The Protective Effect of Zebularine, an Inhibitor of DNA Methyltransferase, on Renal Tubulointerstitial Inflammation and Fibrosis
Abstract
:1. Introduction
2. Results
2.1. Expression of DNMT in Obstructed Kidneys
2.2. Effects of DNMT Inhibition on Fibrotic Changes in Obstructed Kidneys
2.3. Effects of DNMT Inhibition on Epithelial-Mesenchymal Transition (EMT) in Obstructed Kidneys
2.4. Inhibition of DNMT Attenuates Inflammation in Obstructed Kidneys
2.5. Inhibition of DNMT Decreases Oxidative Stress and Increases Antioxidant Enzymes in Obstructed Kidneys
2.6. Effects of DNMT Inhibition on Apoptosis in Obstructed Kidneys
3. Discussion
4. Materials and Methods
4.1. Animal Experiment
4.2. Histology and Immunohistochemistry (IHC)
4.3. Renal Collagen Content Assay
4.4. Immunoblot Analysis
4.5. Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR)
4.6. Statistical Analysis
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Bikbov, B.; Purcell, C.A.; Levey, A.S.; Smith, M.; Abdoli, A.; Abebe, M.; Adebayo, O.; Afarideh, M.; Agarwal, S.; Agudelo-Botero, M.; et al. Global, regional, and national burden of chronic kidney disease, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2020, 395, 709–733. [Google Scholar] [CrossRef] [Green Version]
- Decleves, A.E.; Sharma, K. Novel targets of antifibrotic and anti-inflammatory treatment in CKD. Nat. Rev. Nephrol. 2014, 10, 257–267. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.Y.; Kim, S.I.; Choi, M.E. Therapeutic targets for treating fibrotic kidney diseases. Transl. Res. 2015, 165, 512–530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tampe, D.; Zeisberg, M. Potential approaches to reverse or repair renal fibrosis. Nat. Rev. Nephrol. 2014, 10, 226–237. [Google Scholar] [CrossRef]
- Reddy, M.A.; Natarajan, R. Recent developments in epigenetics of acute and chronic kidney diseases. Kidney Int. 2015, 88, 250–261. [Google Scholar] [CrossRef]
- Skvortsova, K.; Iovino, N.; Bogdanović, O. Functions and mechanisms of epigenetic inheritance in animals. Nat. Rev. Mol. Cell. Biol. 2018, 19, 774–790. [Google Scholar] [CrossRef] [Green Version]
- Pan, Y.; Liu, G.; Zhou, F.; Su, B.; Li, Y. DNA methylation profiles in cancer diagnosis and therapeutics. Clin. Exp. Med. 2018, 18, 1–14. [Google Scholar] [CrossRef]
- Jones, P.A. Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 2012, 13, 484–492. [Google Scholar] [CrossRef]
- Marquez, V.E.; Barchi, J.J.; Kelley, J.A.; Rao, K.V.; Agbaria, R.; Ben-Kasus, T.; Cheng, J.C.; Yoo, C.B.; Jones, P.A. Zebularine: A unique molecule for an epigenetically based strategy in cancer chemotherapy. The magic of its chemistry and biology. Nucleosides Nucleotides Nucleic Acids 2005, 24, 305–318. [Google Scholar] [CrossRef]
- Zhou, L.; Cheng, X.; Connolly, B.A.; Dickman, M.J.; Hurd, P.J.; Hornby, D.P. Zebularine: A novel DNA methylation inhibitor that forms a covalent complex with DNA methyltransferases. J. Mol. Biol. 2002, 321, 591–599. [Google Scholar] [CrossRef]
- Cheng, J.C.; Yoo, C.B.; Weisenberger, D.J.; Chuang, J.; Wozniak, C.; Liang, G.; Marquez, V.E.; Greer, S.; Orntoft, T.F.; Thykjaer, T.; et al. Preferential response of cancer cells to zebularine. Cancer Cell 2004, 6, 151–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cheng, J.C.; Matsen, C.B.; Gonzales, F.A.; Ye, W.; Greer, S.; Marquez, V.E.; Jones, P.A.; Selker, E.U. Inhibition of DNA methylation and reactivation of silenced genes by zebularine. J. Natl. Cancer Inst. 2003, 95, 399–409. [Google Scholar] [CrossRef] [PubMed]
- Yoo, C.B.; Chuang, J.C.; Byun, H.-M.; Egger, G.; Yang, A.S.; Dubeau, L.; Long, T.; Laird, P.W.; Marquez, V.E.; Jones, P.A. Long-term epigenetic therapy with oral zebularine has minimal side effects and prevents intestinal tumors in mice. Cancer Prev. Res. 2008, 1, 233–240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aranyi, T.; Susztak, K. Cytosine methylation studies in patients with diabetic kidney disease. Curr. Diab. Rep. 2019, 19, 91. [Google Scholar] [CrossRef]
- Sheng, X.; Qiu, C.; Liu, H.; Gluck, C.; Hsu, J.Y.; He, J.; Hsu, C.-Y.; Sha, D.; Weir, M.R.; Isakova, T.; et al. Systematic integrated analysis of genetic and epigenetic variation in diabetic kidney disease. Proc. Natl. Acad. Sci. USA 2020, 117, 29013–29024. [Google Scholar] [CrossRef]
- Zhang, L.; Zhang, Q.; Liu, S.; Chen, Y.; Li, R.; Lin, T.; Yu, C.; Zhang, H.; Huang, Z.; Zhao, X.; et al. DNA methyltransferase 1 may be a therapy target for attenuating diabetic nephropathy and podocyte injury. Kidney Int. 2017, 92, 140–153. [Google Scholar] [CrossRef]
- Bechtel, W.; McGoohan, S.; Zeisberg, E.M.; Müller, G.A.; Kalbacher, H.; Salant, D.J.; Müller, C.A.; Kalluri, R.; Zeisberg, M. Methylation determines fibroblast activation and fibrogenesis in the kidney. Nat. Med. 2010, 16, 544–550. [Google Scholar] [CrossRef] [Green Version]
- Ruiz, S.; Pergola, P.E.; Zager, R.A.; Vaziri, N.D. Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease. Kidney Int. 2013, 83, 1029–1041. [Google Scholar] [CrossRef] [Green Version]
- Ucero, A.C.; Benito-Martin, A.; Izquierdo, M.C.; Sanchez-Niño, M.D.; Sanz, A.B.; Ramos, A.M.; Berzal, S.; Ruiz-Ortega, M.; Egido, J.; Ortiz, A. Unilateral ureteral obstruction: Beyond obstruction. Int. Urol. Nephrol. 2014, 46, 765–776. [Google Scholar] [CrossRef]
- Cardenas, H.; Vieth, E.; Lee, J.; Segar, M.; Liu, Y.; Nephew, K.P.; Matei, D. TGF-β induces global changes in DNA methylation during the epithelial-to-mesenchymal transition in ovarian cancer cells. Epigenetics 2014, 9, 1461–1472. [Google Scholar] [CrossRef]
- Chen, L.H.; Hsu, W.L.; Tseng, Y.J.; Liu, D.W.; Weng, C.F. Involvement of DNMT 3B promotes epithelial-mesenchymal transition and gene expression profile of invasive head and neck squamous cell carcinomas cell lines. BMC Cancer 2016, 16, 431. [Google Scholar]
- Güvenç, C.; Neckebroeck, F.; Antoranz, A.; Garmyn, M.; van den Oord, J.; Bosisio, F.M. Bona fide tumor suppressor genes hypermethylated in melanoma: A narrative review. Int. J. Mol. Sci. 2021, 22, 10674. [Google Scholar] [CrossRef]
- Haase, V.H. Oxygen regulates epithelial-to-mesenchymal transition: Insights into molecular mechanisms and relevance to disease. Kidney Int. 2009, 76, 492–499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, S.I.; Choi, M.E. TGF-beta-activated kinase-1: New insights into the mechanism of TGF-beta signaling and kidney disease. Kidney Res. Clin. Pract. 2012, 31, 94–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- De Chiara, L.; Crean, J. Emerging transcriptional mechanisms in the regulation of epithelial to mesenchymal transition and cellular plasticity in the kidney. J. Clin. Med. 2016, 5, 6. [Google Scholar] [CrossRef] [Green Version]
- Lin, K.; Baritaki, S.; Militello, L.; Malaponte, G.; Bevelacqua, Y.; Bonavida, B. The role of B-RAF mutations in melanoma and the induction of EMT via dysregulation of the NF-κB/Snail/RKIP/PTEN circuit. Genes Cancer 2010, 1, 409–420. [Google Scholar] [CrossRef]
- Kaufhold, S.; Bonavida, B. Central role of Snail1 in the regulation of EMT and resistance in cancer: A target for therapeutic intervention. J. Exp. Clin. Cancer Res. 2014, 33, 62. [Google Scholar] [CrossRef]
- Javaid, S.; Zhang, J.; Anderssen, E.; Black, J.C.; Wittner, B.S.; Tajima, K.; Ting, D.T.; Smolen, G.A.; Zubrowski, M.; Desai, R.; et al. Dynamic chromatin modification sustains epithelial-mesenchymal transition following inducible expression of Snail-1. Cell Rep. 2013, 5, 1679–1689. [Google Scholar] [CrossRef] [Green Version]
- Micevic, G.; Theodosakis, N.; Bosenberg, M. Aberrant DNA methylation in melanoma: Biomarker and therapeutic opportunities. Clin. Epigenet. 2017, 9, 34. [Google Scholar] [CrossRef] [Green Version]
- Klose, R.J.; Bird, A.P. Genomic DNA methylation: The mark and its mediators. Trends Biochem. Sci. 2006, 31, 89–97. [Google Scholar] [CrossRef]
- Yin, S.; Zhang, Q.; Yang, J.; Lin, W.; Li, Y.; Chen, F.; Cao, W. TGFβ-incurred epigenetic aberrations of miRNA and DNA methyltransferase suppress Klotho and potentiate renal fibrosis. Biochim. Biophys. Acta Mol. Cell. Res. 2017, 1864, 1207–1216. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Khor, T.O.; Xu, C.; Shen, G.; Jeong, W.S.; Yu, S.; Kong, A.N. Activation of Nrf2-antioxidant signaling attenuates NF-kB-inflammatory response and elicits apoptosis. Biochem. Pharmacol. 2008, 76, 1485–1489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ganesh Yerra, V.; Negi, G.; Sharma, S.S.; Kumar, A. Potential therapeutic effects of the simultaneous targeting of the Nrf2 and NF-κB pathways in diabetic neuropathy. Redox Biol. 2013, 1, 394–397. [Google Scholar] [CrossRef] [Green Version]
- Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules 2020, 25, 5474. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.C. Glutathione synthesis. Biochim. Biophys. Acta 2013, 1830, 3143–3153. [Google Scholar] [CrossRef] [Green Version]
- Meng, T.; Fu, S.; He, D.; Hu, G.; Gao, X.; Zhang, Y.; Huang, B.; Du, J.; Zhou, A.; Su, Y.; et al. Evodiamine inhibits lipopolysaccharide (LPS)-induced inflammation in BV-2 cells via regulating AKT/Nrf2-HO-1/NF-κB signaling axis. Cell. Mol. Neurobiol. 2021, 41, 115–127. [Google Scholar] [CrossRef]
- Liu, Y.; Mayo, M.W.; Nagji, A.S.; Smith, P.W.; Ramsey, C.S.; Li, D.; Jones, D.R. Phosphorylation of RelA/p65 promotes DNMT-1 recruitment to chromatin and represses transcription of the tumor metastasis suppressor gene BRMS1. Oncogene 2012, 31, 1143–1154. [Google Scholar] [CrossRef] [PubMed]
- Klecker, R.W.; Cysyk, R.L.; Collins, J.M. Zebularine metabolism by aldehyde oxidase in hepatic cytosol from humans, monkeys, dogs, rats, and mice: Influence of sex and inhibitors. Bioorg. Med. Chem. 2006, 14, 62–66. [Google Scholar] [CrossRef]
- Holleran, J.L.; Parise, R.A.; Joseph, E.; Eiseman, J.L.; Covey, J.M.; Glaze, E.R.; Lyubimov, A.V.; Chen, Y.-F.; D’Argenio, D.Z.; Egorin, M.J. Plasma pharmacokinetics, oral bioavailability, and interspeciesscaling of the DNA methyltransferase inhibitor, zebularine. Clin. Cancer Res. 2005, 11, 3862–3868. [Google Scholar] [CrossRef] [Green Version]
- Chen, M.; Shabashvili, D.; Nawab, A.; Yang, S.X.; Dyer, L.M.; Brown, K.D.; Hollingshead, M.; Hunter, K.W.; Kaye, F.J.; Hochwald, S.N.; et al. DNA methyltransferase inhibitor, zebularine, delays tumor growth and induces apoptosis in a genetically engineered mouse model of breast cancer. Mol. Cancer Ther. 2012, 11, 370–382. [Google Scholar] [CrossRef] [Green Version]
- Herranz, M.; Martin-Caballero, J.; Fraga, M.F.; Ruiz-Cabello, J.; Flores, J.M.; Desco, M.; Victor, M.; Esteller, M. The novel DNA methylation inhibitor zebularine is effective against the development of murine T-cell lymphoma. Blood 2006, 107, 1174–1177. [Google Scholar] [CrossRef] [PubMed]
- Nittby, H.; Ericsson, P.; Förnvik, K.; Strömblad, S.; Jansson, L.; Xue, Z.; Skagerberg, G.; Widegren, B.; Sjögren, H.-O.; Salford, L.G. Zebularine induces long-term survival of pancreatic islet allotransplants in streptozotocin treated diabetic rats. PLoS ONE 2013, 8, e71981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Johnson, W.D.; Harder, J.B.; Naylor, J.; McCormick, D.L.; Detrisac, C.J.; Glaze, E.R.; Tomaszewski, J.E. A pharmacokinetic/pharmacodynamic approach to evaluating the safety of zebularine in non-human primates. Proc. Amer. Assoc. Cancer Res. 2006, 66, 309–310. [Google Scholar]
- Ning, X.H.; Ge, X.F.; Cui, Y.; An, H.X. Ulinastatin inhibits unilateral ureteral obstruction-induced renal interstitial fibrosis in rats via transforming growth factor β (TGF-β)/Smad signalling pathways. Int. Immunopharmacol. 2013, 15, 406–413. [Google Scholar] [CrossRef] [PubMed]
- Wu, W.P.; Chang, C.H.; Chiu, Y.T.; Ku, C.L.; Wen, M.C.; Shu, K.H.; Wu, M.J. A reduction of unilateral ureteral obstruction-induced renal fibrosis by a therapy combining valsartan with aliskiren. Am. J. Physiol. Renal Physiol. 2010, 299, F929–F941. [Google Scholar] [CrossRef] [PubMed]
- Cai, X.R.; Zhou, Q.-C.; Yu, J.; Feng, Y.-Z.; Xian, Z.-H.; Yang, W.C.; Mo, X.-K. Assessment of Renal Function in Patients with Unilateral Ureteral Obstruction Using Whole-Organ Perfusion Imaging with 320-Detector Row Computed Tomography. PLoS ONE 2015, 10, e0122454. [Google Scholar] [CrossRef] [PubMed]
- Chung, S.; Yoon, H.E.; Kim, S.J.; Kim, S.J.; Koh, E.S.; Hong, Y.A.; Park, C.W.; Chang, Y.S.; Shin, S.J. Oleanolic acid attenuates renal fibrosis in mice with unilateral ureteral obstruction via facilitating nuclear translocation of Nrf2. Nutr. Metab. 2014, 11, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, T.; Liu, T.; Xie, P.; Jiang, S.; Yi, W.; Dai, P.; Guo, X. UPLC-MS-based urine nontargeted metabolic profiling identifies dysregulation of pantothenate and CoA biosynthesis pathway in diabetic kidney disease. Life Sci. 2020, 258, 118160. [Google Scholar] [CrossRef]
- Soji, K.; Doi, S.; Nakashima, A.; Sasaki, K.; Doi, T.; Masaki, T. Deubiquitinase inhibitor PR-619 reduces Smad4 expression and suppresses renal fibrosis in mice with unilateral ureteral obstruction. PLoS ONE 2018, 13, e0202409. [Google Scholar] [CrossRef] [Green Version]
- Chung, S.; Son, M.; Chae, Y.; Oh, S.; Koh, E.S.; Kim, Y.K.; Shin, S.J.; Park, C.W.; Jung, S.C.; Kim, H.-S. Fabry disease exacerbates renal interstitial fibrosis after unilateral ureteral obstruction via impaired autophagy and enhanced apoptosis. Kidney Res. Clin. Pract. 2020, 40, 208–219. [Google Scholar] [CrossRef]
- Chung, S.; Kim, S.; Son, M.; Kim, M.; Koh, E.S.; Shin, S.J.; Park, C.W.; Kim, H.-S. Inhibition of p300/CBP-Associated Factor Attenuates Renal Tubulointerstitial Fibrosis through Modulation of NF-kB and Nrf2. Int. J. Mol. Sci. 2019, 20, 1554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chung, S.; Kim, S.; Kim, M.; Koh, E.S.; Yoon, H.E.; Kim, H.S.; Park, C.W.; Chang, Y.S.; Shin, S.J. T-type calcium channel blocker attenuates unilateral ureteral obstruction-induced renal interstitial fibrosis by activating the Nrf2 antioxidant pathway. Am. J. Transl. Res. 2016, 8, 4574–4585. [Google Scholar] [PubMed]
- Schmittgen, T.D.; Livak, K.J. Analyzing real-time PCR data by the comparative C(T) method. Nat. Protoc. 2008, 3, 1101–1108. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Koh, E.S.; Kim, S.; Son, M.; Park, J.-Y.; Pyo, J.; Kim, W.-Y.; Kim, M.; Chung, S.; Park, C.W.; Kim, H.-S.; et al. The Protective Effect of Zebularine, an Inhibitor of DNA Methyltransferase, on Renal Tubulointerstitial Inflammation and Fibrosis. Int. J. Mol. Sci. 2022, 23, 14045. https://doi.org/10.3390/ijms232214045
Koh ES, Kim S, Son M, Park J-Y, Pyo J, Kim W-Y, Kim M, Chung S, Park CW, Kim H-S, et al. The Protective Effect of Zebularine, an Inhibitor of DNA Methyltransferase, on Renal Tubulointerstitial Inflammation and Fibrosis. International Journal of Molecular Sciences. 2022; 23(22):14045. https://doi.org/10.3390/ijms232214045
Chicago/Turabian StyleKoh, Eun Sil, Soojeong Kim, Mina Son, Ji-Young Park, Jaehyuk Pyo, Wan-Young Kim, Minyoung Kim, Sungjin Chung, Cheol Whee Park, Ho-Shik Kim, and et al. 2022. "The Protective Effect of Zebularine, an Inhibitor of DNA Methyltransferase, on Renal Tubulointerstitial Inflammation and Fibrosis" International Journal of Molecular Sciences 23, no. 22: 14045. https://doi.org/10.3390/ijms232214045
APA StyleKoh, E. S., Kim, S., Son, M., Park, J. -Y., Pyo, J., Kim, W. -Y., Kim, M., Chung, S., Park, C. W., Kim, H. -S., & Shin, S. J. (2022). The Protective Effect of Zebularine, an Inhibitor of DNA Methyltransferase, on Renal Tubulointerstitial Inflammation and Fibrosis. International Journal of Molecular Sciences, 23(22), 14045. https://doi.org/10.3390/ijms232214045