that it can be correlated to inflammasome activation. This contributes to the pathogenesis and chronicity of noninfectious uveitis, and our results emphasize that targeting the inflammasome pathway could be efficacious for noninfectious uveitis treatment.

Keywords

experimental autoimmune uveitis / inflammation / nucleotide-binding oligomerization domain and leucine-rich repeat receptor-3 (NLRP3) inflammasome / uveitis

Cite this article

Download citation ▾
Avik Shome, Ilva D. Rupenthal, Rachael L. Niederer, Odunayo O. Mugisho. The NLRP3 inflammasome pathway contributes to chronic inflammation in experimental autoimmune uveitis. Animal Models and Experimental Medicine, 2025, 8(6): 1080-1094 DOI:10.1002/ame2.70011

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Forrester JV, Klaska IP, Yu T, Kuffova L. Uveitis in mouse and man. Int Rev Immunol. 2013; 32(1): 76-96.

[2]

Durrani OM, Meads CA, Murray PI. Uveitis: a potentially blinding disease. Ophthalmologica. 2004; 218(4): 223-236.

[3]

Barisani-Asenbauer T, Maca SM, Mejdoubi L, Emminger W, Machold K, Auer H. Uveitis- a rare disease often associated with systemic diseases and infections- a systematic review of 2619 patients. Orphanet J Rare Dis. 2012; 7: 57.

[4]

Damsker JM, Hansen AM, Caspi RR. Th1 and Th17 cells. Ann NY Acad Sci. 2010; 1183(1): 211-221.

[5]

Guo P, Hirano M, Herrin BR, et al. Dual nature of the adaptive immune system in lampreys. Nature. 2009; 459(7248): 796-801.

[6]

Sun D, Liang D, Kaplan HJ, Shao H. The role of Th17-associated cytokines in the pathogenesis of experimental autoimmune uveitis (EAU). Cytokine. 2015; 74(1): 76-80.

[7]

Zhong Z, Su G, Kijlstra A, Yang P. Activation of the interleukin-23/interleukin-17 signalling pathway in autoinflammatory and autoimmune uveitis. Prog Retin Eye Res. 2020; 100: 866.

[8]

Karkhur S, Hasanreisoglu M, Vigil E, et al. Interleukin-6 inhibition in the management of non-infectious uveitis and beyond. J Ophthalmic Inflamm Infect. 2019; 9(1): 17.

[9]

Veldhoen M. Interleukin 17 is a chief orchestrator of immunity. Nat Immunol. 2017; 18(6): 612-621.

[10]

Cunha-Vaz J, Bernardes R, Lobo C. Blood-retinal barrier. Eur J Ophthalmol. 2011; 21(6): S3-S9.

[11]

Willermain F, Rosenbaum JT, Bodaghi B, et al. Interplay between innate and adaptive immunity in the development of non-infectious uveitis. Prog Retin Eye Res. 2012; 31(2): 182-194.

[12]

Fusco R, Siracusa R, Genovese T, Cuzzocrea S, Di Paola R. Focus on the role of NLRP3 Inflammasome in diseases. Int J Mol Sci. 2020; 21(12): 4223.

[13]

Shome A, Mugisho OO, Niederer RL, Rupenthal ID. Blocking the inflammasome: a novel approach to treat uveitis. Drug Discov Today. 2021; 26(12): 2839-2857.

[14]

Mugisho OO, Rupenthal ID, Paquet-Durand F, Acosta ML, Green CR. Targeting connexin hemichannels to control the inflammasome: the correlation between connexin43 and NLRP3 expression in chronic eye disease. Expert Opin Ther Targets. 2019; 23(10): 855-863.

[15]

Lim RR, Wieser ME, Ganga RR, et al. NOD-like receptors in the eye: uncovering its role in diabetic retinopathy. Int J Mol Sci. 2020; 21(3): 899.

[16]

Mills KH, Dungan LS, Jones SA, Harris J. The role of inflammasome-derived IL-1 in driving IL-17 responses. J Leukoc Biol. 2013; 93(4): 489-497.

[17]

Zhao R, Zhou H, Zhang J, Liu X, Su SB. Interleukin-1beta promotes the induction of retinal autoimmune disease. Int Immunopharmacol. 2014; 22(2): 285-292.

[18]

Li H, Zhu L, Wang R, et al. Aging weakens Th17 cell pathogenicity and ameliorates experimental autoimmune uveitis in mice. Protein Cell. 2022; 13(6): 422-445.

[19]

Kumar B, Cashman SM, Kumar-Singh R. Complement-mediated activation of the NLRP3 Inflammasome and its inhibition by AAV-mediated delivery of CD59 in a model of uveitis. Mol Ther. 2018; 26(6): 1568-1580.

[20]

Chen H, Deng Y, Gan X, et al. NLRP12 collaborates with NLRP3 and NLRC4 to promote pyroptosis inducing ganglion cell death of acute glaucoma. Mol Neurodegener. 2020; 15(1): 26.

[21]

Ildefonso CJ, Biswal MR, Ahmed CM, Lewin AS. The NLRP3 Inflammasome and its role in age-related macular degeneration. Adv Exp Med Biol. 2016; 854: 59-65.

[22]

Kuo C, Green CR, Rupenthal ID, Mugisho OO. Connexin43 hemichannel block protects against retinal pigment epithelial cell barrier breakdown. Acta Diabetol. 2020; 57(1): 13-22.

[23]

Agarwal RK, Caspi RR. Rodent models of experimental autoimmune uveitis. Methods Mol Med. 2004; 102: 395-419.

[24]

Agarwal RK, Silver PB, Caspi RR. Rodent models of experimental autoimmune uveitis. Methods Mol Biol. 2012; 900: 443-469.

[25]

Harimoto K, Ito M, Karasawa Y, Sakurai Y, Takeuchi M. Evaluation of mouse experimental autoimmune uveoretinitis by spectral domain optical coherence tomography. Br J Ophthalmol. 2014; 98(6): 808-812.

[26]

Shome A, Mugisho OO, Niederer RL, Rupenthal ID. Comprehensive grading system for experimental autoimmune uveitis in mice. Biomedicines. 2023; 11(7): 2022.

[27]

Mugisho OO, Aryal J, Shome A, et al. Orally delivered Connexin43 hemichannel blocker, Tonabersat, inhibits vascular breakdown and Inflammasome activation in a mouse model of diabetic retinopathy. Int J Mol Sci. 2023; 24(4): 3876.

[28]

Fan NW, Li J, Mittal SK, et al. Characterization of clinical and immune responses in an experimental chronic autoimmune uveitis model. Am J Pathol. 2021; 191(3): 425-437.

[29]

Zhang Y, Yang W, Li W, Zhao Y. NLRP3 inflammasome: checkpoint connecting innate and adaptive immunity in autoimmune diseases. Front Immunol. 2021; 12: 732933.

[30]

Balamurugan S, Das D, Hasanreisoglu M, et al. Interleukins and cytokine biomarkers in uveitis. Indian J Ophthalmol. 2020; 68(9): 1750-1763.

[31]

Netea MG, van de Veerdonk FL, van der Meer JWM, Dinarello CA, Joosten LAB. Inflammasome-independent regulation of IL-1-family cytokines. Annu Rev Immunol. 2015; 33(1): 49-77.

[32]

Dick AD, Carter D, Robertson M, et al. Control of myeloid activity during retinal inflammation. J Leukoc Biol. 2003; 74(2): 161-166.

[33]

Okunuki Y, Mukai R, Nakao T, et al. Retinal microglia initiate neuroinflammation in ocular autoimmunity. Proc Natl Acad Sci USA. 2019; 116(20): 9989-9998.

[34]

Rosmus D-D, Wieghofer P. Guardians of the eye: new tales about retinal microglia and other ocular macrophages. Neural Regen Res. 2022; 17(6): 1275-1277.

[35]

Wieghofer P, Hagemeyer N, Sankowski R, et al. Mapping the origin and fate of myeloid cells in distinct compartments of the eye by single-cell profiling. EMBO J. 2021; 40(6): e105123.

[36]

Forrester JV, Kuffova L, Dick AD. Autoimmunity, autoinflammation, and infection in uveitis. Am J Ophthalmol. 2018; 189: 77-85.

[37]

Shao H, Liao T, Ke Y, Shi H, Kaplan HJ, Sun D. Severe chronic experimental autoimmune uveitis (EAU) of the C57BL/6 mouse induced by adoptive transfer of IRBP1-20-specific T cells. Exp Eye Res. 2006; 82(2): 323-331.

[38]

Willbanks GA, Rootman DS, Jay V, Wiggert B, Chamberlain J, Inman RD. Experimental autoimmune uveitis in HLA-B27 transgenic mice. Hum Immunol. 1997; 53(2): 188-194.

[39]

Traugott U. Detailed analysis of early immunopathologic events during lesion formation in acute experimental autoimmune encephalomyelitis. Cell Immunol. 1989; 119(1): 114-129.

[40]

Jiang G, Ke Y, Sun D, Han G, Kaplan HJ, Shao H. Reactivation of Uveitogenic T cells by retinal astrocytes derived from experimental autoimmune uveitis-prone B10RIII mice. Invest Ophthalmol Vis Sci. 2008; 49(1): 282-289.

[41]

Kuo CYJ, Maran JJ, Jamieson EG, Rupenthal ID, Murphy R, Mugisho OO. Characterization of NLRP3 inflammasome activation in the onset of diabetic retinopathy. Int J Mol Sci. 2022; 23(22): 14-471.

[42]

Mugisho OO, Green CR, Squirrell DM, et al. Connexin43 hemichannel block protects against the development of diabetic retinopathy signs in a mouse model of the disease. J Mol Med. 2019; 97(2): 215-229.

[43]

Nor MNM, Rupenthal ID, Green CR, Acosta ML. Connexin hemichannel block using orally delivered Tonabersat improves outcomes in animal models of retinal disease. Neurotherapeutics. 2020; 17(1): 371-387.

[44]

Liu X, Ye F, Xiong H, et al. IL-1β induces IL-6 production in retinal Müller cells predominantly through the activation of P38 MAPK/NF-κB signaling pathway. Exp Cell Res. 2015; 331(1): 223-231.

[45]

Li W, Liu X, Tu Y, et al. Dysfunctional Nurr1 promotes high glucose-induced Müller cell activation by up-regulating the NF-κB/NLRP3 inflammasome axis. Neuropeptides. 2020; 82: 102057.

[46]

Lalor SJ, Dungan LS, Sutton CE, Basdeo SA, Fletcher JM, Mills KHG. Caspase-1-processed cytokines IL-1β and IL-18 promote IL-17 production by γδ and CD4 T cells that mediate autoimmunity. J Immunol. 2011; 186(10): 5738-5748.

[47]

Fankhauser C, Friedlander RM, Gagliardini V. Prevention of nuclear localization of activated caspases correlates with inhibition of apoptosis. Apoptosis. 2000; 5(2): 117-132.

[48]

Mao P-L, Jiang Y, Wee BY, Porter AG. Activation of Caspase-1 in the nucleus requires nuclear translocation of pro-caspase-1 mediated by its prodomain. J Biol Chem. 1998; 273(37): 23-621.

[49]

McKee CM, Coll RC. NLRP3 inflammasome priming: a riddle wrapped in a mystery inside an enigma. J Leukoc Biol. 2020; 108(3): 937-952.

[50]

Cliff CL, Williams BM, Chadjichristos CE, Mouritzen U, Squires PE, Hills CE. Connexin43: a target for the treatment of inflammation in secondary complications of the kidney and eye in diabetes. Int J Mol Sci. 2022; 23(2): 600.

[51]

Xu CY, Zhang WS, Zhang H, Cao Y, Zhou HY. The role of Connexin-43 in the inflammatory process: a new potential therapy to influence keratitis. J Ophthalmol. 2019; 2019: 9312827.

[52]

Li W, Bao GQ, Chen WQ, et al. Connexin43 hemichannel as a novel mediator of sterile and infectious inflammatory diseases. Sci Rep. 2018; 8(1): 1-15.

[53]

Acosta ML, Nor MNM, Guo CX, et al. Connexin therapeutics: blocking connexin hemichannel pores is distinct from blocking pannexin channels or gap junctions. Neural Regen Res. 2020; 16(3): 482.

[54]

Cronin M, Anderson PN, Cook JE, Green CR, Becker DL. Blocking connexin43 expression reduces inflammation and improves functional recovery after spinal cord injury. Mol Cell Neurosci. 2008; 39(2): 152-160.

[55]

Akanuma S-i, Higashi H, Maruyama S, et al. Expression and function of connexin43 protein in mouse and human retinal pigment epithelial cells as hemichannels and gap junction proteins. Exp Eye Res. 2018; 168: 128-137.

[56]

Retamal MA, Altenberg GA. Role and posttranslational regulation of Cx46 Hemichannels and gap junction channels in the eye lens. Front Physiol. 2022; 13: 864948.

RIGHTS & PERMISSIONS

2025 The Author(s). Animal Models and Experimental Medicine published by John Wiley & Sons Australia, Ltd on behalf of The Chinese Association for Laboratory Animal Sciences.

AI Summary AI Mindmap
PDF

10

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/