Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

邮发代号 80-967

2019 Impact Factor: 3.421

Frontiers of Medicine  2023, Vol. 17 Issue (4): 781-795   https://doi.org/10.1007/s11684-023-0986-x
  本期目录
Hyperosmolarity promotes macrophage pyroptosis by driving the glycolytic reprogramming of corneal epithelial cells in dry eye disease
Yu Han, Yu Zhang, Kelan Yuan, Yaying Wu, Xiuming Jin, Xiaodan Huang()
Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, China
 全文: PDF(6388 KB)   HTML
Abstract

Tear film hyperosmolarity plays a core role in the development of dry eye disease (DED) by mediating the disruption of ocular surface homeostasis and triggering inflammation in ocular surface epithelium. In this study, the mechanisms involving the hyperosmolar microenvironment, glycolysis mediating metabolic reprogramming, and pyroptosis were explored clinically, in vitro, and in vivo. Data from DED clinical samples indicated that the expression of glycolysis and pyroptosis-related genes, including PKM2 and GSDMD, was significantly upregulated and that the secretion of IL-1β significantly increased. In vitro, the indirect coculture of macrophages derived from THP-1 and human corneal epithelial cells (HCECs) was used to discuss the interaction among cells. The hyperosmolar environment was found to greatly induce HCECs’ metabolic reprogramming, which may be the primary cause of the subsequent inflammation in macrophages upon the activation of the related gene and protein expression. 2-Deoxy-d-glucose (2-DG) could inhibit the glycolysis of HCECs and subsequently suppress the pyroptosis of macrophages. In vivo, 2-DG showed potential efficacy in relieving DED activity and could significantly reduce the overexpression of genes and proteins related to glycolysis and pyroptosis. In summary, our findings suggested that hyperosmolar-induced glycolytic reprogramming played an active role in promoting DED inflammation by mediating pyroptosis.

Key wordsdry eye disease    glycolytic reprogramming    pyroptosis    inflammation    2-DG
收稿日期: 2022-03-06      出版日期: 2023-10-12
Corresponding Author(s): Xiaodan Huang   
 引用本文:   
. [J]. Frontiers of Medicine, 2023, 17(4): 781-795.
Yu Han, Yu Zhang, Kelan Yuan, Yaying Wu, Xiuming Jin, Xiaodan Huang. Hyperosmolarity promotes macrophage pyroptosis by driving the glycolytic reprogramming of corneal epithelial cells in dry eye disease. Front. Med., 2023, 17(4): 781-795.
 链接本文:  
https://academic.hep.com.cn/fmd/CN/10.1007/s11684-023-0986-x
https://academic.hep.com.cn/fmd/CN/Y2023/V17/I4/781
Gene Forward primer sequence Reverse primer sequence
Human-NLRP3 5′-GATCTTCGCTGCGATCAACAG-3′ 5′-CGTGCATTATCTGAACCCCAC-3′
Human-caspase-1 5′-TTTCCGCAAGGTTCGATTTTCA-3′ 5′-GGCATCTGCGCTCTACCATC-3′
Human-GSDMD 5′-GTGTGTCAACCTGTCTATCAAGG-3′ 5′-CATGGCATCGTAGAAGTGGAAG-3′
Human-IL-1β 5′-ATGATGGCTTATTACAGTGGCAA-3′ 5′-GTCGGAGATTCGTAGCTGGA-3′
Human-HK1 5′-GCTCTCCGATGAAACTCTCATAG-3′ 5′-GGACCTTACGAATGTTGGCAA-3′
Human-TPI1 5′-CTCATCGGCACTCTGAACG-3′ 5′-GCGAAGTCGATATAGGCAGTAGG-3′
Human-GAPDH 5′-GGAGCGAGATCCCTCCAAAAT-3′ 5′- GGCTGTTGTCATACTTCTCATGG-3′
Human-PGAM 5′-TCTGGAGGCGCTCCTATGAT-3′ 5′-TCTGTGAGGTCTGCATACCTG-3′
Human-ENO1 5′-GCCGTGAACGAGAAGTCCTG-3′ 5′-ACGCCTGAAGAGACTCGGT-3′
Human-PKM2 5′-ATGTCGAAGCCCCATAGTGAA-3′ 5′-TGGGTGGTGAATCAATGTCCA-3′
Mouse-Nlrp3 5′-TGTGAGAAGCAGGTTCTACTCT-3′ 5′-TGTAGCGACTGTTGAGGTCCA-3′
Mouse-caspase-1 5′-AATACAACCACTCGTACACGTC-3′ 5′-AGCTCCAACCCTCGGAGAAA-3′
Mouse-Gsdmd 5′-TTCCAGTGCCTCCATGAATGT-3′ 5′-GCTGTGGACCTCAGTGATCT-3′
Mouse-Il-1β 5′-GAAATGCCACCTTTTGACAGTG-3′ 5′-TGGATGCTCTCATCAGGACAG-3′
Mouse-Pkm2 5′-CGCCTGGACATTGACTCTG-3′ 5′-GAAATTCAGCCGAGCCACATT-3′
Tab.1  
Fig.1  
Fig.2  
Fig.3  
Fig.4  
Fig.5  
Fig.6  
1 F Stapleton, M Alves, VY Bunya, I Jalbert, K Lekhanont, F Malet, KS Na, D Schaumberg, M Uchino, J Vehof, E Viso, S Vitale, L Jones. TFOS DEWS II epidemiology report. Ocul Surf 2017; 15(3): 334–365
https://doi.org/10.1016/j.jtos.2017.05.003 pmid: 28736337
2 KF Farrand, M Fridman, IÖ Stillman, DA Schaumberg. Prevalence of diagnosed dry eye disease in the United States among adults aged 18 years and older. Am J Ophthalmol 2017; 182: 90–98
https://doi.org/10.1016/j.ajo.2017.06.033 pmid: 28705660
3 P Song, W Xia, M Wang, X Chang, J Wang, S Jin, J Wang, W Wei, I Rudan. Variations of dry eye disease prevalence by age, sex and geographic characteristics in China: a systematic review and meta-analysis. J Glob Health 2018; 8(2): 020503
https://doi.org/10.7189/jogh.08.020503 pmid: 30206477
4 L Jones, LE Downie, D Korb, JM Benitez-Del-Castillo, R Dana, SX Deng, PN Dong, G Geerling, RY Hida, Y Liu, KY Seo, J Tauber, TH Wakamatsu, J Xu, JS Wolffsohn, JP Craig. TFOS DEWS II management and therapy report. Ocul Surf 2017; 15(3): 575–628
https://doi.org/10.1016/j.jtos.2017.05.006 pmid: 28736343
5 SC Pflugfelder, CS de Paiva. The pathophysiology of dry eye disease: what we know and future directions for research. Ophthalmology 2017; 124(11): S4–S13
https://doi.org/10.1016/j.ophtha.2017.07.010 pmid: 29055361
6 Y Dai, J Zhang, J Xiang, Y Li, D Wu, J Xu. Calcitriol inhibits ROS-NLRP3-IL-1β signaling axis via activation of Nrf2-antioxidant signaling in hyperosmotic stress stimulated human corneal epithelial cells. Redox Biol 2019; 21: 101093
https://doi.org/10.1016/j.redox.2018.101093 pmid: 30611121
7 YF Meng, Q Pu, SY Dai, Q Ma, X Li, W Zhu. Nicotinamide mononucleotide alleviates hyperosmolarity-induced IL-17a secretion and macrophage activation in corneal epithelial cells/macrophage co-culture system. J Inflamm Res 2021; 14: 479–493
https://doi.org/10.2147/JIR.S292764 pmid: 33658825
8 FJ Holly, MA Lemp. Tear physiology and dry eyes. Surv Ophthalmol 1977; 22(2): 69–87
https://doi.org/10.1016/0039-6257(77)90087-X pmid: 335548
9 Y Jiang, C Yang, Y Zheng, Y Liu, Y Chen. A set of global metabolomic biomarker candidates to predict the risk of dry eye disease. Front Cell Dev Biol 2020; 8: 344
https://doi.org/10.3389/fcell.2020.00344 pmid: 32582687
10 MV Riley. Glucose and oxygen utilization by the rabbit cornea. Exp Eye Res 1969; 8(2): 193–200
https://doi.org/10.1016/S0014-4835(69)80031-X pmid: 5786869
11 P Rao, S Suvas. Development of inflammatory hypoxia and prevalence of glycolytic metabolism in progressing herpes stromal keratitis lesions. J Immunol 2019; 202(2): 514–526
https://doi.org/10.4049/jimmunol.1800422 pmid: 30530484
12 V Chayakul, M Reim. The enzymatic activities in the alkali-burnt rabbit cornea. Graefes Arch Clin Exp Ophthalmol 1982; 218(3): 145–148
https://doi.org/10.1007/BF02215652 pmid: 7095440
13 IC You, TG Coursey, F Bian, FL Barbosa, CS de Paiva, SC Pflugfelder. Macrophage phenotype in the ocular surface of experimental murine dry eye disease. Arch Immunol Ther Exp (Warsz) 2015; 63(4): 299–304
https://doi.org/10.1007/s00005-015-0335-0 pmid: 25772203
