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

Advances in the role of epigenetics in homocysteine-related diseases

    Chengyan Wu

    The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China

    ,
    Xulei Duan

    The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China

    ,
    Xuehui Wang

    *Author for correspondence: Tel.: +86 136 5373 9422;

    E-mail Address: 121045@xxmu.edu.cn

    The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China

    &
    Libo Wang

    The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China

    Published Online:https://doi.org/10.2217/epi-2023-0207

    Homocysteine has a wide range of biological effects. However, the specific molecular mechanism of its pathogenicity is still unclear. The diseases induced by hyperhomocysteinemia (HHcy) are called homocysteine-related diseases. Clinical treatment of HHcy is mainly through folic acid and B-complex vitamins, which are not effective in reducing the associated end point events. Epigenetics is the alteration of heritable genes caused by DNA methylation, histone modification, noncoding RNAs and chromatin remodeling without altering the DNA sequence. In recent years the role of epigenetics in homocysteine-associated diseases has been gradually discovered. This article summarizes the latest evidence on the role of epigenetics in HHcy, providing new directions for its prevention and treatment.

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

    References

    • 1. Hankey GJ, Eikelboom JW. Homocysteine and vascular disease. Lancet 345(9176), 407–413 (1999).
    • 2. Lonn E, Yusuf S, Arnold MJ et al. Homocysteine lowering with folic acid and B vitamins in vascular disease. N. Engl. J. Med. 354(15), 1567–1577 (2006).
    • 3. Koklesova L, Mazurakova A, Samec M et al. Homocysteine metabolism as the target for predictive medical approach, disease prevention, prognosis, and treatments tailored to the person. EPMA J. 12(4), 477–505 (2021).
    • 4. Motulsky AG. Nutritional ecogenetics: homocysteine-related arteriosclerotic vascular disease, neural tube defects, and folic acid. Am. J. Hum. Genet. 58(1), 17–20 (1996).
    • 5. Lin C, Chen Y, Leu H et al. Anti-inflammatory strategies for homocysteine-related cardiovascular disease. Front. Biosci. 14(10), 3836–3845 (2009).
    • 6. Ye Z, Wang C, Zhang Q et al. Prevalence of homocysteine-related hypertension in patients with chronic kidney disease. J. Clin. Hypertens. 19(2), 151–160 (2017).
    • 7. Vance SH, Tucci M, Benghuzzi H. Pathophysiological response of rhesus monkey kidney epithelial cells exposed to epigallocatechin-3-gallate. Biomed. Sci. Instrum. 41, 223–228 (2005).
    • 8. McCully KS. Vascular pathology of homocysteinemia: implications for the pathogenesis of arteriosclerosis. Am. J. Pathol. 56(1), 111–128 (1969). •• An early article that found that homocysteinemia (HHcy) can induce disease. The authors concluded that HHcy can induce arteriosclerosis in children.
    • 9. Nygård O, Vollset SE, Refsum H et al. Total plasma homocysteine and cardiovascular risk profile. The Hordaland Homocysteine Study. JAMA 274(19), 1526–1533 (1995).
    • 10. Refsum H, Ueland PM, Nygård O, Vollset SE. Homocysteine and cardiovascular disease. Annu. Rev. Med. 49, 31–62 (1998).
    • 11. Lonn E, Yusuf S, Arnold MJ et al. Homocysteine lowering with folic acid and B vitamins in vascular disease. N. Engl. J. Med. 354(15), 1567–1577 (2006). • Interesting article that discusses folic acid supplementation lowering Hcy without reducing the end point event.
    • 12. Study of the Effectiveness of Additional Reductions in Cholesterol and Homocysteine (SEARCH) Collaborative Group, Armitage JM, Bowman L, Clarke RJ et al. Effects of homocysteine-lowering with folic acid plus vitamin B12 vs placebo on mortality and major morbidity in myocardial infarction survivors: a randomized trial. JAMA 303(24), 2486–2494 (2010). • Interesting article that tells the story of folic acid supplementation lowering Hcy without reducing the end point event.
    • 13. Chan S, Hung C, Shih J et al. Exercise intervention attenuates hyperhomocystei nemia-induced aortic endothelial oxidative injury by regulating SIRT1 through mitigating NADPH oxidase/LOX-1 signaling. Redox. Biol. 14, 116–125 (2018).
    • 14. Chen J, Huang Y, Hu X, Bian X, Nian S. Gastrodin prevents homocysteine-induced human umbilical vein endothelial cells injury via PI3K/Akt/eNOS and Nrf2/ARE pathway. J. Cell. Mol. Med. 25(1), 345–357 (2021).
    • 15. Liu S, Tao J, Duan F, Li H, Tan H. HHcy induces pyroptosis and atherosclerosis via the lipid raft-mediated NOX-ROS-NLRP3 inflammasome pathway in apoE-/- mice. Cells 11(15), 2438 (2022).
    • 16. Seaks CE, Weekman EM, Sudduth TL et al. Apolipoprotein E ε4/4 genotype limits response to dietary induction of hyperhomocysteinemia and resulting inflammatory signaling. J. Cereb. Blood Flow Metab. 42(5), 771–787 (2022).
    • 17. Ji X, Yu H, Zhou G et al. Physcion, a tetra-substituted 9,10-anthraquinone, prevents homocysteine-induced endothelial dysfunction by activating Ca2+- and Akt-eNOS-NO signaling pathways. Phytomedicine 81, 153410 (2021).
    • 18. Carluccio MA, Ancora MA, Massaro M et al. Homocysteine induces VCAM-1 gene expression through NF-kappaB and NAD(P)H oxidase activation: protective role of Mediterranean diet polyphenolic antioxidants. Am. J. Physiol. Heart Circ. Physiol. 293(4), H2344–2354 (2007).
    • 19. Ji C, Yi H, Huang J, Zhang W, Zheng M. Propofol alleviates inflammation and apoptosis in HCY-induced HUVECs by inhibiting endoplasmic reticulum stress. Mol. Med. Rep. 23(5), 333 (2021).
    • 20. Hu H, Wang C, Jin Y et al. Catalpol inhibits homocysteine-induced oxidation and inflammation via inhibiting Nox4/NF-κB and GRP78/PERK pathways in human aorta endothelial cells. Inflammation 42(1), 64–80 (2019).
    • 21. Zhan B, Xu Z, Zhang Y et al. Nicorandil reversed homocysteine-induced coronary microvascular dysfunction via regulating PI3K/Akt/eNOS pathway. Biomed. Pharmacother. 127, 110121 (2020).
    • 22. Xie X, Zhang Z, Wang X et al. Stachydrine protects eNOS uncoupling and ameliorates endothelial dysfunction induced by homocysteine. Mol. Med. 24(1), 10 (2018).
    • 23. Jiang X, Yang F, Tan H et al. Hyperhomocystinemia impairs endothelial function and eNOS activity via PKC activation. Arterioscler. Thromb. Vasc. Biol. 25(12), 2515–2521 (2005).
    • 24. Wu B, Yue H, Zhou G et al. Protective effects of oxymatrine on homocystein-induced endothelial injury: involvement of mitochondria-dependent apoptosis and Akt-eNOS-NO signaling pathways. Eur. J. Pharmacol. 864, 172717 (2019).
    • 25. Liu Y, Zhou G, Song X et al. Emodin protects against homocysteine-induced cardiac dysfunction by inhibiting oxidative stress via MAPK and Akt/eNOS/NO signaling pathways. Eur. J. Pharmacol. 940, 175452 (2023).
    • 26. Flannigan KL, Agbor TA, Blackler RW et al. Impaired hydrogen sulfide synthesis and IL-10 signaling underlie hyperhomocysteinemia-associated exacerbation of colitis. Proc. Natl Acad. Sci. USA 111(37), 13559–13564 (2014).
    • 27. Wang Y, Shi S, Dong S et al. Sodium hydrosulfide attenuates hyperhomocysteinemia rat myocardial injury through cardiac mitochondrial protection. Mol. Cell. Biochem. 399(1–2), 189–200 (2015).
    • 28. Zhao Z, Liu X, Shi S et al. Exogenous hydrogen sulfide protects from endothelial cell damage, platelet activation, and neutrophils extracellular traps formation in hyperhomocysteinemia rats. Exp. Cell Res. 370(2), 434–443 (2018).
    • 29. Nakladal D, Lambooy SPH, Mišúth S et al. Homozygous whole body Cbs knockout in adult mice features minimal pathology during ageing despite severe homocysteinemia. FASEB J. 36(4), e22260 (2022).
    • 30. Han L, Miao Y, Zhao Y et al. The binding of autotaxin to integrins mediates hyperhomocysteinemia-potentiated platelet activation and thrombosis in mice and humans. Blood Adv. 6(1), 46–61 (2022).
    • 31. Fryer RH, Wilson BD, Gubler DB, Fitzgerald LA, Rodgers GM. Homocysteine, a risk factor for premature vascular disease and thrombosis, induces tissue factor activity in endothelial cells. Arterioscler. Thromb. 13(9), 1327–1333 (1993).
    • 32. Kolodziejczyk-Czepas J, Talar B, Nowak P, Olas B, Wachowicz B. Homocysteine and its thiolactone impair plasmin activity induced by urokinase or streptokinase in vitro. Int. J. Biol. Macromol. 50(3), 754–758 (2012).
    • 33. Malinowska J, Tomczynska M, Olas B. Changes of blood platelet adhesion to collagen and fibrinogen induced by homocysteine and its thiolactone. Clin. Biochem. 45(15), 1225–1228 (2012).
    • 34. Sauls DL, Wolberg AS, Hoffman M. Elevated plasma homocysteine leads to alterations in fibrin clot structure and stability: implications for the mechanism of thrombosis in hyperhomocysteinemia. J. Thromb. Haemost. 1(2), 300–306 (2003).
    • 35. Siennicka A, Zuchowski M, Chełstowski K, Cnotliwy M, Clark JS, Jastrzębska M. Homocysteine-enhanced proteolytic and fibrinolytic processes in thin intraluminal thrombus and adjacent wall of abdominal aortic aneurysm: study in vitro. Biomed Res. Int. 2018, 3205324 (2018).
    • 36. Bao XM, Zheng H. Atorvastatin attenuates homocysteine-induced migration of smooth muscle cells through mevalonate pathway involving reactive oxygen species and p38 MAPK. Clin. Exp. Pharmacol. Physiol. 42( 8), 865–873 (2015).
    • 37. Liu J, Yan X, Wang Z, Zhang N, Lin A, Li Z. Adipocyte factor CTRP6 inhibits homocysteine-induced proliferation, migration, and dedifferentiation of vascular smooth muscle cells through PPARγ/NLRP3. Biochem. Cell Biol. 99(5), 596–605 (2021).
    • 38. Ji J, Feng M, Niu X, Zhang X, Wang Y. Liraglutide blocks the proliferation, migration and phenotypic switching of homocysteine (Hcy)-induced vascular smooth muscle cells (VSMCs) by suppressing proprotein convertase subtilisin kexin9 (PCSK9)/low-density lipoprotein receptor (LDLR). Bioengineered 12(1), 8057–8066 (2021).
    • 39. Ma H, Wang L, Lv W, Lv Z. Effects of miR-7 on Hcy-induced rat cerebral arterial vascular smooth muscle cell proliferation, migration and inflammatory factor expression by targeting MMP-14 to regulate TLR4/NF-κB signaling pathway. Cell. Mol. Biol. (Noisy-le-grand) 66(7), 12–17 (2020).
    • 40. Duan H, Li Y, Yan L et al. MicroRNA-217 suppresses homocysteine-induced proliferation and migration of vascular smooth muscle cells via N-methyl-D-aspartic acid receptor inhibition. Clin. Exp. Pharmacol. Physiol. 43(10), 967–975 (2016).
    • 41. Meng L, Liu L, Zhou C et al. Polyphenols and polypeptides in Chinese rice wine inhibit homocysteine-induced proliferation and migration of vascular smooth muscle cells. J. Cardiovasc. Pharmacol. 67(6), 482–490 (2016).
    • 42. Luo X, Xiao Y, Song F, Yang Y, Xia M, Ling W. Increased plasma S-adenosyl-homocysteine levels induce the proliferation and migration of VSMCs through an oxidative stress-ERK1/2 pathway in apoE(-/-) mice. Cardiovasc. Res. 95(2), 241–250 (2012).
    • 43. Chou Y, Lin H, Chen K, Chang C, Lee W, Juan S. Molecular mechanisms underlying the anti-proliferative and anti-migratory effects of folate on homocysteine-challenged rat aortic smooth muscle cells. Br. J. Pharmacol. 169(7), 1447–1460 (2013).
    • 44. Zhang T, Lu R, Chen Y et al. Hyperhomocysteinemia and dyslipidemia in point mutation G307S of cystathionine β-synthase-deficient rabbit generated using CRISPR/Cas9. Lipids Health Dis. 19(1), 224 (2020).
    • 45. Minno AD, Anesi A, Chiesa M et al. Plasma phospholipid dysregulation in patients with cystathionine-β synthase deficiency. Nutr. Metab. Cardiovasc. Dis. 30(12), 2286–2295 (2020).
    • 46. Al-Shabrawey M, Elmarakby A, Samra Y et al. Hyperhomocysteinemia dysregulates plasma levels of polyunsaturated fatty acids-derived eicosanoids. Life Res. (Auckl.) 5(2), 14 (2022).
    • 47. Werstuck GH, Lentz SR, Dayal S et al. Homocysteine-induced endoplasmic reticulum stress causes dysregulation of the cholesterol and triglyceride biosynthetic pathways. J. Clin. Investig. 107(10), 1263–1273 (2001).
    • 48. Mikael EG, Genest J, Rozen R. Elevated homocysteine reduces apolipoprotein A-I expression in hyperhomocysteinemic mice and in males with coronary artery disease. Circ. Res. 98(4), 564–571 (2006).
    • 49. Liao D, Tan H, Hui R et al. Hyperhomocysteinemia decreases circulating high-density lipoprotein by inhibiting apolipoprotein A-I protein synthesis and enhancing HDL cholesterol clearance. Circ. Res. 99(6), 598–606 (2006).
    • 50. Van der PA, Van Gilst WH, Voors AA et al. Treating oxidative stress in heart failure: past, present and future. Eur. J. Heart Fail. 21(4), 425–435 (2019).
    • 51. Liguori I, Russo G, Curcio F et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 13, 757–772 (2018).
    • 52. Richard E, Gallego-Villar L, Rivera-Barahona A et al. Altered redox homeostasis in branched-chain amino acid disorders, organic acidurias, and homocystinuria. Oxid. Med. Cell. Longev. 2018, 1246069 (2018).
    • 53. Ji X, YU H, Zhou G et al. Physcion, a tetra-substituted 9,10-anthraquinone, prevents homocysteine-induced endothelial dysfunction by activating Ca2+- and Akt-eNOS-NO signaling pathways. Phytomedicine 81, 153410 (2021).
    • 54. Esse R, Barroso M, de-Almeida IT, Castro R. The contribution of homocysteine metabolism disruption to endothelial dysfunction: state-of-the-art. Int. J. Mol. Sci. 20(4), 867 (2019).
    • 55. Lai W, Kan M. Homocysteine-induced endothelial dysfunction. Ann. Nutr. Metab. 67(1), 1–12 (2015).
    • 56. Suvorava T, Metry S, Pick S, Kojda G. Alterations in endothelial nitric oxide synthase activity and their relevance to blood pressure. Biochem. Pharmacol. 205, 115256 (2022).
    • 57. Singh J, Lee Y, Kellum JA. A new perspective on NO pathway in sepsis and ADMA lowering as a potential therapeutic approach. Crit. Care 26(1), 246 (2022).
    • 58. Işıklar OO, Barutcuoğlu B, Kabaroğlu C et al. Do cardiac risk factors affect the homocysteine and asymmetric dimethylarginine relationship in patients with coronary artery diseases? Clin. Biochem. 45(16–17), 1325–1330 (2012).
    • 59. Zhang J, Liu J, Li Z, Wang L, Jiang Y, Wang S. Dysfunction of endothelial NO system originated from homocysteine-induced aberrant methylation pattern in promoter region of DDAH2 gene. Chin. Med. J. 120(23), 2132–2137 (2007).
    • 60. Zhao Z, Liu X, Shi S et al. Exogenous hydrogen sulfide protects from endothelial cell damage, platelet activation, and neutrophils extracellular traps formation in hyperhomocysteinemia rats. Exp. Cell Res. 370(2), 434–443 (2018).
    • 61. Kumar M, Sandhir R. Hydrogen sulfide attenuates hyperhomocysteinemia-induced blood-brain barrier permeability by inhibiting MMP-9. Int. J. Neurosci. 132(11), 1061–1071 (2022).
    • 62. Han L, Miao Y, Zhao Y et al. The binding of autotaxin to integrins mediates hyperhomocysteinemia-potentiated platelet activation and thrombosis in mice and humans. Blood Adv. 6(1), 46–61 (2022).
    • 63. Tsai JC, Wang H, Perrella MA et al. Induction of cyclin A gene expression by homocysteine in vascular smooth muscle cells. J. Clin. Investig. 97(1), 146–153 (1996).
    • 64. Han X, Zhang H, Cao C et al. Aberrant DNA methylation of the PDGF gene in homocysteinemediated VSMC proliferation and its underlying mechanism. Mol. Med. Rep. 10(2), 947–954 (2014).
    • 65. Xu L, Hao H, Hao Y et al. Aberrant MFN2 transcription facilitates homocysteine-induced VSMCs proliferation via the increased binding of c-Myc to DNMT1 in atherosclerosis. J. Cell. Mol. Med. 23(7), 4611–4626 (2019).
    • 66. Blachier F, Andriamihaja M, Blais A. Sulfur-containing amino acids and lipid metabolism. J. Nutr. 150(Suppl. 1), S2524–S2531 (2020).
    • 67. Visram M, Radulovic M, Steiner S et al. Homocysteine regulates fatty acid and lipid metabolism in yeast. J. Biol. Chem. 293(15), 5544–5555 (2018).
    • 68. Wang X, Zhang Y, Ma H. Targeted profiling of amino acid metabolome in serum by a liquid chromatography–mass spectrometry method: application to identify potential markers for diet-induced hyperlipidemia. Anal. Methods 12(18), 2355–2362 (2020).
    • 69. Velez-Carrasco W, Merkel M, Twiss CO, Smith JD. Dietary methionine effects on plasma homocysteine and HDL metabolism in mice. J. Nutr. Biochem. 19(6), 362–370 (2008).
    • 70. Liu Y, Tian T, Zhang H, Gao L, Zhou X. The effect of homocystein-lowering therapy with folic acid on flow-mediated vasodilation in patients with coronary artery disease: a meta-analysis of randomized controlled trials. Atherosclerosis 235(1), 31–35 (2014).
    • 71. Li Y. Modern epigenetics methods in biological research. Methods 187, 104–113 (2021).
    • 72. Mattei AL, Bailly N, Meissner A. DNA methylation: a historical perspective. Trends Genet. 38(7), 676–707 (2022).
    • 73. Ram O, Goren A, Amit I et al. Combinatorial patterning of chromatin regulators uncovered by genome-wide location analysis in human cells. Cell 147, 1628–1639 (2011).
    • 74. Yun J, Jialal I, Devaraj S. Epigenetic regulation of high glucoseinduced proinflammatory cytokine production in monocytes by curcumin. J. Nutr. Biochem. 22(5), 450–458 (2011).
    • 75. Reddy MA, Natarajan R. Epigenetic mechanisms in diabetic vascular complications. Cardiovasc. Res. 90(3), 421–429 (2011).
    • 76. Dozmorov MG, Giles CB, Koelsch KA, Wren JD. Systematic classification of non-coding RNAs by epigenomic similarity. BMC Bioinform. 14(Suppl. 14), S2 (2013).
    • 77. Chen L, Heikkinen L, Wang C, Yang Y, Sun H, Wong G. Trends in the development of miRNA bioinformatics tools. Brief. Bioinformatics 20(5), 1836–1852 (2019).
    • 78. Bridges MC, Daulagala AC, Kourtidis A. LNCcation: lncRNA localization and function. J. Cell Biol. 220(2), e202009045 (2021).
    • 79. Chen L, Wang C, Sun H et al. The bioinformatics toolbox for circRNA discovery and analysis. Brief. Bioinformatics 22(2), 1706–1728 (2021).
    • 80. Rafnar T, Sulem P, Thorleifsson G et al. Genome-wide association study yields variants at 20p12.2 that associate with urinary bladder cancer. Hum. Mol. Genet. 23(20), 5545–5557 (2014).
    • 81. Heshmatzad K, Naderi N, Maleki M et al. Role of non-coding variants in cardiovascular disease. J. Cell. Mol. Med. 27(12), 1621–1636 (2023).
    • 82. Dong S, Zhang Y, Chen Y et al. Comprehensive review and annotation of susceptibility SNPs associated with obesity-related traits. Obes. Rev. 19(7), 917–930 (2018).
    • 83. Rojano E, Ranea JA, Perkins JR. Characterisation of non-coding genetic variation in histamine receptors using AnNCR-SNP. Amino Acids 48(10), 2433–2442 (2016).
    • 84. Gate RE, Cheng CS, Aiden AP et al. Genetic determinants of co-accessible chromatin regions in activated T cells across humans. Nat. Genet. 50(8), 1140–1150 (2018).
    • 85. Loscalzo J, Handy DE. Epigenetic modifications: basic mechanisms and role in cardiovascular disease (2013 Grover Conference series). Pulm. Circ. 4(2), 169–174 (2014).
    • 86. Pogribny IP, Karpf AR, James SR, Melnyk S, Han T, Tryndyak VP. Epigenetic alterations in the brains of Fisher 344 rats induced by long-term administration of folate/methyl-deficient diet. Brain Res. 1237, 25–34 (2008). • It is widely believed that homocysteine induces the reduction of the SAM/SAH ratio, which in turn induces DNA hypomethylation and promotes the occurrence of the disease. However, this article shows that this is not absolute and the association between SAM/SAH and gene-specific DNA methylation is tissue-specific.
    • 87. Glier MB, Ngai YF, Sulistyoningrum DC, Aleliunas RE, Bottiglieri T, Devlin AM. Tissue-specific relationship of s-adenosylhomocysteine with allele-specific H19/IGF2 methylation and imprinting in mice with hyperhomocysteinemia. Epigenetics 8(1), 44–53 (2013).
    • 88. Esse R, Imbard A, Florindo C et al. Protein arginine hypomethylation in a mouse model of cystathionine beta-synthase deficiency. FASEB J. 28(6), 2686–2695 (2014).
    • 89. Lee H, Wang L, Kuo Y et al. Lack of global epigenetic methylation defects in CBS deficient mice. J. Inherit. Metab. Dis. 40(1), 113–120 (2017).
    • 90. Fu Y, Wang X, Kong W. Hyperhomocysteinaemia and vascular injury: advances in mechanisms and drug targets. Br. J. Pharmacol. 175(8), 1173–1189 (2018).
    • 91. Wang W, Jin H. Homocysteine: a potential common route for cardiovascular risk and DNA methylation in psoriasis. Chin. Med. J. 130(16), 1980–1986 (2017).
    • 92. Schumann NAB, Mendonça AS, Silveira MM et al. Procaine and S-adenosyl-l-homocysteine affect the expression of genes related to the epigenetic machinery and change the DNA methylation status of in vitro cultured bovine skin fibroblasts. DNA Cell Biol. 39(1), 37–49 (2020).
    • 93. Zhang D, Chen Y, Xie X et al. Homocysteine activates vascular smooth muscle cells by DNA demethylation of platelet-derived growth factor in endothelial cells. J. Mol. Cell. Cardiol. 53(4), 487–496 (2012).
    • 94. Jiang Y, Jiang J, Xiong J et al. Homocysteine-induced extracellular superoxide dismutase and its epigenetic mechanisms in monocytes. J. Exp. Biol. 211(Pt 6), 911–920 (2008).
    • 95. Li J, Barrero C, Gupta S, Kruger WD, Merali S, Praticò D. Homocysteine modulates 5-lipoxygenase expression level via DNA methylation. Aging Cell 16(2), 273–280 (2017).
    • 96. Ma S, Hao Y, Jiao Y et al. Homocysteine-induced oxidative stress througH TLR4/NF-ΚB/DNMT1-mediated LOX-1 DNA methylation in endothelial cells. Mol. Med. Rep. 16(6), 9181–9188 (2017).
    • 97. Wu H, Li Z, Yang Y et al. Rap1A accelerates homocysteine-induced ANA-1 cells inflammation via synergy of FoxO1 and DNMT3a. Cell. Signal. 106, 110627 (2023).
    • 98. Xu L, Hao H, Hao Y et al. Aberrant MFN2 transcription facilitates homocysteine-induced VSMCs proliferation via the increased binding of c-Myc to DNMT1 in atherosclerosis. J. Cell. Mol. Med. 23(7), 4611–4626 (2019).
    • 99. Guo W, Zhang H, Yang A et al. Homocysteine accelerates atherosclerosis by inhibiting scavenger receptor class B member1 via DNMT3b/SP1 pathway. J. Mol. Cell. Cardiol. 138, 34–48 (2020).
    • 100. Wang H, Yoshizumi M, Lai K et al. Inhibition of growth and p21RAS methylation in vascular endothelial cells by homocysteine but not cysteine. J. Biol. Chem. 272(40), 25380–25385 (1997).
    • 101. Gou Y, Ye Q, Liang X et al. Homocysteine restrains hippocampal neurogenesis in focal ischemic rat brain by inhibiting DNA methylation. Neurochem. Int. 147, 105065 (2021).
    • 102. Yang Z, Wang L, Zhang W, Wang X, Zhou S. Plasma homocysteine involved in methylation and expression of thrombomodulin in cerebral infarction. Biochem. Biophys. Res. Commun. 473(4), 1218–1222 (2016).
    • 103. Pushpakumar S, Kundu S, Narayanan N, Sen U. DNA hypermethylation in hyperhomocysteinemia contributes to abnormal extracellular matrix metabolism in the kidney. FASEB J. 29(11), 4713–4725 (2015).
    • 104. Ding N, Xie L, Ma F et al. miR-30a-5p promotes glomerular podocyte apoptosis via DNMT1-mediated hypermethylation under hyperhomocysteinemia. Acta. Biochim. Biophys. Sin. 54(1), 126–136 (2022).
    • 105. Xie L, Ma S, Ding N et al. Homocysteine induces podocyte apoptosis by regulating miR-1929-5p expression through c-Myc, DNMT1 and EZH2. Mol. Oncol. 15(11), 3203–3221 (2021).
    • 106. Scarpa S, Fuso A, D’Anselmi F, Cavallaro RA. Presenilin 1 gene silencing by S-adenosylmethionine: a treatment for Alzheimer disease? FEBS Lett. 541(1–3), 145–148 (2003).
    • 107. Pi T, Lang G, Liu B, Shi J. Protective effects of Dendrobium nobile Lindl. alkaloids on Alzheimer’s disease-like symptoms induced by high-methionine diet. Curr. Neuropharmacol. 20(5), 983–997 (2022).
    • 108. Zeng P, Shi Y, Wang X et al. Emodin rescued hyperhomocysteinemia-induced dementia and Alzheimer’s disease-like features in rats. Int. J. Neuropsychopharmacol. 22(1), 57–70 (2019).
    • 109. Wang S, Wang X, Zhao Y et al. Homocysteine-induced disturbances in DNA methylation contribute to development of stress-associated cognitive decline in rats. Neurosci. Bull. 38(8), 887–900 (2022).
    • 110. Shen J, Jiao Y, Ding N et al. Homocysteine facilitates endoplasmic reticulum stress and apoptosis of hepatocytes by suppressing ERO1α expression via cooperation between DNMT1 and G9a. Cell Biol. Int. 46(8), 1236–1248 (2022).
    • 111. Yang A, Zeng W, Zhang H et al. Homocysteine accelerates hepatocyte autophagy by upregulating TFEB via DNMT3b-mediated DNA hypomethylation. Acta Biochim. Biophys. Sin. 55(8), 1184–1192 (2023).
    • 112. Kowluru RA. Diabetic retinopathy: mitochondria caught in a muddle of homocysteine. J. Clin. Med. 9(9), 3019 (2020).
    • 113. Mohammad G, Kowluru RA. Homocysteine disrupts balance between MMP-9 and its tissue inhibitor in diabetic retinopathy: the role of DNA methylation. Int. J. Mol. Sci. 21(5), 1771 (2020).
    • 114. Tao H, Shi P, Xuan H, Ding X. DNA methyltransferase-1 inactivation of androgen receptor axis triggers homocysteine induced cardiac fibroblast autophagy in diabetic cardiac fibrosis. Arch. Biochem. Biophys. 692, 108521 (2020).
    • 115. Zhang D, Lou J, Zhang X et al. Hyperhomocysteinemia results from and promotes hepatocellular carcinoma via CYP450 metabolism by CYP2J2 DNA methylation. Oncotarget 8(9), 15377–15392 (2017).
    • 116. Naushad SM, Reddy CA, Kumaraswami K et al. Impact of hyperhomocysteinemia on breast cancer initiation and progression: epigenetic perspective. Cell Biochem. Biophys. 68(2), 397–406 (2014).
    • 117. Zhang H, Wang Y, Ma S et al. Homocysteine inhibits endothelial progenitor cells proliferation via DNMT1-mediated hypomethylation of cyclin A. Exp. Cell Res. 362(1), 217–226 (2018).
    • 118. Xiao Y, Xia J, Cheng J et al. Inhibition of S-adenosylhomocysteine hydrolase induces endothelial dysfunction via epigenetic regulation of p66shc-mediated oxidative stress pathway. Circulation 139(19), 2260–2277 (2019).
    • 119. Jamaluddin MS, Yang X, Wang H. Hyperhomocysteinemia, DNA methylation and vascular disease. Clin. Chem. Lab. Med. 45(12), 1660–1666 (2007).
    • 120. Esse R, Teerlink T, Koolwijk P et al. Folinic acid increases protein arginine methylation in human endothelial cells. Nutrients 10(4), 404 (2018).
    • 121. Ma S, Lu G, Zhang Q et al. Extracellular-superoxide dismutase DNA methylation promotes oxidative stress in homocysteine-induced atherosclerosis. Acta Biochim. Biophys. Sin. 54(9), 1222–1233 (2022).
    • 122. Feng P, Liang Y, Lin W et al. Homocysteine induced oxidative stress in human umbilical vein endothelial cells via regulating methylation of SORBS1. Eur. Rev. Med. Pharmacol. Sci. 22(20), 6948–6958 (2018).
    • 123. Wei L, Chao N, Gao S et al. Homocysteine induces vascular inflammatory response via SMAD7 hypermethylation in human umbilical vein smooth muscle cells. Microvasc. Res. 120, 8–12 (2018).
    • 124. Li L, Xie J, Zhang M, Wang S. Homocysteine harasses the imprinting expression of IGF2 and H19 by demethylation of differentially methylated region between IGF2/H19 genes. Acta Biochim. Biophys. Sin. 41(6), 464–471 (2009).
    • 125. Zhang D, Wen X, Wu W, Xu E, Zhang Y, Cui W. Homocysteine-related HTERT DNA demethylation contributes to shortened leukocyte telomere length in atherosclerosis. Atherosclerosis 231(1), 173–179 (2013).
    • 126. Spence JD. Nutrition and risk of stroke. Nutrients 11(3), 647 (2019).
    • 127. Wang C, Xu G, Wen Q et al. CBS promoter hypermethylation increases the risk of hypertension and stroke. Clinics (Sao Paulo) 74, e630 (2019).
    • 128. Zhang D, Wen X, Zhang L, Cui W. DNA methylation of human telomerase reverse transcriptase associated with leukocyte telomere length shortening in hyperhomocysteinemia-type hypertension in humans and in a rat model. Circ. J. 78(8), 1915–1923 (2014).
    • 129. Wan C, Zong R, Chen X. The new mechanism of cognitive decline induced by hypertension: high homocysteine-mediated aberrant DNA methylation. Front. Cardiovasc. Med. 9, 928701 (2022).
    • 130. Hu J, Zhu H, Xu G et al. Significant association between DHFR promoter methylation and ischemic stroke in a Chinese hypertensive population. J. Clin. Lab. Anal. 34(8), e23322 (2020).
    • 131. Li D, Zhao Q, Zhang C, Huang X, Godfrey O, Zhang W. Associations of MTRR A66G polymorphism and promoter methylation with ischemic stroke in patients with hyperhomocysteinemia. J. Gene Med. 22(5), e3170 (2020).
    • 132. Lin Y, Kong J, Tian T et al. Identification of novel phenotypes correlated with CKD: a phenotype-wide association study. Int. J. Med. Sci. 19(13), 1920–1928 (2022).
    • 133. Yang X, Cao R, Yu Y et al. A study on the correlation between MTHFR promoter methylation and diabetic nephropathy. Am. J. Transl. Res. 8(11), 4960–4967 (2016).
    • 134. Ingrosso D, Cimmino A, Perna AF et al. Folate treatment and unbalanced methylation and changes of allelic expression induced by hyperhomocysteinaemia in patients with uraemia. Lancet 361(9370), 1693–1699 (2003).
    • 135. Li J, Barrero C, Gupta S, Kruger WD, Merali S, Praticò D. Homocysteine modulates 5-lipoxygenase expression level via DNA methylation. Aging Cell 16(2), 273–280 (2017).
    • 136. Pi T, Liu B, Shi J. Abnormal homocysteine metabolism: an insight of Alzheimer’s disease from DNA methylation. Behav. Neurol. 2020, 8438602 (2020).
    • 137. Ouyang Y, Wu Q, Li J, Sun S, Sun S. S-adenosylmethionine: a metabolite critical to the regulation of autophagy. Cell Prolif. 53(11), e12891 (2020).
    • 138. Kowluru RA, Mohammad G, Sahajpal N. Faulty homocysteine recycling in diabetic retinopathy. Eye Vis. (Lond). 7, 4 (2020).
    • 139. Wu L, Wu J. Hyperhomocysteinemia is a risk factor for cancer and a new potential tumor marker. Clin. Chim. Acta 22(1–2), 21–28 (2002).
    • 140. Sullivan MR, Darnell AM, Reilly MF et al. Methionine synthase is essential for cancer cell proliferation in physiological folate environments. Nat. Metab. 3(11), 1500–1511 (2021).
    • 141. Sedillo JC, Cryns VL. Targeting the methionine addiction of cancer. Am. J. Cancer Res. 12(5), 2249–2276 (2022).
    • 142. Ding W, Zhou D, Jiang X, Lu L. Methionine synthase A2756G polymorphism and risk of colorectal adenoma and cancer: evidence based on 27 studies. PLoS ONE 8(4), e60508 (2013).
    • 143. Vantaku V, Amara CS, Piyarathna DWB et al. DNA methylation patterns in bladder tumors of African American patients point to distinct alterations in xenobiotic metabolism. Carcinogenesis 40(11), 1332–1340 (2019).
    • 144. Yadav S, Longkumer I, Garg PR et al. Association of air pollution and homocysteine with global DNA methylation: a population-based study from North India. PLoS ONE 16(12), e0260860 (2021).
    • 145. McCaddon A, Regland B. COVID-19: a methyl-group assault? Med. Hypotheses 149, 110543 (2021).
    • 146. Jia L, Zeng Y, Hu Y et al. Homocysteine impairs porcine oocyte quality via deregulation of one-carbon metabolism and hypermethylation of mitochondrial DNA. Biol. Reprod. 100(4), 907–916 (2019).
    • 147. Hoeijmakers L, Kempe H, Verschure PJ. Epigenetic imprinting during assisted reproductive technologies: the effect of temporal and cumulative fluctuations in methionine cycling on the DNA methylation state. Mol. Reprod. Dev. 83(2), 94–107 (2016).
    • 148. Stillman B. Histone modifications: insights into their influence on gene expression. Cell 175(1), 6–9 (2018).
    • 149. Demetriadou C, Koufaris C, Kirmizis A. Histone N-alpha terminal modifications: genome regulation at the tip of the tail. Epigenetics Chromatin 13(1), 29 (2020).
    • 150. Roth SY, Denu JM, Allis CD. Histone acetyltransferases. Annu. Rev. Biochem. 70, 81–120 (2001).
    • 151. Zhao Q, Li S, Li N et al. miR-34a targets HDAC1-regulated H3K9 acetylation on lipid accumulation induced by homocysteine in foam cells. J. Cell. Biochem. 118(12), 4617–4627 (2017).
    • 152. Xiong J, Ma F, Ding N et al. miR-195-3p alleviates homocysteine-mediated atherosclerosis by targeting IL-31 through its epigenetics modifications. Aging Cell 20(10), e13485 (2021).
    • 153. Qin X, Shan Y, Dou M, Li F, Guo Y. Notch1 signaling activation alleviates coronary microvascular dysfunction through histone modification of Nrg-1 via the interaction between NICD and GCN5. Apoptosis 28(1–2), 124–135 (2023).
    • 154. Elmasry K, Mohamed R, Sharma I et al. Epigenetic modifications in hyperhomocysteinemia: potential role in diabetic retinopathy and age-related macular degeneration. Oncotarget 9(16), 12562–12590 (2018).
    • 155. Tóthová B, Kovalská M, Kalenská D, Tomašcová A, Lehotský J. Histone hyperacetylation as a response to global brain ischemia associated with hyperhomocysteinemia in rats. Int. J. Mol. Sci. 19(10), 3147 (2018).
    • 156. Won SB, Han A, Kwon YH. Maternal consumption of low-isoflavone soy protein isolate alters hepatic gene expression and liver development in rat offspring. J. Nutr. Biochem. 42, 51–61 (2017).
    • 157. Greer EL, Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 13(5), 343–357 (2012).
    • 158. Cong G, Yan R, Huang H et al. Involvement of histone methylation in macrophage apoptosis and unstable plaque formation in methionine-induced hyperhomocysteinemic ApoE-/- mice. Life Sci. 173, 135–144 (2017).
    • 159. Xie L, Ding N, Zhang H et al. SNF5 promotes IL-1β expression via H3K4me1 in atherosclerosis induced by homocysteine. Int. J. Biochem. Cell Biol. 135, 105974 (2021).
    • 160. Whalen CA, Mattie FJ, Florindo C et al. No effect of diet-induced mild hyperhomocysteinemia on vascular methylating capacity, atherosclerosis progression, and specific histone methylation. Nutrients 12(8), 2182 (2020).
    • 161. Yang A, Jiao Y, Yang S et al. Homocysteine activates autophagy by inhibition of CFTR expression via interaction between DNA methylation and H3K27me3 in mouse liver. Cell Death Dis. 9(2), 169 (2018).
    • 162. Aoki Y, Tome Y, Han Q et al. Histone H3 lysine-trimethylation markers are decreased by recombinant methioninase and increased by methotrexate at concentrations which inhibit methionine-addicted osteosarcoma cell proliferation. Biochem. Biophys. Rep. 28, 101177 (2021).
    • 163. Cunningham A, Erdem A, Alshamleh I et al. Dietary methionine starvation impairs acute myeloid leukemia progression. Blood 140(19), 2037–2052 (2022).
    • 164. Jan M, Cueto R, Jiang X et al. Molecular processes mediating hyperhomocysteinemia-induced metabolic reprogramming, redox regulation and growth inhibition in endothelial cells. Redox. Biol. 45, 102018 (2021).
    • 165. Qin X, Shan Y, Gao J, Li F, Guo Y. E3 ubiquitin ligase mind bomb 1 overexpression reduces apoptosis and inflammation of cardiac microvascular endothelial cells in coronary microvascular dysfunction. Cell. Signal. 91, 110223 (2022).
    • 166. Sun W, Wang X, Novakovic A, Wang J, He G, Yang Q. Protection of dilator function of coronary arteries from homocysteine by tetramethylpyrazine: role of ER stress in modulation of BKCa channels. Vascul. Pharmacol. 113, 27–37 (2019).
    • 167. Derouiche F, Bôle-Feysot C, Naïmi D, Coëffier M. Hyperhomocysteinemia-induced oxidative stress differentially alters proteasome composition and activities in heart and aorta. Biochem. Biophys. Res. Commun. 452(3), 740–745 (2014).
    • 168. Wang L, Lai G, Chu G, Liang X, Zhao Y. cMyBP-C was decreased via KLHL3-mediated proteasomal degradation in congenital heart diseases. Exp. Cell Res. 355(1), 18–25 (2017).
    • 169. Tripathi M, Singh BK, Zhou J et al. Vitamin B12 and folate decrease inflammation and fibrosis in NASH by preventing syntaxin 17 homocysteinylation. J. Hepatol. 77(5), 1246–1255 (2022).
    • 170. Xiang W, Yang Y, Weng L et al. Hyperhomocysteinemia activates NLRP3 inflammasome to cause hepatic steatosis and insulin resistance via MDM2-mediated ubiquitination of HSF1. Int. Immunopharmacol. 118, 110085 (2023).
    • 171. Tian X, Gong L, Jin A, Wang Y, Zhou X, Tan Y. E3 ubiquitin ligase SIAH-1 nuclear accumulation is critical for homocysteine induced impairment of C6 astroglioma cells. Mol. Med. Rep. 20(3), 2227–2235 (2019).
    • 172. Bretes E, Zimny J. Homocysteine thiolactone affects protein ubiquitination in yeast. Acta Biochim. Pol. 60(3), 485–488 (2013).
    • 173. Rossetto D, Avvakumov N, Côté J. Histone phosphorylation: a chromatin modification involved in diverse nuclear events. Epigenetics 7(10), 1098–1108 (2012).
    • 174. Lowndes NF, Toh GW. DNA repair: the importance of phosphorylating histone H2AX. Curr. Biol. 15(3), R99–R102 (2005).
    • 175. Palmer AM, Kamynina E, Field MS, Stover PJ. Folate rescues vitamin B12 depletion-induced inhibition of nuclear thymidylate biosynthesis and genome instability. Proc. Natl Acad. Sci. USA 114(20), E4095–E4102 (2017).
    • 176. Siddiqui MS, Francois M, Hecker J, Faunt J, Fenech MF, Leifert WR. γH2AX is increased in peripheral blood lymphocytes of Alzheimer’s disease patients in the South Australian Neurodegeneration, Nutrition and DNA Damage (SAND) study of aging. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 829–830, 6–18 (2018).
    • 177. Clapier CR, Iwasa J, Cairns BR. Mechanisms of action and regulation of ATP-dependent chromatin-remodelling complexes. Nat. Rev. Mol. Cell Biol. 18(7), 407–422 (2017).
    • 178. Mayes K, Qiu Z, Alhazmi A, Landry JW. ATP-dependent chromatin remodeling complexes as novel targets for cancer therapy. Adv. Cancer Res. 121, 183–233 (2014).
    • 179. Dechassa ML, Sabri A, Pondugula S et al. SWI/SNF has intrinsic nucleosome disassembly activity that is dependent on adjacent nucleosomes. Mol. Cell 38, 590–602 (2010).
    • 180. Clapier CR, Kasten MM, Parnell TJ et al. Regulation of DNA translocation efficiency within the chromatin remodeler RSC/Sth1 potentiates nucleosome sliding and ejection. Mol. Cell 62, 453–461 (2016).
    • 181. Papamichos-Chronakis M, Watanabe S, Rando OJ, Peterson CL. Global regulation of H2A.Z localization by the INO80 chromatin-remodeling enzyme is essential for genome integrity. Cell 144, 200–213 (2011).
    • 182. Brahma S, Udugama MI, Kim J et al. INO80 exchanges H2A.Z for H2A by translocating on DNA proximal to histone dimers. Nat. Commun. 8, 15616 (2017).
    • 183. Eustermann S, Schall K, Kostrewa D et al. Structural basis for ATP-dependent chromatin remodelling by the INO80 complex. Nature 556, 386–390 (2018).
    • 184. Hasan N, Ahuja N. The emerging roles of ATP-dependent chromatin remodeling complexes in pancreatic cancer. Cancers (Basel) 11(12), 1859 (2019).
    • 185. Quan J, Yusufzai T. The tumor suppressor chromodomain helicase DNA-binding protein 5 (CHD5) remodels nucleosomes by unwrapping. J. Biol. Chem. 289, 20717–201726 (2014).
    • 186. Zhang H, Wang Y, Cao C et al. A regulatory circuit involving miR-143 and DNMT3a mediates vascular smooth muscle cell proliferation induced by homocysteine. Mol. Med. Rep. 13(1), 483–490 (2016).
    • 187. Zhang M, Li F, Wang X et al. MiR-145 alleviates Hcy-induced VSMC proliferation, migration, and phenotypic switch through repression of the PI3K/Akt/mTOR pathway. Histochem. Cell Biol. 153(5), 357–366 (2020).
    • 188. Li Y, Chen F, Guo R, Jia S, Li W, Zhang B. Tanshinone IIA inhibits homocysteine-induced proliferation of vascular smooth muscle cells via miR-145/CD40 signaling. Biochem. Biophys. Res. Commun. 522(1), 157–163 (2020).
    • 189. Liu K, Xuekelati S, Zhang Y et al. Expression levels of atherosclerosis-associated miR-143 and miR-145 in the plasma of patients with hyperhomocysteinaemia. BMC Cardiovasc. Disord. 17(1), 163 (2017).
    • 190. Yan P, Sun C, Ma J, Jin Z, Guo R, Yang B. MicroRNA-128 confers protection against cardiac microvascular endothelial cell injury in coronary heart disease via negative regulation of IRS1. J. Cell. Physiol. 234(8), 13452–13463 (2019).
    • 191. Griñán R, Escolà-Gil JC, Julve J, Benítez S, Rotllan N. Epigenetic regulation by microRNAs in hyperhomocysteinemia-accelerated atherosclerosis. Int. J. Mol. Sci. 23(20), 12452 (2022).
    • 192. Xie L, Ma S, Ding N et al. Homocysteine induces podocyte apoptosis by regulating miR-1929-5p expression through c-Myc, DNMT1 and EZH2. Mol. Oncol. 15(11), 3203–3221 (2021).
    • 193. Jie Y, Ding N, Xie L et al. [Homocysteine induces glomerular podocyte apoptosis via up-regulation of miR-488-3p expression in MPC-5 mice]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 38(9), 801–806 (2022).
    • 194. Li H, Liu M, Yang W et al. Naringenin induces neuroprotection against homocysteine-induced PC12 cells via the upregulation of superoxide dismutase 1 expression by decreasing miR-224-3p expression. J. Biol. Regul. Homeost. Agents 34(2), 421–433 (2020).
    • 195. Veeranki S, Winchester LJ, Tyagi SC. Hyperhomocysteinemia associated skeletal muscle weakness involves mitochondrial dysfunction and epigenetic modifications. Biochim. Biophys. Acta 1852(5), 732–741 (2015).
    • 196. Wang R, Han Z, Song G, Cui Y, Xia H, Ma X. Homocysteine-induced neural tube defects in chick embryos via oxidative stress and DNA methylation associated transcriptional down-regulation of miR-124. Toxicol. Res. 10(3), 425–435 (2021).
    • 197. Arutjunyan AV, Milyutina YP, Shcherbitskaia AD, Kerkeshko GO, Zalozniaia IV. Epigenetic mechanisms involved in the effects of maternal hyperhomocysteinemia on the functional state of placenta and nervous system plasticity in the offspring. Biochemistry 88(4), 435–456 (2023).
    • 198. An F, Yin Y, Ju W. Long noncoding RNA DANCR expression and its predictive value in patients with atherosclerosis. Bioengineered 13(3), 6919–6928 (2022).
    • 199. Safaei S, Tahmasebi-Birgani M, Bijanzadeh M, Seyedian SM. Increased expression level of long noncoding RNA H19 in plasma of patients with myocardial infarction. Int. J. Mol. Med. 9(2), 122–129 (2020).
    • 200. Huang J, Yang J, Li J et al. Association of long noncoding RNA H19 polymorphisms with the susceptibility and clinical features of ischemic stroke in southern Chinese Han population. Metab. Brain Dis. 34(4), 1011–1021 (2019).
    • 201. Li J, Luo M, Xie N, Wang H, Wang J. Bioinformatics-based analysis of the involvement of AC005550.3, RP11-415D17.3, and RP1-140K8.5 in homocysteine-induced vascular endothelial injury. Am. J. Transl. Res. 10(7), 2126–2136 (2018).
    • 202. Li W, Li Y, Cui S et al. Se alleviates homocysteine-induced fibrosis in cardiac fibroblasts via downregulation of lncRNA MEG3. Exp. Ther. Med. 22(5), 1269 (2021).
    • 203. Diao L, Bai L, Jiang X, Li J, Zhang Q. Long-chain noncoding RNA GAS5 mediates oxidative stress in cardiac microvascular endothelial cells injury. J. Cell. Physiol. 234(10), 17649–17662 (2019).
    • 204. Gu L, Xu L, Yang A, Jiao Y, Zhang H, Jiang Y. [Long non-coding RNA growth arrest specific transcript 5 (lncGAS5) promotes autophagy of hepatocytes induced by homocysteine]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 37(3), 240–245 (2021).
    • 205. Zheng M, Zheng Y, Gao M et al. Expression and clinical value of lncRNA MALAT1 and lncRNA ANRIL in glaucoma patients. Exp. Ther. Med. 19(2), 1329–1335 (2020).
    • 206. Ding S, Zhu Y, Liang Y. Circular RNAs in vascular functions and diseases. Adv. Exp. Med. Biol. 1087, 287–297 (2018).
    • 207. Singh M, George AK, Homme RP et al. Circular RNAs profiling in the cystathionine-β-synthase mutant mouse reveals novel gene targets for hyperhomocysteinemia induced ocular disorders. Exp. Eye Res. 174, 80–92 (2018).
    • 208. Singh M, George AK, Homme RP et al. Expression analysis of the circular RNA molecules in the human retinal cells treated with homocysteine. Curr. Eye Res. 44(3), 287–293 (2019).
    • 209. Eyob W, George AK, Homme RP et al. Regulation of the parental gene GRM4 by circGrm4 RNA transcript and glutamate-mediated neurovascular toxicity in eyes. Mol. Cell. Biochem. 476(2), 663–673 (2021).
    • 210. Hu F, Sha W, Dai H et al. Lower expression of Hsa_circRNA_102682 in diabetic hyperhomocysteinemia negatively related to creatinemia is associated with TGF-β and CTGF. J. Clin. Lab. Anal. 35(8), e23860 (2021).
    • 211. Gao L, Wu Q, Yin H et al. [The nuclear translocation of circRNA CPSF6 promotes apoptosis of trophoblast cells induced by homocysteine]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 38(2), 146–152 (2022).
    • 212. Sharma GS, Singh LR. Conformational status of cytochrome C upon N-homocysteinylation: implications to cytochrome C release. Arch. Biochem. Biophys. 614, 23–27 (2017).
    • 213. Gurda D, Handschuh L, Kotkowiak W, Jakubowski H. Homocysteine thiolactone and N-homocysteinylated protein induce pro-atherogenic changes in gene expression in human vascular endothelial cells. Amino Acids 47(7), 1319–1339 (2015).
    • 214. Moshtaghie E, Nayeri H, Moshtaghie AA, Asgary S. The effect of homocysteine thiolactone on paraoxonase and aryl esterase activity of human serum purified paraoxonase 1 in vitro experiments. ARYA Atheroscler. 18(2), 1–6 (2022).
    • 215. Genoud V, Quintana PG, Gionco S, Baldessari A, Quintana I. Structural changes of fibrinogen molecule mediated by the N-homocysteinylation reaction. J. Thromb. Thrombolysis 45(1), 66–76 (2018).
    • 216. Xu L, Chen J, Gao J, Yu H, Yang P. Crosstalk of homocysteinylation, methylation and acetylation on histone H3. Analyst 140(9), 3057–3063 (2015).
    • 217. AnandBabu K, Sen P, Angayarkanni N. Oxidized LDL, homocysteine, homocysteine thiolactone and advanced glycation end products act as pro-oxidant metabolites inducing cytokine release, macrophage infiltration and pro-angiogenic effect in ARPE-19 cells. PLoS ONE 14(5), e0216899 (2019).
    • 218. Jakubowski H. Homocysteine modification in protein structure/function and human disease. Physiol. Rev. 99(1), 555–604 (2019).
    • 219. Jakubowsk H, Boers GH, Strauss KA. Mutations in cystathionine beta-synthase or methylenetetrahydrofolate reductase gene increase N-homocysteinylated protein levels in humans. FASEB J. 22(12), 4071–4076 (2008).
    • 220. Kameda T, Horiuchi Y, Shimano S et al. Effect of myeloperoxidase oxidation and N-homocysteinylation of high-density lipoprotein on endothelial repair function. J. Biol. Chem. 403(3), 265–277 (2021).
    • 221. Bossenmeyer-Pourié C, Smith AD, Lehmann S et al. N-homocysteinylation of tau and MAP1 is increased in autopsy specimens of Alzheimer’s disease and vascular dementia. J. Pathol. 248(3), 291–303 (2019).
    • 222. Jakubowski H. Homocysteine modification in protein structure/function and human disease. Physiol. Rev. 99(1), 555–604 (2019).
    • 223. Zhang Q, Bai B, Mei X et al. Elevated H3K79 homocysteinylation causes abnormal gene expression during neural development and subsequent neural tube defects. Nat. Commun. 9(1), 3436 (2018).
    • 224. Gu Y, Zhang Q, Guo J et al. Higher serum homocysteine and lower thyroid hormone levels in pregnant women are associated with neural tube defects. J. Trace Elem. Med. Biol. 68, 126814 (2021).
    • 225. Yu Y, Jia C, Shi Q, Zhu Y, Liu Y. Hyperhomocysteinemia in men with a reproductive history of fetal neural tube defects: three case reports and literature review. Medicine (Baltimore) 98(2), e13998 (2019).
    • 226. Genoud V, Quintana PG, Gionco S, Baldessari A, Quintana I. Structural changes of fibrinogen molecule mediated by the N-homocysteinylation reaction. J. Thromb. Thrombolysis 45(1), 66–76 (2018).
    • 227. Ma S, Zhang H, Jiao Y et al. Homocysteine-induced proliferation of vascular smooth muscle cells occurs via PTEN hypermethylation and is mitigated by Resveratrol. Mol. Med. Rep. 17(4), 5312–5319 (2018).
    • 228. Carrizzo A, Iside C, Nebbioso A et al. SIRT1 pharmacological activation rescues vascular dysfunction and prevents thrombosis in MTHFR deficiency. Cell. Mol. Life Sci. 79(8), 410 (2022).
    • 229. Xiang Y, Liang B, Zhang X et al. Atheroprotective mechanism by which folic acid regulates monocyte subsets and function through DNA methylation. Clin. Epigenetics 14(1), 32 (2022).
    • 230. Pizzolo F, Blom HJ, Choi SW et al. Folic acid effects on s-adenosylmethionine, s-adenosylhomocysteine, and DNA methylation in patients with intermediate hyperhomocysteinemia. Am. J. Coll. Nutr. 30(1), 11–18 (2011).
    • 231. Zhou S, Zhang Z, Xu G. Notable epigenetic role of hyperhomocysteinemia in atherogenesis. Lipids Health Dis. 13, 134 (2014).
    • 232. Rysz J, Franczyk B, Rysz-Górzyńska M, Gluba-Brzózka A. Are alterations in DNA methylation related to CKD development? Int. J. Mol. Sci. 23(13), 7108 (2022).
    • 233. Capelli I, Cianciolo G, Gasperoni L et al. Folic acid and vitamin B12 administration in CKD, why not? Nutrients 11(2), 383 (2019).