Korean J Physiol Pharmacol.  2023 May;27(3):231-240. 10.4196/kjpp.2023.27.3.231.

Lyso-globotriaosylsphingosine induces endothelial dysfunction via autophagy-dependent regulation of necroptosis

Affiliations
  • 1Department of Pharmacology, Yeungnam University College of Medicine, Daegu 42415, Korea
  • 2Department of Physiology, Ewha Womans University College of Medicine, Seoul 07084, Korea

Abstract

Fabry disease is a lysosomal storage disorder characterized by the lysosomal accumulations of glycosphingolipids in a variety of cytotypes, which include endothelial cells. The disease is inherited and originates from an error in glycosphingolipid catabolism caused by insufficient α-galactosidase A activity, which causes uncontrolled progressive storage of intracellular globotriaosylceramide (Gb3) in the vasculature and extracellular accumulation of lyso-Gb3 (a deacetylated soluble form of Gb3). Necrosis can lead to inflammation, which exacerbates necrosis and creates a positive feedback loop that triggers necroinflammation. However, the role played by necroptosis, a form of programmed necrotic cell death, in the cell-to-cell inflammatory reaction between epithelial and endothelial cells is unclear. Thus, the present study was undertaken to determine whether lyso-Gb3 induces necroptosis and whether necroptosis inhibition protects endothelial dysfunction against lyso-Gb3 inflamed retinal pigment epithelial cells. We found lyso-Gb3 induced necroptosis of a retinal pigment epithelial cell line (ARPE-19) in an autophagy-dependent manner and that conditioned media (CM) from ARPE-19 cells treated with lyso-Gb3 induced the necroptosis, inflammation, and senescence of human umbilical vein endothelial cells. In addition, a pharmacological study showed CM from lyso-Gb3 treated ARPE-19 cells induced endothelial necroptosis, inflammation, and senescence were significantly inhibited by an autophagy inhibitor (3-MA) and by two necroptosis inhibitors (necrostatin and GSK-872), respectively. These results demonstrate lyso-Gb3 induces necroptosis via autophagy and suggest that lyso-Gb3 inflamed retinal pigment epithelial cells trigger endothelial dysfunction via the autophagy-dependent necroptosis pathway. This study suggests the involvement of a novel autophagy-dependent necroptosis pathway in the regulation of endothelial dysfunction in Fabry disease.

Keyword

Autophagy; Cellular senescence; Glycosphingolipids; Inflammation; Necroptosis

Figure

  • Fig. 1 Lyso-Gb3 induces autophagy in ARPE-19 cells. (A) ARPE-19 cells were exposed to the indicated concentrations of lyso-Gb3 for 24 h. Transmission electron microscopic image of an autophagosome. Scale bar, 1 μm. (B) ARPE-19 cells were pretreated with 3-MA (10 mM) for 1 h and then incubated with lyso-Gb3 (0.5 μM) for 24 h. Autophagy was confirmed by indirect immunofluorescent staining using LC3 antibodies. Nuclei were stained with DAPI. Images were captured at an original magnification of ×600. Scale bar, 50 μm. (C) ARPE-19 cells were exposed to lyso-Gb3 (0.5 μM) for 24 h, and protein expressions were assessed by immunoblotting with anti-LC3B, anti-beclin-1, and anti-tubulin antibodies. Results are representative of three independent experiments. Bar graphs show densitometric quantifications of Western blot bands. (D) Cellular morphologies were observed under a light microscope using the same conditions. (E) Cell viabilities were determined using an MTT assay under the same conditions. Results are representative of three independent experiments. 3-MA, 3-Methyladenine. **p < 0.01 vs. vehicle controls, one-way ANOVA followed by Bonferroni’s post-hoc test.

  • Fig. 2 Lyso-Gb3 induces necroptosis in an autophagy-dependent manner in ARPE-19 cells. (A) ARPE-19 cells were exposed to the indicated concentrations of lyso-Gb3 for 24 h. Protein expressions were determined by immunoblotting with anti-RIP1, anti-RIP3, anti-MLKL, and anti-tubulin antibodies. Bar graphs show densitometric quantifications of Western blot bands. (B) ARPE-19 cells were first incubated with 3-MA (10 mM) for 1 h and then incubated with lyso-Gb3 (0.5 μM) for 24 h. Protein expression was determined by immunoblotting with anti-LC3, anti-beclin-1, anti-RIP1, anti-RIP3, anti-MLKL, and anti-tubulin antibodies. Bar graphs present densitometric quantifications of Western blot bands. (C) ARPE-19 cells were first incubated with 3-MA (10 mM) for 1 h and then incubated with lyso-Gb3 (0.5 μM) for 24 h. The cell lysates were resolved on non-reducing gel and analyzed by immunoblotting with anti-MLKL antibody. (D) The mRNA expressions of RIP1, RIP3, and MLKL were determined by quantitative real time-PCR analysis in ARPE-19 cells treated with lyso-Gb3 in the presence or absence of 3-MA (10 mM). qRT-PCR analysis was performed in triplicate. (E) ELISA assay was conducted to analyze the effect of 3-MA on IL-1β, IFN-γ, and TNF-α secretion by lyso-Gb3. ARPE-19 cells were pretreated with 3-MA (10 mM) for 1 h, followed by 24-h incubation with lyso-Gb3 (0.5 μM). (F) ARPE-19 cells were first incubated with 3-MA (10 mM) for 1 h and then incubated with TNF-α (5 ng/ml), IL-1β (10 ng/ml) and IFN-γ (10 ng/ml) for 24 h. Protein expressions were determined by immunoblotting with anti-RIP1, anti-RIP3, anti-MLKL, and anti-tubulin antibodies. Bar graphs show densitometric quantifications of Western blot bands. The results represent three independent experiments. 3-MA, 3-methyladenine; TNF-α, tumor necrosis factor α; IL, interleukin; IFN-γ, interferon gamma. *p < 0.05 and **p < 0.01 vs. vehicle controls, #p < 0.05 and ##p < 0.01 vs. lyso-Gb3 treated cells and pro-inflammatory cytokine treated cells, one-way ANOVA followed by Bonferroni’s post-hoc test.

  • Fig. 3 Conditioned media of lyso-Gb3 treated ARPE-19 cells increases endothelial necroptosis and inflammation. (A) Schematic view of the experiment design. (B–D) ARPE-19 cells were pretreated with necrostatin (30 μM), GSK’872 (30 μM), or 3-MA (10 mM) for 1 h and then incubated with lyso-Gb3 (0.5 μM) for 3 h. Media were then replaced with fresh medium. After overnight (O/N) incubation, the conditioned medium (CM) was recovered and then transferred to HUVECs for further incubation for 24 h. ARPE-19 cells and HUVECs were immunoblotted using anti-RIP1, anti-RIP3, anti-MLKL, anti-LC3, anti-beclin-1, anti-VCAM-1, anti-ICAM-1, and anti-tubulin antibodies. Bar graphs show densitometric quantifications of Western blot bands. 3-MA, 3-methyladenine; HUVEC, human umbilical vein endothelial cell. *p < 0.05 and **p < 0.01 vs. vehicle controls, #p < 0.05 and ##p < 0.01 vs. lyso-Gb3 treated cells, one-way ANOVA followed by Bonferroni’s post -hoc test.

  • Fig. 4 Conditioned media from lyso-Gb3 treated ARPE-19 cells increases endothelial senescence. (A) Endothelial senescence was assessed by senescence-associated β-galactosidase (SA-β-Gal) staining in HUVECs treated with conditioned medium (CM) from lyso-Gb3 treated ARPE-19 cells. (B) The effects of necrostatin, GSK’872, or 3-MA on lyso-Gb3 treated ARPE-19 CM-induced HUVEC senescence as determined by SA-β-Gal assays. Images are representative of three independent experiments (B). Scale bar, 50 μm. Bar graphs show expressions as mean percentages of SA-β-Gal positive cells. 3-MA, 3-methyladenine; HUVEC, human umbilical vein endothelial cell. **p < 0.01 vs. vehicle control, ##p < 0.01 vs. lyso-Gb3 treated cells, one-way ANOVA followed by Bonferroni’s post-hoc test.


Reference

1. Brady RO, Gal AE, Bradley RM, Martensson E, Warshaw AL, Laster L. 1967; Enzymatic defect in Fabry's disease. Ceramidetrihexosidase deficiency. N Engl J Med. 276:1163–1167. DOI: 10.1056/NEJM196705252762101. PMID: 6023233.
2. Lücke T, Höppner W, Schmidt E, Illsinger S, Das AM. 2004; Fabry disease: reduced activities of respiratory chain enzymes with decreased levels of energy-rich phosphates in fibroblasts. Mol Genet Metab. 82:93–97. DOI: 10.1016/j.ymgme.2004.01.011. PMID: 15110329.
Article
3. Levine B, Klionsky DJ. 2004; Development by self-digestion: molecular mechanisms and biological functions of autophagy. Dev Cell. 6:463–477. DOI: 10.1016/S1534-5807(04)00099-1. PMID: 15068787.
4. Choi ME, Price DR, Ryter SW, Choi AMK. 2019; Necroptosis: a crucial pathogenic mediator of human disease. JCI Insight. 4:e128834. DOI: 10.1172/jci.insight.128834. PMID: 31391333. PMCID: PMC6693822.
Article
5. Pasparakis M, Vandenabeele P. 2015; Necroptosis and its role in inflammation. Nature. 517:311–320. DOI: 10.1038/nature14191. PMID: 25592536.
Article
6. Vitner EB, Salomon R, Farfel-Becker T, Meshcheriakova A, Ali M, Klein AD, Platt FM, Cox TM, Futerman AH. 2014; RIPK3 as a potential therapeutic target for Gaucher's disease. Nat Med. 20:204–208. DOI: 10.1038/nm.3449. PMID: 24441827.
Article
7. Rodier F, Campisi J. 2011; Four faces of cellular senescence. J Cell Biol. 192:547–556. DOI: 10.1083/jcb.201009094. PMID: 21321098. PMCID: PMC3044123.
Article
8. Nigro P, Abe J, Woo CH, Satoh K, McClain C, O'Dell MR, Lee H, Lim JH, Li JD, Heo KS, Fujiwara K, Berk BC. 2010; PKCzeta decreases eNOS protein stability via inhibitory phosphorylation of ERK5. Blood. 116:1971–1979. DOI: 10.1182/blood-2010-02-269134. PMID: 20538799. PMCID: PMC3173991.
Article
9. Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. 2009; Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo. Nat Protoc. 4:1798–1806. DOI: 10.1038/nprot.2009.191. PMID: 20010931.
Article
10. El-Awady AR, Miles B, Scisci E, Kurago ZB, Palani CD, Arce RM, Waller JL, Genco CA, Slocum C, Manning M, Schoenlein PV, Cutler CW. 2015; Porphyromonas gingivalis evasion of autophagy and intracellular killing by human myeloid dendritic cells involves DC-SIGN-TLR2 crosstalk. PLoS Pathog. 10:e1004647. DOI: 10.1371/journal.ppat.1004647. PMID: 25679217. PMCID: PMC4352937. PMID: 8c26c24211924711b92b30d02e63acb4.
Article
11. Zhang JM, An J. 2007; Cytokines, inflammation, and pain. Int Anesthesiol Clin. 45:27–37. DOI: 10.1097/AIA.0b013e318034194e. PMID: 17426506. PMCID: PMC2785020.
Article
12. Hanada T, Yoshimura A. 2002; Regulation of cytokine signaling and inflammation. Cytokine Growth Factor Rev. 13:413–421. DOI: 10.1016/S1359-6101(02)00026-6. PMID: 12220554.
Article
13. Chan AH, Schroder K. 2020; Inflammasome signaling and regulation of interleukin-1 family cytokines. J Exp Med. 217:e20190314. DOI: 10.1084/jem.20190314. PMID: 31611248. PMCID: PMC7037238.
Article
14. van Eijk M, Ferraz MJ, Boot RG, Aerts JMFG. 2020; Lyso-glycosphingolipids: presence and consequences. Essays Biochem. 64:565–578. DOI: 10.1042/EBC20190090. PMID: 32808655. PMCID: PMC7517347.
Article
15. Rozenfeld P, Feriozzi S. 2017; Contribution of inflammatory pathways to Fabry disease pathogenesis. Mol Genet Metab. 122:19–27. DOI: 10.1016/j.ymgme.2017.09.004. PMID: 28947349.
Article
16. Sanchez-Niño MD, Carpio D, Sanz AB, Ruiz-Ortega M, Mezzano S, Ortiz A. 2015; Lyso-Gb3 activates Notch1 in human podocytes. Hum Mol Genet. 24:5720–5732. DOI: 10.1093/hmg/ddv291. PMID: 26206887.
Article
17. Ferraz MJ, Kallemeijn WW, Mirzaian M, Herrera Moro D, Marques A, Wisse P, Boot RG, Willems LI, Overkleeft HS, Aerts JM. 2014; Gaucher disease and Fabry disease: new markers and insights in pathophysiology for two distinct glycosphingolipidoses. Biochim Biophys Acta. 1841:811–825. DOI: 10.1016/j.bbalip.2013.11.004. PMID: 24239767.
Article
18. Stepien KM, Roncaroli F, Turton N, Hendriksz CJ, Roberts M, Heaton RA, Hargreaves I. 2020; Mechanisms of mitochondrial dysfunction in lysosomal storage disorders: a review. J Clin Med. 9:2596. DOI: 10.3390/jcm9082596. PMID: 32796538. PMCID: PMC7463786. PMID: c0ad71572dab44ccb9a6096a74d2e4b0.
Article
19. Sanchez-Niño MD, Sanz AB, Carrasco S, Saleem MA, Mathieson PW, Valdivielso JM, Ruiz-Ortega M, Egido J, Ortiz A. 2011; Globotriaosylsphingosine actions on human glomerular podocytes: implications for Fabry nephropathy. Nephrol Dial Transplant. 26:1797–1802. DOI: 10.1093/ndt/gfq306. PMID: 20504837.
Article
20. Yang Z, Wang Y, Zhang Y, He X, Zhong CQ, Ni H, Chen X, Liang Y, Wu J, Zhao S, Zhou D, Han J. 2018; RIP3 targets pyruvate dehydrogenase complex to increase aerobic respiration in TNF-induced necroptosis. Nat Cell Biol. 20:186–197. DOI: 10.1038/s41556-017-0022-y. PMID: 29358703.
Article
21. Cougnoux A, Cluzeau C, Mitra S, Li R, Williams I, Burkert K, Xu X, Wassif CA, Zheng W, Porter FD. 2016; Necroptosis in Niemann-Pick disease, type C1: a potential therapeutic target. Cell Death Dis. 7:e2147. DOI: 10.1038/cddis.2016.16. PMID: 26986514. PMCID: PMC4823930.
Article
22. Zhu K, Liang W, Ma Z, Xu D, Cao S, Lu X, Liu N, Shan B, Qian L, Yuan J. 2018; Necroptosis promotes cell-autonomous activation of proinflammatory cytokine gene expression. Cell Death Dis. 9:500. DOI: 10.1038/s41419-018-0524-y. PMID: 29703889. PMCID: PMC5923285.
Article
23. Zu R, Yu Z, Zhao J, Lu X, Liang W, Sun L, Si C, Zhu K, Zhang T, Li G, Zhang M, Zhang Y, Liu N, Yuan J, Shan B. 2021; Quantitative analysis of phosphoproteome in necroptosis reveals a role of TRIM28 phosphorylation in promoting necroptosis-induced cytokine production. Cell Death Dis. 12:994. DOI: 10.1038/s41419-021-04290-7. PMID: 34689152. PMCID: PMC8542044. PMID: 70ad286ccd1f4e56af15db5ed08e7b33.
Article
24. Kaczmarek A, Vandenabeele P, Krysko DV. 2013; Necroptosis: the release of damage-associated molecular patterns and its physiological relevance. Immunity. 38:209–223. DOI: 10.1016/j.immuni.2013.02.003. PMID: 23438821.
Article
25. Jung S, Seo DJ, Yeo D, Wang Z, Min A, Zhao Z, Song M, Choi IS, Myoung J, Choi C. 2020; Experimental infection of hepatitis E virus induces pancreatic necroptosis in miniature pigs. Sci Rep. 10:12022. DOI: 10.1038/s41598-020-68959-3. PMID: 32694702. PMCID: PMC7374588.
Article
26. Frank D, Vince JE. 2019; Pyroptosis versus necroptosis: similarities, differences, and crosstalk. Cell Death Differ. 26:99–114. DOI: 10.1038/s41418-018-0212-6. PMID: 30341423. PMCID: PMC6294779.
Article
27. Godo S, Shimokawa H. 2017; Endothelial functions. Arterioscler Thromb Vasc Biol. 37:e108–e114. DOI: 10.1161/ATVBAHA.117.309813. PMID: 28835487.
Article
28. Rodgers JL, Jones J, Bolleddu SI, Vanthenapalli S, Rodgers LE, Shah K, Karia K, Panguluri SK. 2019; Cardiovascular risks associated with gender and aging. J Cardiovasc Dev Dis. 6:19. DOI: 10.3390/jcdd6020019. PMID: 31035613. PMCID: PMC6616540. PMID: 35945fd9b2c54bb092e36026be981167.
Article
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr