open access

Vol 59, No 2 (2021)
Original paper
Submitted: 2021-01-26
Accepted: 2021-05-10
Published online: 2021-05-19
Get Citation

Astragalus polysaccharide protects formaldehyde-induced toxicity by promoting NER pathway in bone marrow mesenchymal stem cells

Yali She12, Xiaowen Zhao1, Pingfan Wu1, Ling Xue1, Zhe Liu1, Meng Zhu1, Jie Yang1, Yaling Li12
·
Pubmed: 34008856
·
Folia Histochem Cytobiol 2021;59(2):124-133.
Affiliations
  1. Department of Pathology, School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
  2. Provincial-Level Key Laboratory of Molecular Medicine of Major Diseases and Study on Prevention and Treatment of Traditional Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu, China

open access

Vol 59, No 2 (2021)
ORIGINAL PAPERS
Submitted: 2021-01-26
Accepted: 2021-05-10
Published online: 2021-05-19

Abstract

Introduction. In our previous study, it has been confirmed that formaldehyde (FA) not only inhibits the proliferative activity, but also causes DNA-protein crosslinks (DPCs) formation in bone marrow mesenchymal stem cells (BMSCs). The purpose of this study was to detect the protective effect of astragalus polysaccharide (APS) against the cytotoxicity and genotoxicity of BMSCs exposed to FA, and to explore potential molecular mechanisms of APS activity. Material and methods. Human BMSCs were cultured in vitro and randomly divided into control cells (Ctrl group), FA-treated cells (FA group, 120 μmol/L), and cells incubated with FA and increasing concentrations (40, 100, or 400 μg/mL) of APS (FA + APS groups). Cytotoxicity was measured by MTT assay. DNA strand breakage, DNA-protein crosslinks (DPCs), and micronucleus formation were respectively detected by comet assay, KCl-SDS precipitation assay, and micronucleus assay. The mRNA and protein expression level of xeroderma pigmentosum group A (XPA), xeroderma pigmentosum group C (XPC), excision repair cross-complementation group 1 (ERCC1), replication protein A1 (RPA1), and replication protein A2 (RPA2) were all detected by qRT-PCR and Western Blot. Results. Compared with the FA group, the cytotoxicity, DNA strand breakage, DPCs, and micronucleus levels were decreased significantly in FA + APS groups (P < 0.01). Meanwhile, the mRNA and protein expression of XPA, XPC, ERCC1, RPA1, and RPA2 were up regulated significantly in the FA + APS groups (P < 0.05) with the most prominent effect of the 100 μg/mL APS. Conclusions. Our results suggest that APS can protect the cytotoxicity and genotoxicity of human BMSCs induced by FA. The mechanism may be associated with up-regulated expression of XPA, XPC, ERCC1, RPA1, and RPA2 in the nucleotide excision repair (NER) pathway which promotes DNA damage repair.

Abstract

Introduction. In our previous study, it has been confirmed that formaldehyde (FA) not only inhibits the proliferative activity, but also causes DNA-protein crosslinks (DPCs) formation in bone marrow mesenchymal stem cells (BMSCs). The purpose of this study was to detect the protective effect of astragalus polysaccharide (APS) against the cytotoxicity and genotoxicity of BMSCs exposed to FA, and to explore potential molecular mechanisms of APS activity. Material and methods. Human BMSCs were cultured in vitro and randomly divided into control cells (Ctrl group), FA-treated cells (FA group, 120 μmol/L), and cells incubated with FA and increasing concentrations (40, 100, or 400 μg/mL) of APS (FA + APS groups). Cytotoxicity was measured by MTT assay. DNA strand breakage, DNA-protein crosslinks (DPCs), and micronucleus formation were respectively detected by comet assay, KCl-SDS precipitation assay, and micronucleus assay. The mRNA and protein expression level of xeroderma pigmentosum group A (XPA), xeroderma pigmentosum group C (XPC), excision repair cross-complementation group 1 (ERCC1), replication protein A1 (RPA1), and replication protein A2 (RPA2) were all detected by qRT-PCR and Western Blot. Results. Compared with the FA group, the cytotoxicity, DNA strand breakage, DPCs, and micronucleus levels were decreased significantly in FA + APS groups (P < 0.01). Meanwhile, the mRNA and protein expression of XPA, XPC, ERCC1, RPA1, and RPA2 were up regulated significantly in the FA + APS groups (P < 0.05) with the most prominent effect of the 100 μg/mL APS. Conclusions. Our results suggest that APS can protect the cytotoxicity and genotoxicity of human BMSCs induced by FA. The mechanism may be associated with up-regulated expression of XPA, XPC, ERCC1, RPA1, and RPA2 in the nucleotide excision repair (NER) pathway which promotes DNA damage repair.

Get Citation

Keywords

astragalus polysaccharide; formaldehyde; bone marrow mesenchymal stem cells; toxicity; NER pathway; DNA strand breakage; DNA-protein crosslinks; micronucleus formation

About this article
Title

Astragalus polysaccharide protects formaldehyde-induced toxicity by promoting NER pathway in bone marrow mesenchymal stem cells

Journal

Folia Histochemica et Cytobiologica

Issue

Vol 59, No 2 (2021)

Article type

Original paper

Pages

124-133

Published online

2021-05-19

Page views

1737

Article views/downloads

814

DOI

10.5603/FHC.a2021.0013

Pubmed

34008856

Bibliographic record

Folia Histochem Cytobiol 2021;59(2):124-133.

Keywords

astragalus polysaccharide
formaldehyde
bone marrow mesenchymal stem cells
toxicity
NER pathway
DNA strand breakage
DNA-protein crosslinks
micronucleus formation

Authors

Yali She
Xiaowen Zhao
Pingfan Wu
Ling Xue
Zhe Liu
Meng Zhu
Jie Yang
Yaling Li

References (44)
  1. Moore KA, Lemischka IR. Stem cells and their niches. Science. 2006; 311(5769): 1880–1885.
  2. Méndez-Ferrer S, Michurina TV, Ferraro F, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010; 466(7308): 829–834.
  3. Zhang L, Freeman LE, Nakamura J, et al. Formaldehyde and leukemia: epidemiology, potential mechanisms, and implications for risk assessment. Environ Mol Mutagen. 2010; 51(3): 181–191.
  4. Chemical agents and related occupations. IARC Monogr Eval Carcinog Risks Hum. 2012; 100: 9–562.
  5. Zhang L, Tang X, Rothman N, et al. Occupational exposure to formaldehyde, hematotoxicity, and leukemia-specific chromosome changes in cultured myeloid progenitor cells. Cancer Epidemiol Biomarkers Prev. 2010; 19(1): 80–88.
  6. Liang X, Zhang J, Song W, et al. Formaldehyde Exposure in Indoor Air From Public Places and Its Associated Health Risks in Kunshan City, China. Asia Pac J Public Health. 2018; 30(6): 551–560.
  7. She Y, Li Yi, Liu Y, et al. Formaldehyde induces toxic effects and regulates the expression of damage response genes in BM-MSCs. Acta Biochim Biophys Sin (Shanghai). 2013; 45(12): 1011–1020.
  8. Ortega-Atienza S, Wong VC, DeLoughery Z, et al. ATM and KAT5 safeguard replicating chromatin against formaldehyde damage. Nucleic Acids Res. 2016; 44(1): 198–209.
  9. Zhang S, Chen H, Zhang J, et al. The multiplex interactions and molecular mechanism on genotoxicity induced by formaldehyde and acrolein mixtures on human bronchial epithelial BEAS-2B cells. Environ Int. 2020; 143: 105943.
  10. Liang FQ, Godley B. Oxidative stress-induced mitochondrial DNA damage in human retinal pigment epithelial cells: a possible mechanism for RPE aging and age-related macular degeneration. Exp Eye Res. 2003; 76(4): 397–403.
  11. Hubal EA, Schlosser PM, Conolly RB, et al. Comparison of inhaled formaldehyde dosimetry predictions with DNA-protein cross-link measurements in the rat nasal passages. Toxicol Appl Pharmacol. 1997; 143(1): 47–55.
  12. Latimer JJ, Johnson JM, Kelly CM, et al. Nucleotide excision repair deficiency is intrinsic in sporadic stage I breast cancer. Proc Natl Acad Sci U S A. 2010; 107(50): 21725–21730.
  13. Vrouwe MG, Pines A, Overmeer RM, et al. UV-induced photolesions elicit ATR-kinase-dependent signaling in non-cycling cells through nucleotide excision repair-dependent and -independent pathways. J Cell Sci. 2011; 124(Pt 3): 435–446.
  14. Salem AMH, Nakano T, Takuwa M, et al. Genetic analysis of repair and damage tolerance mechanisms for DNA-protein cross-links in Escherichia coli. J Bacteriol. 2009; 191(18): 5657–5668.
  15. Yeung K, Gubili J, Cassileth B. An Evidence-based Review of Astragalus membranaceus (Astragalus) for Cancer Patients. Evidence-based Anticancer Materia Medica. 2011: 65–84.
  16. Jung Y, Jerng U, Lee S. A systematic review of anticancer effects of radix astragali. Chin J Integr Med. 2016; 22(3): 225–236.
  17. Wu J, Yu J, Wang J, et al. Astragalus polysaccharide enhanced antitumor effects of Apatinib in gastric cancer AGS cells by inhibiting AKT signalling pathway. Biomed Pharmacother. 2018; 100: 176–183.
  18. Liu T, Zhang M, Niu H, et al. Astragalus polysaccharide from Astragalus Melittin ameliorates inflammation via suppressing the activation of TLR-4/NF-κB p65 signal pathway and protects mice from CVB3-induced virus myocarditis. Int J Biol Macromol. 2019; 126: 179–186.
  19. Zheng Y, Ren W, Zhang L, et al. A Review of the Pharmacological Action of Astragalus Polysaccharide. Front Pharmacol. 2020; 11: 349.
  20. Chen M, May BH, Zhou IW, et al. Oxaliplatin-based chemotherapy combined with traditional medicines for neutropenia in colorectal cancer: A meta-analysis of the contributions of specific plants. Crit Rev Oncol Hematol. 2016; 105: 18–34.
  21. Zhang PP, Meng ZT, Wang LC, et al. Astragalus polysaccharide promotes the release of mature granulocytes through the L-selectin signaling pathway. Chin Med. 2015; 10: 17.
  22. Zhang LY, Yong WX, Wang L, et al. Astragalus Polysaccharide Eases G1 Phase-Correlative Bystander Effects through Mediation of TGF- R/MAPK/ROS Signal Pathway After Carbon Ion Irradiation in BMSCs. Am J Chin Med. 2019; 47(3): 595–612.
  23. Yang F, Yan G, Li Y, et al. Astragalus Polysaccharide Attenuated Iron Overload-Induced Dysfunction of Mesenchymal Stem Cells via Suppressing Mitochondrial ROS. Cell Physiol Biochem. 2016; 39(4): 1369–1379.
  24. Liu Y, Li CM, Lu Z, et al. Studies on formation and repair of formaldehyde-damaged DNA by detection of DNA-protein crosslinks and DNA breaks. Front Biosci. 2006; 11: 991–997.
  25. Formaldehyde, 2-butoxyethanol and 1-tert-butoxypropan-2-ol. IARC Monogr Eval Carcinog Risks Hum. 2006; 88: 1–478.
  26. Ji S, Thomforde J, Rogers C, et al. Transcriptional Bypass of DNA-Protein and DNA-Peptide Conjugates by T7 RNA Polymerase. ACS Chem Biol. 2019; 14(12): 2564–2575.
  27. Ji S, Fu I, Naldiga S, et al. 5-Formylcytosine mediated DNA-protein cross-links block DNA replication and induce mutations in human cells. Nucleic Acids Res. 2018; 46(13): 6455–6469.
  28. Permana PA, Snapka RM. Aldehyde-induced protein-DNA crosslinks disrupt specific stages of SV40 DNA replication. Carcinogenesis. 1994; 15(5): 1031–1036.
  29. Zhang L, Luo Y, Lu Z, et al. Astragalus Polysaccharide Inhibits Ionizing Radiation-Induced Bystander Effects by Regulating MAPK/NF-kB Signaling Pathway in Bone Mesenchymal Stem Cells (BMSCs). Med Sci Monit. 2018; 24: 4649–4658.
  30. Zhu J, Fu W, Jia W, et al. Association between NER Pathway Gene Polymorphisms and Wilms Tumor Risk. Mol Ther Nucleic Acids. 2018; 12: 854–860.
  31. Kojima Y, Machida YJ. DNA-protein crosslinks from environmental exposure: Mechanisms of formation and repair. Environ Mol Mutagen. 2020; 61(7): 716–729.
  32. Baker DJ, Wuenschell G, Xia L, et al. Nucleotide excision repair eliminates unique DNA-protein cross-links from mammalian cells. J Biol Chem. 2007; 282(31): 22592–22604.
  33. Buschta-Hedayat N, Buterin T, Hess MT, et al. Recognition of nonhybridizing base pairs during nucleotide excision repair of DNA. Proc Natl Acad Sci U S A. 1999; 96(11): 6090–6095.
  34. Wood R. DNA damage recognition during nucleotide excision repair in mammalian cells. Biochimie. 1999; 81(1-2): 39–44.
  35. Yasuda G, Nishi R, Watanabe E, et al. In vivo destabilization and functional defects of the xeroderma pigmentosum C protein caused by a pathogenic missense mutation. Mol Cell Biol. 2007; 27(19): 6606–6614.
  36. Servant G, Streva VA, Derbes RS, et al. The Nucleotide Excision Repair Pathway Limits L1 Retrotransposition. Genetics. 2017; 205(1): 139–153.
  37. Sijbers A, Laat Wde, Ariza R, et al. Xeroderma Pigmentosum Group F Caused by a Defect in a Structure-Specific DNA Repair Endonuclease. Cell. 1996; 86(5): 811–822.
  38. O'Donovan A, Davies AA, Moggs JG, et al. XPG endonuclease makes the 3' incision in human DNA nucleotide excision repair. Nature. 1994; 371(6496): 432–435.
  39. Cassee FR, de Burbure CY, Rambali B, et al. Subchronic inhalation of mixtures of cigarette smoke constituents in Xpa-/-p53+/- knock-out mice: a comparison of intermittent with semi-continuous exposure to acetaldehyde, formaldehyde, and acrolein. Food Chem Toxicol. 2008; 46(2): 527–536.
  40. Saijo M, Kuraoka I, Masutani C, et al. Sequential binding of DNA repair proteins RPA and ERCC1 to XPA in vitro. Nucleic Acids Res. 1996; 24(23): 4719–4724.
  41. Lindsey-Boltz LA, Kemp MG, Reardon JT, et al. Coupling of human DNA excision repair and the DNA damage checkpoint in a defined in vitro system. J Biol Chem. 2014; 289(8): 5074–5082.
  42. Byrne BM, Oakley GG. Replication protein A, the laxative that keeps DNA regular: The importance of RPA phosphorylation in maintaining genome stability. Semin Cell Dev Biol. 2019; 86: 112–120.
  43. Krasikova YS, Rechkunova NI, Maltseva EA, et al. Localization of xeroderma pigmentosum group A protein and replication protein A on damaged DNA in nucleotide excision repair. Nucleic Acids Res. 2010; 38(22): 8083–8094.
  44. Krasikova YS, Rechkunova NI, Maltseva EA, et al. RPA and XPA interaction with DNA structures mimicking intermediates of the late stages in nucleotide excision repair. PLoS One. 2018; 13(1): e0190782.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk

tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl