Vitamin D3 Protects against Diabetic Retinopathy by Inhibiting High-Glucose-Induced Activation of the ROS/TXNIP/NLRP3 Inflammasome Pathway

Li Lu, Qianyi Lu, Wei Chen, Jingwen Li, Chunxia Li, Zhi Zheng, Li Lu, Qianyi Lu, Wei Chen, Jingwen Li, Chunxia Li, Zhi Zheng

Abstract

Purpose: This study aimed to evaluate the mechanisms underlying the effects of 1,25-dihydroxyvitamin D (vitamin D3) on diabetes-induced retinal vascular damage and retinal vascular endothelial cell apoptosis.

Methods: Diabetic and control rats were randomly assigned to receive vitamin D3 or vehicle for 6 months. Additionally, human retinal microvascular endothelial cells (HRMECs) were incubated in normal or high-glucose medium with or without vitamin D3. Morphological changes in retinal tissues and retinal vascular permeability were examined, and cellular apoptosis was detected by fluorescence staining. Intracellular reactive oxygen species (ROS) levels were determined using fluorescent probes. Proteins were examined by Western blotting.

Results: Vitamin D3 significantly downregulated intracellular ROS and inhibited TRX-interacting protein (TXNIP)/NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome pathway activation. Additionally, vitamin D3 reduced vascular endothelial growth factor (VEGF) expression and the Bax/Bcl-2 ratio. These changes were associated with retinal recovery and with decreases in retinal vascular permeability and retinal capillary cell apoptosis.

Conclusions: Vitamin D3 decreases diabetes-induced ROS and exerts protective effects against retinal vascular damage and cell apoptosis in association with inhibition of the ROS/TXNIP/NLRP3 inflammasome pathway. Understanding the mechanisms of action of vitamin D3 has important implications for preventing and treating inflammatory-related illnesses such as diabetic retinopathy.

Figures

Figure 1
Figure 1
Vitamin D3 inhibits the high-glucose-induced death of HRMECs by inhibiting expression of proteins in the NLRP3 inflammasome pathway. (a) Western blot analysis of proteins in the NLRP3 inflammasome pathway in the NG, NG + VD, HG, HG + VD, and NG + NLRP3-overexpressing and HG + NLRP3-knockdown groups. (b) Quantitative analysis of the bands. (c) Transfection of HRMECs with plasmid DNA (CTR: control group; scramble: scrambled shRNA control group; vector: empty vector control group). (d) Co-staining of Hoechst33238 and propidium iodide was performed to assess apoptosis of HRMECs. (e) Quantification of apoptotic cells. The results are expressed as the mean percentage of apoptotic cells ± SD. n = 6 (∗∗: NG versus HG, P < 0.05; ##: HG versus HG + VD, P < 0.05; ∗: NG + NLRP3 Op versus NG, P < 0.05; #: HG + NLRP3 KD versus HG, P < 0.05).
Figure 2
Figure 2
(a) Representative images of retinas at month 6. (b-c) The retinal thickness and cell numbers in the GCL (cells per 100 μm). At month 6, the GCL and IPL were obvious and well organized in the NC group. The retinal thickness in diabetic rats was reduced and the number of GCL cells decreased (DM), whereas in diabetic rats treated with vitamin D3, the retinas were more normal in structure and thicker than those in diabetic rats (NC: normal control group; NC + VD: normal control group + vitamin D3; DM: diabetes mellitus group; IPL: inner plexiform layer; GCL: ganglion cell layer). Data represent mean ± SD (n = 10). ∗∗DM versus NC, P < 0.05; ##DM + VD versus DM, P < 0.05. Data are expressed as the mean ± standard deviation. Scale bar, 30 μm.
Figure 3
Figure 3
Protective effect of vitamin D3 against apoptosis of retinal cells at month 6. (a) No TUNEL-positive cells were seen in the NC or NC + VD groups; abundant red fluorescent nuclei were observed in the DM group. Sparse red fluorescent nuclei were found in the DM + VD group. GCL: ganglion cell layer; INL: inner nuclear layer; ONL: outer nuclear layer. (b) Western blot of retinal extracts using anti-Bcl-2 and anti-Bax antibodies; β-tubulin was used as the loading control. (c) Bax/Bcl-2 ratios (data represent mean ± SD (n = 10) ∗∗: NC versus DM, P < 0.05, ##: DM versus DM + VD, P < 0.05. NC: normal control group; NC + VD: normal control group + vitamin D3; DM: diabetes mellitus group; DM + VD: diabetes mellitus group + vitamin D3. Scale bar, 30 μm.
Figure 4
Figure 4
Vitamin D3 attenuates the elevated BRB breakdown in diabetic rats by inhibiting VEGF expression and the TXNIP/NLRP3 pathway. (a) Retinal vascular permeability was quantified by detecting the retinal Evans blue leakage (data are the means ± SD, n = 10∗∗: DM versus NC, P < 0.05 and ##: DM + VD versus DM, P < 0.05). (b, c) Retinal VEGF protein expression was inhibited by vitamin D3 (data are the means ± SD, n = 10∗∗: DM versus NC, P < 0.05 and ##: DM + VD versus DM, P < 0.05). (d, e) Quantitative Western blot analyses of TXNIP and the NLRP3 inflammasome in whole retinal extracts. Expression was normalized to that of β-tubulin. Data are the means ± SD, n = 10 (∗∗: NC versus DM, P < 0.05; ##: DM + VD versus DM, P < 0.05).
Figure 5
Figure 5
Vitamin D3 inhibits glucose-induced ROS production in HRMECs. (a) To measure ROS production, cells were labelled with H2DCF-DA. The figures show representative data from three independent experiments. (b) Quantitative analysis of the measured fluorescence intensity was performed. Values are the means ± SD of three independent experiments (n = 6, ∗∗: HG versus NG, P < 0.05, ##: HG + VD and HG + NAC versus HG, P < 0.05). (c) Western blot and quantitative analyses of TXNIP from whole retinal extracts. Expression was normalized to that of β-tubulin. Data are the means ± SD, n = 6 (∗∗: HG versus NG, P < 0.05, ##: HG + VD and HG + NAC versus HG, P < 0.05).

References

    1. Wild S., Roglic G., Green A., Sicree R., King H. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care. 2004;27(5):1047–1053. doi: 10.2337/diacare.27.5.1047.
    1. Yau J. W. Y., Rogers S. L., Kawasaki R., et al. Global prevalence and major risk factors of diabetic retinopathy. Diabetes Care. 2012;35(3):556–564. doi: 10.2337/dc11-1909.
    1. Plum L. A., DeLuca H. F. Vitamin D, disease and therapeutic opportunities. Nature Reviews Drug Discovery. 2010;9(12):941–955. doi: 10.1038/nrd3318.
    1. Prietl B., Treiber G., Pieber T. R., Amrein K. Vitamin D and immune function. Nutrients. 2013;5(7):2502–2521. doi: 10.3390/nu5072502.
    1. Chagas C. E. A., Borges M. C., Martini L. A., Rogero M. M. Focus on vitamin D, inflammation and type 2 diabetes. Nutrients. 2012;4(12):52–67. doi: 10.3390/nu4010052.
    1. Guillot X., Semerano L., Saidenberg-Kermanac’h N., Falgarone G., Boissier M.-C. Vitamin D and inflammation. Joint, Bone, Spine. 2010;77(6):552–557. doi: 10.1016/j.jbspin.2010.09.018.
    1. Tarcin O., Yavuz D. G., Ozben B., et al. Effect of vitamin D deficiency and replacement on endothelial function in asymptomatic subjects. The Journal of Clinical Endocrinology & Metabolism. 2009;94(10):4023–4030. doi: 10.1210/jc.2008-1212.
    1. Aksoy H., Akçay F., Kurtul N., Baykal O., Avci B. Serum 1,25 dihydroxy vitamin D (1,25(OH)2D3), 25 hydroxy vitamin D (25(OH)D) and parathormone levels in diabetic retinopathy. Clinical Biochemistry. 2000;33(1):47–51. doi: 10.1016/S0009-9120(99)00085-5.
    1. Kaur H., Donaghue K. C., Chan A. K., et al. Vitamin D deficiency is associated with retinopathy in children and adolescents with type 1 diabetes. Diabetes Care. 2011;34(6):1400–1402. doi: 10.2337/dc11-0103.
    1. Patrick P. A., Visintainer P. F., Shi Q., Weiss I. A., Brand D. A. Vitamin D and retinopathy in adults with diabetes mellitus. Archives of Ophthalmology. 2012;130(6):756–760. doi: 10.1001/archophthalmol.2011.2749.
    1. Longo-Mbenza B., Mvitu Muaka M., Masamba W., et al. Retinopathy in non diabetics, diabetic retinopathy and oxidative stress: a new phenotype in Central Africa? International Journal of Ophthalmology. 2014;7(2):293–301. doi: 10.3980/j.issn.2222-3959.2014.02.18.
    1. Shimo N., Yasuda T., Kaneto H., et al. Vitamin D deficiency is significantly associated with retinopathy in young Japanese type 1 diabetic patients. Diabetes Research & Clinical Practice. 2014;106(2):e41–e43. doi: 10.1016/j.diabres.2014.08.005.
    1. Taverna M. J., Selam J.-L., Slama G. Association between a protein polymorphism in the start codon of the vitamin D receptor gene and severe diabetic retinopathy in C-peptide-negative type 1 diabetes. The Journal of Clinical Endocrinology & Metabolism. 2005;90(8):4803–4808. doi: 10.1210/jc.2004-2407.
    1. van Etten E., Verlinden L., Giulietti A., et al. The vitamin D receptor gene FokI polymorphism: functional impact on the immune system. European Journal of Immunology. 2007;37(2):395–405. doi: 10.1002/eji.200636043.
    1. Taverna M. J., Sola A., Guyot-Argenton C., et al. Taq I polymorphism of the vitamin D receptor and risk of severe diabetic retinopathy. Diabetologia. 2002;45(3):436–442. doi: 10.1007/s00125-001-0769-2.
    1. Zhong X., Du Y., Lei Y., Liu N., Guo Y., Pan T. Effects of vitamin D receptor gene polymorphism and clinical characteristics on risk of diabetic retinopathy in Han Chinese type 2 diabetes patients. Gene. 2015;566(2):212–216. doi: 10.1016/j.gene.2015.04.045.
    1. Payne J., Ray R., Watson D., et al. Vitamin D insufficiency in diabetic retinopathy. Endocrine Practice. 2012;18(2):185–193. doi: 10.4158/EP11147.OR.
    1. Ren Z., Li W., Zhao Q., Ma L., Zhu J. The impact of 1, 25-dihydroxy vitamin D3 on the expressions of vascular endothelial growth factor and transforming growth factor-β1 in the retinas of rats with diabetes. Diabetes Research and Clinical Practice. 2012;98(3):474–480. doi: 10.1016/j.diabres.2012.09.028.
    1. Adamis A. P. Is diabetic retinopathy an inflammatory disease? British Journal of Ophthalmology. 2002;86(4):363–365. doi: 10.1136/bjo.86.4.363.
    1. Kern T. S. Contributions of inflammatory processes to the development of the early stages of diabetic retinopathy. Experimental Diabetes Research. 2007;2007:14. doi: 10.1155/2007/95103.95103
    1. Tang J., Kern T. S. Inflammation in diabetic retinopathy. Progress in Retinal and Eye Research. 2011;30(5):343–358. doi: 10.1016/j.preteyeres.2011.05.002.
    1. Kocak N., Alacacioglu I., Kaynak S., et al. Comparison of vitreous and plasma levels of vascular endothelial growth factor, interleukin-6 and hepatocyte growth factor in diabetic and non-diabetic retinal detachment cases. Annals of Ophthalmology. 2010;42:10–14.
    1. Vincent J. A., Mohr S. Inhibition of caspase-1/interleukin-1β signaling prevents degeneration of retinal capillaries in diabetes and galactosemia. Diabetes. 2007;56(1):224–230. doi: 10.2337/db06-0427.
    1. Behl Y., Krothapalli P., Desta T., DiPiazza A., Roy S., Graves D. T. Diabetes-enhanced tumor necrosis factor-α production promotes apoptosis and the loss of retinal microvascular cells in type 1 and type 2 models of diabetic retinopathy. The American Journal of Pathology. 2008;172(5):1411–1418. doi: 10.2353/ajpath.2008.071070.
    1. Ting J. P.-Y., Willingham S. B., Bergstralh D. T. NLRs at the intersection of cell death and immunity. Nature Reviews Immunology. 2008;8(5):372–379. doi: 10.1038/nri2296.
    1. Schroder K., Tschopp J. The inflammasomes. Cell. 2010;140(6):821–832. doi: 10.1016/j.cell.2010.01.040.
    1. Li P., Xu X., Zheng Z., Zhu B., Shi Y., Liu K. Protective effects of rosiglitazone on retinal neuronal damage in diabetic rats. Current Eye Research. 2011;36(7):673–679. doi: 10.3109/02713683.2011.572220.
    1. Leal E. C., Martins J., Voabil P., et al. Calcium dobesilate inhibits the alterations in tight junction proteins and leukocyte adhesion to retinal endothelial cells induced by diabetes. Diabetes. 2010;59(10):2637–2645. doi: 10.2337/db09-1421.
    1. Qaum T., Xu Q., Joussen A. M., et al. VEGF-initiated blood-retinal barrier breakdown in early diabetes. Investigative Ophthalmology & Visual Science. 2001;42(10):2408–2413.
    1. Hewison M. Vitamin D and the immune system: new perspectives on an old theme. Rheumatic Diseases Clinics of North America. 2012;38(1):125–139. doi: 10.1016/j.rdc.2012.03.012.
    1. Gao X.-Y., Kuang H.-Y., Zou W., Liu X. M., Lin H. B., Yang Y. The timing of re-institution of good blood glucose control affects apoptosis and expression of Bax and Bcl-2 in the retina of diabetic rats. Molecular Biology Reports. 2009;36(7):1977–1982. doi: 10.1007/s11033-008-9407-0.
    1. Kowluru R. A., Mohammad G., Santos J. M., Tewari S., Zhong Q. Interleukin-1β and mitochondria damage, and the development of diabetic retinopathy. Journal of Ocular Biology, Diseases, and Informatics. 2011;4(1-2):3–9. doi: 10.1007/s12177-011-9074-6.
    1. Podestà F., Romeo G., Liu W.-H., et al. Bax is increased in the retina of diabetic subjects and is associated with pericyte apoptosis in vivo and in vitro. The American Journal of Pathology. 2000;156(3):1025–1032. doi: 10.1016/S0002-9440(10)64970-X.
    1. Devi T. S., Lee I., Hüttemann M., Kumar A., Nantwi K. D., Singh L. P. TXNIP links innate host defense mechanisms to oxidative stress and inflammation in retinal Muller glia under chronic hyperglycemia: implications for diabetic retinopathy. Experimental Diabetes Research. 2012;2012:19. doi: 10.1155/2012/438238.438238
    1. Zheng Z., Chen H., Ke G., et al. Protective effect of perindopril on diabetic retinopathy is associated with decreased vascular endothelial growth factor–to–pigment epithelium–derived factor ratio involvement of a mitochondria–reactive oxygen species pathway. Diabetes. 2009;58(4):954–964. doi: 10.2337/db07-1524.
    1. Zheng Z., Chen H., Wang H., et al. Improvement of retinal vascular injury in diabetic rats by statins is associated with the inhibition of mitochondrial reactive oxygen species pathway mediated by peroxisome proliferator–activated receptor γ coactivator 1α. Diabetes. 2010;59(9):2315–2325. doi: 10.2337/db10-0638.
    1. Forbes J. M., Coughlan M. T., Cooper M. E. Oxidative stress as a major culprit in kidney disease in diabetes. Diabetes. 2008;57(6):1446–1454. doi: 10.2337/db08-0057.
    1. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001;414(6865):813–820. doi: 10.1038/414813a.
    1. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes. 2005;54(6):1615–1625. doi: 10.2337/diabetes.54.6.1615.
    1. Tschopp J., Schroder K. NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production? Nature Reviews Immunology. 2010;10(3):210–215. doi: 10.1038/nri2725.
    1. Lawlor K. E., Vince J. E. Ambiguities in NLRP3 inflammasome regulation: is there a role for mitochondria? Biochimica et Biophysica Acta (BBA)-General Subjects. 2014;1840(4):1433–1440. doi: 10.1016/j.bbagen.2013.08.014.
    1. Schroder K., Zhou R., Tschopp J. The NLRP3 inflammasome: a sensor for metabolic danger? Science. 2010;327(5963):296–300. doi: 10.1126/science.1184003.

Source: PubMed

3
Sottoscrivi