14 D Zhou, YT Chen, F Chen, M Gallup, T Vijmasi, AF Bahrami, LB Noble, N van Rooijen, NA McNamara. Critical involvement of macrophage infiltration in the development of Sjögren’s syndrome-associated dry eye. Am J Pathol 2012; 181(3): 753–760
https://doi.org/10.1016/j.ajpath.2012.05.014 pmid: 22770665
15 L Yu, C Yu, H Dong, Y Mu, R Zhang, Q Zhang, W Liang, W Li, X Wang, L Zhang. Recent developments about the pathogenesis of dry eye disease: based on immune inflammatory mechanisms. Front Pharmacol 2021; 12: 732887
https://doi.org/10.3389/fphar.2021.732887 pmid: 34421626
16 MS Milner, KA Beckman, JI Luchs, QB Allen, RM Awdeh, J Berdahl, TS Boland, C Buznego, JP Gira, DF Goldberg, D Goldman, RK Goyal, MA Jackson, J Katz, T Kim, PA Majmudar, RP Malhotra, MB McDonald, RK Rajpal, T Raviv, S Rowen, N Shamie, JD Solomon, K Stonecipher, S Tauber, W Trattler, KA Walter, GO 4th Waring, RJ Weinstock, WF Wiley, E Yeu. Dysfunctional tear syndrome: dry eye disease and associated tear film disorders—new strategies for diagnosis and treatment. Curr Opin Ophthalmol 2017; 27(Suppl 1): 3–47
https://doi.org/10.1097/01.icu.0000512373.81749.b7 pmid: 28099212
17 S Wang, YH Yuan, NH Chen, HB Wang. The mechanisms of NLRP3 inflammasome/pyroptosis activation and their role in Parkinson’s disease. Int Immunopharmacol 2019; 67: 458–464
https://doi.org/10.1016/j.intimp.2018.12.019 pmid: 30594776
18 A Wree, A Eguchi, MD McGeough, CA Pena, CD Johnson, A Canbay, HM Hoffman, AE Feldstein. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology 2014; 59(3): 898–910
https://doi.org/10.1002/hep.26592 pmid: 23813842
19 G Lorenz, MN Darisipudi, HJ Anders. Canonical and non-canonical effects of the NLRP3 inflammasome in kidney inflammation and fibrosis. Nephrol Dial Transplant 2014; 29(1): 41–48
https://doi.org/10.1093/ndt/gft332 pmid: 24026244
20 SB Kovacs, EA Miao. Gasdermins: effectors of pyroptosis. Trends Cell Biol 2017; 27(9): 673–684
https://doi.org/10.1016/j.tcb.2017.05.005 pmid: 28619472
21 Q Ji, L Wang, J Liu, Y Wu, H Lv, Y Wen, L Shi, B Qu, N Szentmáry. Aspergillus fumigatus-stimulated human corneal epithelial cells induce pyroptosis of THP-1 macrophages by secreting TSLP. Inflammation 2021; 44(2): 682–692
https://doi.org/10.1007/s10753-020-01367-x pmid: 33118609
22 JS Wolffsohn, R Arita, R Chalmers, A Djalilian, M Dogru, K Dumbleton, PK Gupta, P Karpecki, S Lazreg, H Pult, BD Sullivan, A Tomlinson, L Tong, E Villani, KC Yoon, L Jones, JP Craig. TFOS DEWS II diagnostic methodology report. Ocul Surf 2017; 15(3): 539–574
https://doi.org/10.1016/j.jtos.2017.05.001 pmid: 28736342
23 J Gao, G Morgan, D Tieu, TA Schwalb, JY Luo, LA Wheeler, ME Stern. ICAM-1 expression predisposes ocular tissues to immune-based inflammation in dry eye patients and Sjögrens syndrome-like MRL/lpr mice. Exp Eye Res 2004; 78(4): 823–835
https://doi.org/10.1016/j.exer.2003.10.024 pmid: 15037117
24 X Ma, J Zou, L He, Y Zhang. Dry eye management in a Sjögren’s syndrome mouse model by inhibition of p38-MAPK pathway. Diagn Pathol 2014; 9: 5
https://doi.org/10.1186/1746-1596-9-5 pmid: 2444398
25 Z Liu, D Chen, X Chen, F Bian, W Qin, N Gao, Y Xiao, J Li, SC Pflugfelder, DQ Li. Trehalose induces autophagy against inflammation by activating TFEB signaling pathway in human corneal epithelial cells exposed to hyperosmotic stress. Invest Ophthalmol Vis Sci 2020; 61(10): 26
https://doi.org/10.1167/iovs.61.10.26 pmid: 32785678
26 Q Zheng, Y Ren, PS Reinach, Y She, B Xiao, S Hua, J Qu, W Chen. Reactive oxygen species activated NLRP3 inflammasomes prime environment-induced murine dry eye. Exp Eye Res 2014; 125: 1–8
https://doi.org/10.1016/j.exer.2014.05.001 pmid: 24836981
27 CA Dinarello. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 2009; 27(1): 519–550
https://doi.org/10.1146/annurev.immunol.021908.132612 pmid: 19302047
28 M Christofi, Sommer S Le, C Mölzer, IP Klaska, L Kuffova, JV Forrester. Low-dose 2-deoxy glucose stabilises tolerogenic dendritic cells and generates potent in vivo immunosuppressive effects. Cell Mol Life Sci 2021; 78(6): 2857–2876
https://doi.org/10.1007/s00018-020-03672-y pmid: 33074350
29 X Zhang, VJ M, Y Qu, X He, S Ou, J Bu, C Jia, J Wang, H Wu, Z Liu, W Li. Dry eye management: targeting the ocular surface microenvironment. Int J Mol Sci 2017; 18(7): 1398
https://doi.org/10.3390/ijms18071398 pmid: 28661456
30 A Jr deRoetth. Glycolytic activity of the cornea. AMA Arch Opthalmol 1951; 45(2): 139–148
https://doi.org/10.1001/archopht.1951.01700010144002 pmid: 14799010
31 RS Thies, LJ Mandel. Role of glucose in corneal metabolism. Am J Physiol 1985; 249(5): C409–C416
https://doi.org/10.1152/ajpcell.1985.249.5.C409 pmid: 3933363
32 X Chen, J Rao, Z Zheng, Y Yu, S Lou, L Liu, Q He, L Wu, X Sun. Integrated tear proteome and metabolome reveal panels of inflammatory-related molecules via key regulatory pathways in dry eye syndrome. J Proteome Res 2019; 18(5): 2321–2330
https://doi.org/10.1021/acs.jproteome.9b00149 pmid: 30966751
33 V Pucino, M Certo, V Bulusu, D Cucchi, K Goldmann, E Pontarini, R Haas, J Smith, SE Headland, K Blighe, M Ruscica, F Humby, MJ Lewis, JJ Kamphorst, M Bombardieri, C Pitzalis, C Mauro. Lactate buildup at the site of chronic inflammation promotes disease by inducing CD4+ T cell metabolic rewiring. Cell Metab 2019; 30(6): 1055–1074.e8
https://doi.org/10.1016/j.cmet.2019.10.004 pmid: 31708446
34 S Hui, JM Ghergurovich, RJ Morscher, C Jang, X Teng, W Lu, LA Esparza, T Reya, Zhan Le, Guo J Yanxiang, E White, JD Rabinowitz. Glucose feeds the TCA cycle via circulating lactate. Nature 2017; 551(7678): 115–118
https://doi.org/10.1038/nature24057 pmid: 29045397
35 JY Zhang, B Zhou, RY Sun, YL Ai, K Cheng, FN Li, BR Wang, FJ Liu, ZH Jiang, WJ Wang, D Zhou, HZ Chen, Q Wu. The metabolite α-KG induces GSDMC-dependent pyroptosis through death receptor 6-activated caspase-8. Cell Res 2021; 31(9): 980–997
https://doi.org/10.1038/s41422-021-00506-9 pmid: 34012073
36 JP Craig, KK Nichols, EK Akpek, B Caffery, HS Dua, CK Joo, Z Liu, JD Nelson, JJ Nichols, K Tsubota, F Stapleton. TFOS DEWS II definition and classification report. Ocul Surf 2017; 15(3): 276–283
https://doi.org/10.1016/j.jtos.2017.05.008 pmid: 28736335
37 JJ López-Cano, MA González-Cela-Casamayor, V Andrés-Guerrero, R Herrero-Vanrell, JM Benítez-Del-Castillo, IT Molina-Martínez. Combined hyperosmolarity and inflammatory conditions in stressed human corneal epithelial cells and macrophages to evaluate osmoprotective agents as potential DED treatments. Exp Eye Res 2021; 211: 108723
https://doi.org/10.1016/j.exer.2021.108723 pmid: 34384756
38 H Chen, X Gan, Y Li, J Gu, Y Liu, Y Deng, X Wang, Y Hong, Y Hu, L Su, W Chi. NLRP12- and NLRC4-mediated corneal epithelial pyroptosis is driven by GSDMD cleavage accompanied by IL-33 processing in dry eye. Ocul Surf 2020; 18(4): 783–794
https://doi.org/10.1016/j.jtos.2020.07.001 pmid: 32735949
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed