We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Identification of histone acetylation modification sites in the striatum of subchronically manganese-exposed rats

    Chunyan Ao

    School of Public Health, the Key Laboratory of Environmental Pollution Monitoring & Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 561113, China

    ,
    Shunfang Tang

    School of Public Health, the Key Laboratory of Environmental Pollution Monitoring & Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 561113, China

    ,
    Yue Yang

    School of Public Health, the Key Laboratory of Environmental Pollution Monitoring & Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 561113, China

    ,
    Ying Liu

    School of Public Health, the Key Laboratory of Environmental Pollution Monitoring & Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 561113, China

    ,
    Hua Zhao

    School of Public Health, the Key Laboratory of Environmental Pollution Monitoring & Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 561113, China

    ,
    Jiaqi Ban

    School of Public Health, the Key Laboratory of Environmental Pollution Monitoring & Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 561113, China

    &
    Jun Li

    *Author for correspondence:

    E-mail Address: gygwlj@163.com

    School of Public Health, the Key Laboratory of Environmental Pollution Monitoring & Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 561113, China

    Published Online:https://doi.org/10.2217/epi-2023-0364

    Aim: To explore the specific histone acetylation sites and oxidative stress-related genes that are associated with the pathogenesis of manganese toxicity. Methods: We employed liquid chromatography–tandem mass spectrometry and bioinformatics analysis to identify acetylated proteins in the striatum of subchronic manganese-intoxicated rats. Results: We identified a total of 12 differentially modified histone acetylation sites: H3K9ac, H3K14ac, H3K18ac, H3K56ac and H3K79ac were upregulated and H3K27ac, H3K36ac, H4K91ac, H4K79ac, H4K31ac, H2BK16ac and H2BK20ac were downregulated. Additionally, we found that CAT, SOD1 and SOD2 might be epigenetically regulated and involved in the pathogenesis of manganism. Conclusion: This study identified histone acetylation sites and oxidative stress-related genes associated with the pathogenesis of manganese toxicity, and these findings are useful in the search for potential epigenetic targets for manganese toxicity.

    Tweetable abstract

    We identified specific histone acetylation sites associated with manganese toxicity, which will help to explore targets for pharmacological intervention in manganese toxicity as well as pathogenesis studies.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Horning KJ, Caito SW, Tipps KG, Bowman AB, Aschner M. Manganese is essential for neuronal health. Annu. Rev. Nutr. 35, 71–108 (2015).
    • 2. Chen P. Manganese metabolism in humans. Front. Biosci. 23(9), 1655–1679 (2018).
    • 3. Rahman SM, Kippler M, Ahmed S, Palm B, El Arifeen S, Vahter M. Manganese exposure through drinking water during pregnancy and size at birth: a prospective cohort study. Reprod. Toxicol. 53, 68–74 (2015).
    • 4. Racette BA, Nelson G, Dlamini WW et al. Environmental manganese exposure and cognitive control in a South African population. Neurotoxicology 89, 31–40 (2022).
    • 5. Wu J, Zhang Q, Sun P et al. Gray matter microstructural alterations in manganese-exposed welders: a preliminary neuroimaging study. Eur. Radiol. 32(12), 8649–8658 (2022).
    • 6. Huang S, Liu Z, Ge X et al. Occupational exposure to manganese and risk of creatine kinase and creatine kinase-MB elevation among ferromanganese refinery workers. Am. J. Ind. Med. 63(5), 394–401 (2020).
    • 7. Chen X, Liu Z, Ge X et al. Associations between manganese exposure and multiple immunological parameters in manganese-exposed workers healthy cohort. J. Trace Elem. Med. Biol. 59, 126454 (2020).
    • 8. Pajarillo E, Nyarko-Danquah I, Digman A et al. Mechanisms of manganese-induced neurotoxicity and the pursuit of neurotherapeutic strategies. Front. Pharmacol. 13, 1011947 (2022).
    • 9. Mehrifar Y, Bahrami M, Sidabadi E, Pirami H. The effects of occupational exposure to manganese fume on neurobehavioral and neurocognitive functions: an analytical cross-sectional study among welders. EXCLI J. 19, 372–386 (2020).
    • 10. Yamamoto M, Eguchi A, Sakurai K et al. Longitudinal analyses of maternal and cord blood manganese levels and neurodevelopment in children up to 3 years of age: the Japan Environment and Children’s Study (JECS). Environ. Int. 161, 107126 (2022). • Explored the effects of manganese exposure during pregnancy on child neurodevelopment through a birth cohort study.
    • 11. Dent SE, King DP, Osterberg VR et al. Phosphorylation of the aggregate-forming protein alpha-synuclein on serine-129 inhibits its DNA-bending properties. J. Biol. Chem. 298(2), 101552 (2021).
    • 12. Choi H, Kim HJ, Yang J et al. Acetylation changes tau interactome to degrade tau in Alzheimer’s disease animal and organoid models. Aging Cell 19(1), e13081 (2020).
    • 13. Nucifora FC, Nucifora LG, Ng C-H et al. Ubiqutination via K27 and K29 chains signals aggregation and neuronal protection of LRRK2 by WSB1. Nat. Commun. 7(1), 11792 (2016).
    • 14. Li M, Xiong J, Yang L et al. Acetylation of p62 regulates base excision repair through interaction with APE1. Cell Rep. 40(3), 111116 (2022).
    • 15. Li H, Chen J-A, Ding Q-Z et al. Behavioral sensitization induced by methamphetamine causes differential alterations in gene expression and histone acetylation of the prefrontal cortex in rats. BMC Neurosci. 22, 24 (2021).
    • 16. Gruber JJ, Geller B, Lipchik AM et al. HAT1 coordinates histone production and acetylation via H4 promoter binding. Mol. Cell 75(4), 711–724.e5 (2019).
    • 17. Liu X, Tan Y, Zhang C et al. NAT10 regulates p53 activation through acetylating p53 at K120 and ubiquitinating Mdm2. EMBO Rep. 17(3), 349–366 (2016).
    • 18. Lozoya OA, Wang T, Grenet D et al. Mitochondrial acetyl-CoA reversibly regulates locus-specific histone acetylation and gene expression. Life Sci. Alliance 2(1), e201800228 (2019).
    • 19. Nativio R, Lan Y, Donahue G et al. An integrated multi-omics approach identifies epigenetic alterations associated with Alzheimer’s disease. Nat. Genet. 52(10), 1024–1035 (2020).
    • 20. Toker L, Tran GT, Sundaresan J et al. Genome-wide histone acetylation analysis reveals altered transcriptional regulation in the Parkinson’s disease brain. Mol. Neurodegener. 16, 31 (2021). •• Differential modification of histone acetylation sites in brain tissue of healthy controls and Parkinson's disease patients was analyzed by genome-wide analysis. RNA sequencing data were also used to assess the impact of histone acetylation alterations on gene expression.
    • 21. Cong L, Lei M-Y, Liu Z-Q et al. Resveratrol attenuates manganese-induced oxidative stress and neuroinflammation through SIRT1 signaling in mice. Food Chem. Toxicol. 153, 112283 (2021).
    • 22. Li S-J, Li Y, Chen J-W et al. Sodium para-aminosalicylic acid protected primary cultured basal ganglia neurons of rat from manganese-induced oxidative impairment and changes of amino acid neurotransmitters. Biol. Trace Elem. Res. 170(2), 357–365 (2016).
    • 23. Yang Y, Liu Y, Zhang A-L et al. Curcumin protects against manganese-induced neurotoxicity in rat by regulating oxidative stress-related gene expression via H3K27 acetylation. Ecotoxicol. Environ. Saf. 236, 113469 (2022).
    • 24. Liu L, Wang H, Rao X et al. Comprehensive analysis of the lysine acetylome and succinylome in the hippocampus of gut microbiota-dysbiosis mice. J. Adv. Res. 30, 27–38 (2020). •• Analyzed alterations in hippocampal lysine acetyl and succinic acid in mice receiving gut microbiota from fecal samples from patients with major depressive disorder or healthy control patients using HPLC–tandem mass spectrometry.
    • 25. Li D, Wan C, Bai B, Cao H, Liu C, Zhang Q. Identification of histone acetylation markers in human fetal brains and increased H4K5ac expression in neural tube defects. Mol. Genet. Genomic Med. 7(12), e1002 (2019). • Provides a comprehensive map of histone H4 modifications in the human fetal brain.
    • 26. Cordova FM, Aguiar AS, Peres TV et al. Oxidative stress is involved in striatal neurotoxicity in manganese-exposed developing rats. Arch. Toxicol. 87(7), 1231–1244 (2013).
    • 27. Dong J, He J, Zhang Z et al. Identification of lysine acetylome of oral squamous cell carcinoma by label-free quantitative proteomics. J. Proteomics 262, 104598 (2022).
    • 28. Wang L, Nwosu C, Gao Y, Zhu MM. Signature ions triggered Electron-Transfer/Higher-Energy Collisional Dissociation (EThcD) for specific and confident glycation site mapping in therapeutic proteins. J. Am. Soc. Mass Spectrom. 31(3), 473–478 (2020).
    • 29. Guo Z, Zhang Z, Wang Q et al. Manganese chloride induces histone acetylation changes in neuronal cells: its role in manganese-induced damage. Neurotoxicology 65, 255–263 (2018).
    • 30. Zhang Z, Guo Z, Zhan Y, Li H, Wu S. Role of histone acetylation in activation of nuclear factor erythroid 2-related factor 2/heme oxygenase 1 pathway by manganese chloride. Toxicol. Appl. Pharm. 336, 94–100 (2017).
    • 31. Dlamini WW, Nelson G, Nielsen SS, Racette BA. Manganese exposure, parkinsonian signs, and quality of life in South African mine workers. Am. J. Ind. Med. 63(1), 36–43 (2020).
    • 32. Nakayasu ES, Burnet MC, Walukiewicz HE et al. Ancient regulatory role of lysine acetylation in central metabolism. mBio. 8(6), e01894-17 (2017).
    • 33. Drazic A, Myklebust LM, Ree R, Arnesen T. The world of protein acetylation. BBA - Proteins Proteom. 1864(10), 1372–1401 (2016).
    • 34. Arenas A, Chen J, Kuang L et al. Lysine acetylation regulates the RNA binding, subcellular localization and inclusion formation of FUS. Hum. Mol. Genet. 29(16), 2684–2697 (2020).
    • 35. Di Cerbo V, Mohn F, Ryan DP et al. Acetylation of histone H3 at lysine 64 regulates nucleosome dynamics and facilitates transcription. eLife 3, e01632 (2014).
    • 36. Roy D, Paul A, Roy A, Ghosh R, Ganguly P, Chaudhuri S. Differential acetylation of histone H3 at the regulatory region of OsDREB1b promoter facilitates chromatin remodelling and transcription activation during cold stress. PLoS ONE 9(6), e100343 (2014).
    • 37. Arima Y, Nakagawa Y, Takeo T et al. Murine neonatal ketogenesis preserves mitochondrial energetics by preventing protein hyperacetylation. Nat. Metab. 3(2), 196–210 (2021).
    • 38. Lohr KM, Masoud ST, Salahpour A, Miller GW. Membrane transporters as mediators of synaptic dopamine dynamics: implications for disease. Eur. J. Neurosci. 45(1), 20–33 (2017).
    • 39. Wang L, Zhang Z, Hou L et al. Phytic acid attenuates upregulation of GSK-3β and disturbance of synaptic vesicle recycling in MPTP-induced Parkinson’s disease models. Neurochem. Int. 129, 104507 (2019).
    • 40. Liu Y, Tong S, Ding L, Liu N, Gao D. Serum levels of glial cell line-derived neurotrophic factor and multiple neurotransmitters: in relation to cognitive performance in Parkinson’s disease with mild cognitive impairment. Int. J. Geriatr. Psychiatry 35(2), 153–162 (2020).
    • 41. Wei B, Liu M, Chen Z, Wei M. Schisandrin ameliorates cognitive impairment and attenuates Aβ deposition in APP/PS1 transgenic mice: involvement of adjusting neurotransmitters and their metabolite changes in the brain. Acta Pharmacol. Sin. 39(4), 616–625 (2018).
    • 42. Bridi JC, Hirth F. Mechanisms of α-synuclein induced synaptopathy in Parkinson’s disease. Front. Neurosci. 12, 80 (2018).
    • 43. Garcia-Reitböck P, Anichtchik O, Bellucci A et al. SNARE protein redistribution and synaptic failure in a transgenic mouse model of Parkinson’s disease. Brain 133(7), 2032–2044 (2010).
    • 44. Yan F, Li J, Milosevic J et al. KAT6A and ENL form an epigenetic transcriptional control module to drive critical leukemogenic gene expression programs. Cancer Discov. 12(3), 792–811 (2022).
    • 45. Xu Y, Liao R, Li N, Xiang R, Sun P. Phosphorylation of Tip60 by p38α regulates p53-mediated PUMA induction and apoptosis in response to DNA damage. Oncotarget 5(24), 12555–12572 (2014).
    • 46. Varga J, Korbai S, Neller A, Zsindely N, Bodai L. Hat1 acetylates histone H4 and modulates the transcriptional program in Drosophila embryogenesis. Sci. Rep. 9, 17973 (2019).
    • 47. Valerio DG, Xu H, Eisold ME, Woolthuis CM, Pandita TK, Armstrong SA. Histone acetyltransferase activity of MOF is required for adult but not early fetal hematopoiesis in mice. Blood 129(1), 48–59 (2017).
    • 48. Cai L, Sutter BM, Li B, Tu BP. Acetyl-CoA induces cell growth and proliferation by promoting the acetylation of histones at growth genes. Mol. Cell 42(4), 426–437 (2011).
    • 49. Chai G, Gong J, Wu J et al. Danggui Buxue decoction ameliorates mitochondrial biogenesis and cognitive deficits through upregulating histone H4 lysine 12 acetylation in APP/PS1 mice. J. Ethnopharmacol. 313, 116554 (2023). •• The Chinese medicine Danggui Buxue decoction improves cognitive deficits in Alzheimer’s disease mice and may be a promising complementary candidate for Alzheimer’s treatment.
    • 50. Rossaert E, Pollari E, Jaspers T et al. Restoration of histone acetylation ameliorates disease and metabolic abnormalities in a FUS mouse model. Acta Neuropathol. Commun. 7, 107 (2019).
    • 51. Gebremedhin KG, Rademacher DJ. Histone H3 acetylation in the postmortem Parkinson’s disease primary motor cortex. Neurosci. Lett. 627, 121–125 (2016).
    • 52. Feng Q, Chai G-S, Wang Z-H et al. Knockdown of pp32 increases histone acetylation and ameliorates cognitive deficits. Front. Aging Neurosci. 9, 104 (2017).
    • 53. Wang Y, Branicky R, Noë A, Hekimi S. Superoxide dismutases: dual roles in controlling ROS damage and regulating ROS signaling. J. Cell Biol. 217(6), 1915–1928 (2018).
    • 54. Lee JN, Dutta RK, Maharjan Y et al. Catalase inhibition induces pexophagy through ROS accumulation. Biochem. Biophys. Res. Commun. 501(3), 696–702 (2018).
    • 55. Wang X, Gao X-Q, Wang X-Y et al. Bioaccumulation of manganese and its effects on oxidative stress and immune response in juvenile groupers (Epinephelus moara ♀ × E. lanceolatus ♂). Chemosphere 297, 134235 (2022).
    • 56. Bahar E, Kim J-Y, Yoon H. Quercetin attenuates manganese-induced neuroinflammation by alleviating oxidative stress through regulation of apoptosis, iNOS/NF-κB and HO-1/Nrf2 pathways. Int. J. Mol. Sci. 18(9), 1989 (2017).
    • 57. Yakes FM, Van Houten B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc. Natl Acad. Sci. USA 94(2), 514–519 (1997).
    • 58. Tong S, Xia M, Xu Y et al. Identification and validation of a novel prognostic signature based on mitochondria and oxidative stress related genes for glioblastoma. J. Transl. Med. 21, 136 (2023).
    • 59. Wang G, Song L, Bai T, Liang W. BcSas2-mediated histone H4K16 acetylation is critical for virulence and oxidative stress response of Botrytis cinerea. Mol. Plant Microbe Interact. 33(10), 1242–1251 (2020).
    • 60. Tokarz P, Kaarniranta K, Blasiak J. Inhibition of DNA methyltransferase or histone deacetylase protects retinal pigment epithelial cells from DNA damage induced by oxidative stress by the stimulation of antioxidant enzymes. Eur. J. Pharmacol. 776, 167–175 (2016).