Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Research Article

Integrated Network Pharmacology and Cellular Assay to Explore the Mechanisms of Selenized Tripterine Phytosomes (Se@Tri-PTs) Alleviating Podocyte Injury in Diabetic Nephropathy

Author(s): Shiping Zhu, Qiubo Liu, Yuling Chang, Chunhua Luo, Xingwang Zhang* and Shengyun Sun*

Volume 29, Issue 38, 2023

Published on: 13 November, 2023

Page: [3073 - 3086] Pages: 14

DOI: 10.2174/0113816128275079231102071508

Price: $65

conference banner
Abstract

Aim: This work aimed to elucidate the mechanisms of Se@Tri-PTs in alleviating podocyte injury via network pharmacology and in vitro cellular assay.

Background: Selenized tripterine phytosomes (Se@Tri-PTs) have been confirmed to undertake synergistic and sensitized effects on inflammation, which may be curatively promising for diabetic nephropathy (DN). However, the mechanisms of Se@Tri-PTs in alleviating podocyte injury, a major contributor to DN, still remain unclear.

Objective: The objective of the study was to find out the underlying mechanisms of Se@Tri-PTs in alleviating podocyte injury in diabetic nephropathy.

Methods: The key components and targets of Tripterygium wilfordii (TW) significant for DN as well as the signaling pathways involved have been identified. A high glucose-induced podocyte injury model was established and verified by western blot. The protective concentration of Se@Tri-PTs was screened by CCK-8 assay. Podocytes cultured with high glucose were treated with Se@Tri-PTs under protective levels. The expression of key protective proteins, nephrin and desmin, in podocytes, was assayed by western blot. Further, autophagy- related proteins and factors, like NLRP3, Beclin-1, LC3II/LC3, P62, and SIRT1, were analyzed, which was followed by apoptosis detection.

Results: Network pharmacology revealed that several monomeric components of TW, especially Tri, act on DN through multiple targets and pathways, including the NLRP3-mediated inflammatory pathway. Se@Tri-PTs improved the viability of podocytes and alleviated their injury induced by high glucose at 5 μg/L or above. High-glucose induction promoted the expression of NLRP3 in podocytes, while a low concentration of Se@Tri-PTs suppressed the expression. A long-term exposure of high glucose significantly inhibited the autophagic activity of podocytes, as manifested by decreased Beclin-1 level, lower ratio of LC3 II/LC3 I, and up- regulation of P62. This abnormality was efficiently reversed by Se@Tri-PTs. Importantly, the expression of SIRT1 was up-regulated and podocyte apoptosis was reduced.

Conclusion: Se@Tri-PTs can alleviate podocyte injury associated with DN by modulating NLRP3 expression through the pathway of SIRT1-mediated autophagy.

Keywords: Tripterine, phytosomes, network pharmacology, diabetic nephropathy, podocyte injury, autophagy.

« Previous
[1]
Guan Y, Davis L, Breyer MD, Hao CM. Cyclooxygenase-2 contributes to diabetic nephropathy through glomerular EP4 receptor. Prostaglandins Other Lipid Mediat 2022; 159: 106621.
[http://dx.doi.org/10.1016/j.prostaglandins.2022.106621] [PMID: 35131551]
[2]
Perkovic V, Craig JC, Chailimpamontree W, et al. Action plan for optimizing the design of clinical trials in chronic kidney disease. Kidney Int Suppl 2017; 7(2): 138-44.
[http://dx.doi.org/10.1016/j.kisu.2017.07.009] [PMID: 30675428]
[3]
Yuan Z, Pan Y, Leng T, et al. Progress and prospects of research ideas and methods in the network pharmacology of traditional Chinese medicine. J Pharm Pharm Sci 2022; 25: 218-26.
[http://dx.doi.org/10.18433/jpps32911] [PMID: 35760072]
[4]
Ding Y, Fu X, Wang Q, Liu H, Wang H, Wu D. The complex interplay between autophagy and NLRP3 inflammasome in renal diseases. Int J Mol Sci 2021; 22(23): 12766.
[http://dx.doi.org/10.3390/ijms222312766] [PMID: 34884572]
[5]
Litwiniuk A, Bik W, Kalisz M, Baranowska-Bik A. Inflammasome NLRP3 potentially links obesity-associated low-grade systemic inflammation and insulin resistance with Alzheimer’s disease. Int J Mol Sci 2021; 22(11): 5603.
[http://dx.doi.org/10.3390/ijms22115603] [PMID: 34070553]
[6]
Hong YA, Kim JE, Jo M, Ko GJ. The role of Sirtuins in kidney diseases. Int J Mol Sci 2020; 21(18): 6686.
[http://dx.doi.org/10.3390/ijms21186686] [PMID: 32932720]
[7]
Wang F, Li R, Zhao L, Ma S, Qin G. Resveratrol ameliorates renal damage by inhibiting oxidative stress-mediated apoptosis of podocytes in diabetic nephropathy. Eur J Pharmacol 2020; 885: 173387.
[http://dx.doi.org/10.1016/j.ejphar.2020.173387] [PMID: 32710953]
[8]
Lv H, Jiang L, Zhu M, et al. The genus Tripterygium: A phytochemistry and pharmacological review. Fitoterapia 2019; 137: 104190.
[http://dx.doi.org/10.1016/j.fitote.2019.104190] [PMID: 31163199]
[9]
Liu S, Chen Q, Yan L, et al. Phytosomal tripterine with selenium modification attenuates the cytotoxicity and restrains the inflammatory evolution via inhibiting NLRP3 inflammasome activation and pyroptosis. Int Immunopharmacol 2022; 108: 108871.
[http://dx.doi.org/10.1016/j.intimp.2022.108871] [PMID: 35605435]
[10]
Zhang X, Zhang T, Zhou X, et al. Enhancement of oral bioavailability of tripterine through lipid nanospheres: Preparation, characterization, and absorption evaluation. J Pharm Sci 2014; 103(6): 1711-9.
[http://dx.doi.org/10.1002/jps.23967] [PMID: 24700417]
[11]
Zhao J, Chen X, Ho KH, et al. Nanotechnology for diagnosis and therapy of rheumatoid arthritis: Evolution towards theranostic approaches. Chin Chem Lett 2021; 32(1): 66-86.
[http://dx.doi.org/10.1016/j.cclet.2020.11.048]
[12]
Cui Y, Zhu T, Zhang X, et al. Oral delivery of superoxide dismutase by lipid polymer hybrid nanoparticles for the treatment of ulcerative colitis. Chin Chem Lett 2022; 33(10): 4617-22.
[http://dx.doi.org/10.1016/j.cclet.2022.03.077]
[13]
Guan B, Yan R, Li R, Zhang X. Selenium as a pleiotropic agent for medical discovery and drug delivery. Int J Nanomedicine 2018; 13: 7473-90.
[http://dx.doi.org/10.2147/IJN.S181343] [PMID: 30532534]
[14]
Sagoo MK, Gnudi L. Diabetic nephropathy: An overview. Methods Mol Biol 2020; 2067: 3-7.
[http://dx.doi.org/10.1007/978-1-4939-9841-8_1] [PMID: 31701441]
[15]
Yu X, Meng X, Xu M, et al. Celastrol ameliorates cisplatin nephrotoxicity by inhibiting NF-κB and improving mitochondrial function. EBioMedicine 2018; 36: 266-80.
[http://dx.doi.org/10.1016/j.ebiom.2018.09.031] [PMID: 30268831]
[16]
Zhang M, Chen Y, Yang M, et al. Celastrol attenuates renal injury in diabetic rats via MAPK/NF-κB pathway. Phytother Res 2019; 33(4): 1191-8.
[http://dx.doi.org/10.1002/ptr.6314] [PMID: 30768745]
[17]
Zhu S, Luo C, Feng W, et al. Selenium-deposited tripterine phytosomes ameliorate the antiarthritic efficacy of the phytomedicine via a synergistic sensitization. Int J Pharm 2020; 578: 119104.
[http://dx.doi.org/10.1016/j.ijpharm.2020.119104] [PMID: 32018017]
[18]
Guo Z, Shi L, Feng H, et al. Reduction-sensitive nanomicelles: Delivery celastrol for retinoblastoma cells effective apoptosis. Chin Chem Lett 2021; 32(3): 1046-50.
[http://dx.doi.org/10.1016/j.cclet.2020.03.066]
[19]
Gil CL, Hooker E, Larrivée B. Diabetic kidney disease, endothelial damage, and podocyte-endothelial crosstalk. Kidney Med 2021; 3(1): 105-15.
[http://dx.doi.org/10.1016/j.xkme.2020.10.005] [PMID: 33604542]
[20]
Reiser J, Sever S. Podocyte biology and pathogenesis of kidney disease. Annu Rev Med 2013; 64(1): 357-66.
[http://dx.doi.org/10.1146/annurev-med-050311-163340] [PMID: 23190150]
[21]
Zhu M, Zhu S, Liu Q, Ren Y, Ma Z, Zhang X. Selenized liposomes with ameliorative stability that achieve sustained release of emodin but fail in bioavailability. Chin Chem Lett 2023; 34(1): 107482.
[http://dx.doi.org/10.1016/j.cclet.2022.04.080]
[22]
Clark AR, Marshall J, Zhou Y, et al. Single-cell transcriptomics reveal disrupted kidney filter cell-cell interactions after early and selective podocyte injury. Am J Pathol 2022; 192(2): 281-94.
[http://dx.doi.org/10.1016/j.ajpath.2021.11.004] [PMID: 34861215]
[23]
Sun Q, Fan J, Billiar TR, Scott MJ. Inflammasome and autophagy regulation - A two-way street. Mol Med 2017; 23(1): 188-95.
[http://dx.doi.org/10.2119/molmed.2017.00077] [PMID: 28741645]
[24]
Liu W, Huang G, Rui H, et al. Course monitoring of membranous nephropathy: Both autoantibodies and podocytes require multidimensional attention. Autoimmun Rev 2022; 21(2): 102976.
[http://dx.doi.org/10.1016/j.autrev.2021.102976] [PMID: 34757091]
[25]
Tortora SC, Bodiwala VM, Quinn A, Martello LA, Vignesh S. Microbiome and colorectal carcinogenesis: Linked mechanisms and racial differences. World J Gastrointest Oncol 2022; 14(2): 375-95.
[http://dx.doi.org/10.4251/wjgo.v14.i2.375] [PMID: 35317317]
[26]
Li KX, Ji MJ, Sun HJ. An updated pharmacological insight of resveratrol in the treatment of diabetic nephropathy. Gene 2021; 780: 145532.
[http://dx.doi.org/10.1016/j.gene.2021.145532] [PMID: 33631244]
[27]
Teh YM, Mualif SA, Lim SK. A comprehensive insight into autophagy and its potential signaling pathways as a therapeutic target in podocyte injury. Int J Biochem Cell Biol 2022; 143: 106153.
[http://dx.doi.org/10.1016/j.biocel.2021.106153] [PMID: 34974186]
[28]
Liang Y, Liu H, Zhu J, et al. Inhibition of p53/miR-34a/SIRT1 axis ameliorates podocyte injury in diabetic nephropathy. Biochem Biophys Res Commun 2021; 559: 48-55.
[http://dx.doi.org/10.1016/j.bbrc.2021.04.025] [PMID: 33932899]
[29]
Liu WJ, Ye L, Huang WF, et al. p62 links the autophagy pathway and the ubiqutin–proteasome system upon ubiquitinated protein degradation. Cell Mol Biol Lett 2016; 21(1): 29.
[http://dx.doi.org/10.1186/s11658-016-0031-z] [PMID: 28536631]
[30]
Yang G, Li Q, Peng J, et al. Fucoxanthin regulates Nrf2 signaling to decrease oxidative stress and improves renal fibrosis depending on Sirt1 in HG-induced GMCs and STZ-induced diabetic rats. Eur J Pharmacol 2021; 913: 174629.
[http://dx.doi.org/10.1016/j.ejphar.2021.174629] [PMID: 34780751]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy