Skip to main content

Advertisement

Log in

Trimethylamine N-oxide promotes oxidative stress and lipid accumulation in macrophage foam cells via the Nrf2/ABCA1 pathway

  • Original Article
  • Published:
Journal of Physiology and Biochemistry Aims and scope Submit manuscript

Abstract

Recently, trimethylamine N-oxide (TMAO) has been considered a risk factor for cardiovascular disease and has a proatherogenic effect. Many studies have found that TMAO is involved in plaque oxidative stress and lipid metabolism, but the specific mechanism is still unclear. In our study, meta-analysis and bioinformatic analysis were firstly conducted in the database, and found that the effect of high plasma TMAO levels on promoting atherosclerotic plaque may be related to the expression of key antioxidant genes nuclear factor erytheroid-derived-2-like 2 (NFE2L2/Nrf2) decreased. Next, we assessed the role of Nrf2-mediated signaling pathway in TMAO-treated foam cells. Our results showed that TMAO can inhibit the expression of Nrf2 and its downstream antioxidant response element such as heme oxygenase-1 (HO-1) and glutathione peroxidase4 (GPX4), resulting in increased production of reactive oxygen species and decreased activity of superoxide dismutase, promoting oxidative stress. And TMAO can also promote lipid accumulation in foam cells by inhibiting cholesterol efflux protein expression. In addition, upregulation of Nrf2 expression partially rescues TMAO-induced oxidative stress and reduces ATP-binding cassette A1 (ABCA1)–mediated lipid accumulation. Therefore, TMAO promotes oxidative stress and lipid accumulation in macrophage foam cells through the Nrf2/ABCA1 pathway, which may provide a potential mechanism for the proatherogenic effect of TMAO.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Data availability

All data used to support the current study are available from the corresponding author on reasonable request. The links for the database included in the study are as follows: GEO database (http://www.ncbi.nlm.nih.gov/geo); Genecard database (http://www.genecards.org/).

Abbreviations

ABCA1:

ATP-binding cassette A1

ARE:

Antioxidant response element

AS:

Atherosclerosis

CVD:

Cardiovascular diseases

GPX4:

Glutathione peroxidase4

GEO:

Gene Expression Omnibus

HO-1:

Heme oxygenase-1

NFE2L2/Nrf2:

Nuclear factor erytheroid–derived-2-like 2

Ox-LDL:

Oxidized low-density lipoprotein

ROS:

Reactive oxygen species

SOD:

Super oxide dismutase

tBHQ:

Tert-butylhydroquinone

TMAO:

Trimethylamine N-oxide

References

  1. Alonso-Pineiro JA, Gonzalez-Rovira A, Sanchez-Gomar I et al (2021) Nrf2 and heme oxygenase-1 involvement in atherosclerosis related oxidative stress. Antioxidants (Basel) 9

  2. Barajas B, Che N, Yin F et al (2011) NF-E2-related factor 2 promotes atherosclerosis by effects on plasma lipoproteins and cholesterol transport that overshadow antioxidant protection. Arterioscler Thromb Vasc Biol 1:58–66

    Article  Google Scholar 

  3. Bekkering S, Quintin J, Joosten LA et al (2014) Oxidized low-density lipoprotein induces long-term proinflammatory cytokine production and foam cell formation via epigenetic reprogramming of monocytes. Arterioscler Thromb Vasc Biol 8:1731–1738

    Article  Google Scholar 

  4. Bolen DW, Rose GD (2008) Structure and energetics of the hydrogen-bonded backbone in protein folding. Annu Rev Biochem:339–362

  5. Brunt VE, Gioscia-Ryan RA, Richey JJ et al (2019) Suppression of the gut microbiome ameliorates age-related arterial dysfunction and oxidative stress in mice. J Physiol 9:2361–2378

    Article  Google Scholar 

  6. Brunt VE, Gioscia-Ryan RA, Casso AG et al (2020) Trimethylamine-N-oxide promotes age-related vascular oxidative stress and endothelial dysfunction in mice and healthy humans. Hypertension 1:101–112

    Article  Google Scholar 

  7. Brunt VE, Casso AG, Gioscia-Ryan RA et al (2021) Gut microbiome-derived metabolite trimethylamine N-oxide induces aortic stiffening and increases systolic blood pressure with aging in mice and humans. Hypertension 2:499–511

    Article  Google Scholar 

  8. Chávez-Sánchez L, Garza-Reyes MG, Espinosa-Luna JE et al (2014) The role of TLR2, TLR4 and CD36 in macrophage activation and foam cell formation in response to oxLDL in humans. Hum Immunol 4:322–329

    Article  Google Scholar 

  9. Chen ML, Zhu XH, Ran L et al (2017) Trimethylamine-N-Oxide Induces Vascular Inflammation by Activating the NLRP3 Inflammasome Through the SIRT3-SOD2-mtROS Signaling Pathway. J Am Heart Assoc 9

  10. Chen S, Henderson A, Petriello MC et al (2019) Trimethylamine N-oxide binds and activates PERK to promote metabolic dysfunction. Cell Metab 6:1141–1151.e5

    Article  Google Scholar 

  11. Chen YY, Ye ZS, Xia NG et al (2022) TMAO as a novel predictor of major adverse vascular events and recurrence in patients with large artery atherosclerotic ischemic stroke. Clin Appl Thromb Hemost:10760296221090503

  12. Cuadrado A, Rojo AI, Wells G et al (2019) Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat Rev Drug Discov 4:295–317

    Article  Google Scholar 

  13. da Costa RM, Rodrigues D, Pereira CA et al (2019) Nrf2 as a potential mediator of cardiovascular risk in metabolic diseases. Front Pharmacol 382

  14. DeFilippis AP, Trainor PJ, Thanassoulis G et al (2022) Atherothrombotic factors and atherosclerotic cardiovascular events: the multi-ethnic study of atherosclerosis. Eur Heart J 10:971–981

    Article  Google Scholar 

  15. Fan J, Watanabe T (2022) Atherosclerosis: known and unknown. Pathol Int 3:151–160

    Article  Google Scholar 

  16. Folkersen L, Persson J, Ekstrand J et al (2012) Prediction of ischemic events on the basis of transcriptomic and genomic profiling in patients undergoing carotid endarterectomy. Mol Med 4:669–675

    Article  Google Scholar 

  17. Freigang S, Ampenberger F, Spohn G et al (2011) Nrf2 is essential for cholesterol crystal-induced inflammasome activation and exacerbation of atherosclerosis. Eur J Immunol 7:2040–2051

    Article  Google Scholar 

  18. Fukami K, Yamagishi S, Sakai K et al (2015) Oral L-carnitine supplementation increases trimethylamine-N-oxide but reduces markers of vascular injury in hemodialysis patients. J Cardiovasc Pharmacol 3:289–295

    Article  Google Scholar 

  19. Gao J, Yan KT, Wang JX et al (2020) Gut microbial taxa as potential predictive biomarkers for acute coronary syndrome and post-STEMI cardiovascular events. Sci Rep 1:2639

    Article  Google Scholar 

  20. Geng J, Yang C, Wang B et al (2018) Trimethylamine N-oxide promotes atherosclerosis via CD36-dependent MAPK/JNK pathway. Biomed Pharmacother:941–947

  21. Gonzalez-Correa C, Moleon J, Minano S et al (2021) Trimethylamine N-oxide promotes autoimmunity and a loss of vascular function in toll-like receptor 7-driven lupus mice. Antioxidants (Basel) 1

  22. Guasti L, Galliazzo S, Molaro M et al (2021) TMAO as a biomarker of cardiovascular events: a systematic review and meta-analysis. Intern Emerg Med 1:201–207

    Article  Google Scholar 

  23. Guo J, Ma J, Cai K et al (2022) Isoflavones from semen sojae preparatum improve atherosclerosis and oxidative stress by modulating Nrf2 signaling pathway through estrogen-like effects. Evid Based Complement Alternat Med:4242099

  24. He LH, Gao JH, Yu XH et al (2020) Artesunate inhibits atherosclerosis by upregulating vascular smooth muscle cells-derived LPL expression via the KLF2/NRF2/TCF7L2 pathway. Eur J Pharmacol:173408

  25. Hu CY, Lynch GC, Kokubo H et al (2010) Trimethylamine N-oxide influence on the backbone of proteins: an oligoglycine model. Proteins 3:695–704

    Article  Google Scholar 

  26. Jiang X, Li Y, Wang W et al (2020) Nuclear factor erythroid 2 related factor 2 activator JC-5411 inhibits atherosclerosis through suppression of inflammation and regulation of lipid metabolism. Front Pharmacol:532568

  27. Kattoor AJ, Pothineni NVK, Palagiri D et al (2017) Oxidative stress in atherosclerosis. Curr Atheroscler Rep 11:42

    Article  Google Scholar 

  28. Khosravi M, Poursaleh A, Ghasempour G et al (2019) The effects of oxidative stress on the development of atherosclerosis. Biol Chem 6:711–732

    Article  Google Scholar 

  29. Koeth RA, Wang Z, Levison BS et al (2013) Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med 5:576–585

    Article  Google Scholar 

  30. Li J, Meng Q, Fu Y et al (2021) Novel insights: dynamic foam cells derived from the macrophage in atherosclerosis. J Cell Physiol 9:6154–6167

    Article  Google Scholar 

  31. Li L, Du Z, Rong B et al (2020) Foam cells promote atherosclerosis progression by releasing CXCL12. Biosci Rep 1

  32. Liu J, Wang C, Li J et al (2021) Autophagy blockage promotes the pyroptosis of ox-LDL-treated macrophages by modulating the p62/Nrf2/ARE axis. J Physiol Biochem 3:419–429

    Article  Google Scholar 

  33. Liu X, Xie Z, Sun M et al (2018) Plasma trimethylamine N-oxide is associated with vulnerable plaque characteristics in CAD patients as assessed by optical coherence tomography. Int J Cardiol:18–23

  34. Liu Y, Dai M (2020) Trimethylamine N-oxide generated by the gut microbiota is associated with vascular inflammation: new insights into atherosclerosis. Mediators Inflamm:4634172

  35. Livak KJ, Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 4:402–408

    Article  Google Scholar 

  36. Lu Q, Tang SL, Liu XY et al (2013) Tertiary-butylhydroquinone upregulates expression of ATP-binding cassette transporter A1 via nuclear factor E2-related factor 2/heme oxygenase-1 signaling in THP-1 macrophage-derived foam cells. Circ J 9:2399–2408

    Article  Google Scholar 

  37. Ma G, Pan B, Chen Y et al (2017) Trimethylamine N-oxide in atherogenesis: impairing endothelial self-repair capacity and enhancing monocyte adhesion. Biosci Rep 2

  38. Mimura J, Itoh K (2015) Role of Nrf2 in the pathogenesis of atherosclerosis. Free Radic Biol Med (Pt B):221–232

  39. Mohammadi A, Najar AG, Yaghoobi MM et al (2016) Trimethylamine-N-oxide treatment induces changes in the ATP-binding cassette transporter A1 and scavenger receptor A1 in murine macrophage J774A.1 cells. Inflammation 1:393–404

    Article  Google Scholar 

  40. Mohammadi A, Vahabzadeh Z, Jamalzadeh S et al (2018) Trimethylamine-N-oxide, as a risk factor for atherosclerosis, induces stress in J774A.1 murine macrophages. Adv Med Sci 1:57–63

    Article  Google Scholar 

  41. Oakley CI, Sanborn D, Rafie N et al (2018) The direct effect of trimethylamine N-oxide (TMAO) on cardiac muscle contractile mechanics. J Clin Transl Sci S1:30–30

    Article  Google Scholar 

  42. Olek RA, Samulak JJ, Sawicka AK et al (2019) Increased trimethylamine N-oxide is not associated with oxidative stress markers in healthy aged women. Oxid Med Cell Longev:6247169

  43. Peng Q, Liu H, Luo Z et al (2022) Effect of autophagy on ferroptosis in foam cells via Nrf2. Mol Cell Biochem 5:1597–1606

    Article  Google Scholar 

  44. Randrianarisoa E, Lehn-Stefan A, Wang X et al (2016) Relationship of serum trimethylamine N-oxide (TMAO) levels with early atherosclerosis in humans. Sci Rep:26745

  45. Rittig ST, Gaal L, Schubert MR et al (2021) Vascular impairment by trimethylamine N-oxide (TMAO) uptake and the first description of a TMAO transporter in endothelial cells. Circulation (Suppl_1)

  46. Ruotsalainen AK, Inkala M, Partanen ME et al (2013) The absence of macrophage Nrf2 promotes early atherogenesis. Cardiovasc Res 1:107–115

    Article  Google Scholar 

  47. Safran M, Dalah I, Alexander J et al (2010) GeneCards Version 3: the human gene integrator. Database (Oxford):baq020

  48. Senthong V, Li XS, Hudec T et al (2016) Plasma trimethylamine N-oxide, a gut microbe-generated phosphatidylcholine metabolite, is associated with atherosclerotic burden. J Am Coll Cardiol 22:2620–2628

    Article  Google Scholar 

  49. Sheng Z, Tan Y, Liu C et al (2019) Relation of circulating trimethylamine N-oxide with coronary atherosclerotic burden in patients with ST-segment elevation myocardial infarction. Am J Cardiol 6:894–898

    Article  Google Scholar 

  50. Shi W, Huang Y, Yang Z et al (2021) Reduction of TMAO level enhances the stability of carotid atherosclerotic plaque through promoting macrophage M2 polarization and efferocytosis. Biosci Rep 6

  51. Steinke I, Ghanei N, Govindarajulu M et al (2020) Drug discovery and development of novel therapeutics for inhibiting TMAO in models of atherosclerosis and diabetes. Front Physiol:567899

  52. Sun T, Zhang Y, Yin J et al (2021) Association of gut microbiota-dependent metabolite trimethylamine N-oxide with first ischemic stroke. J Atheroscler Thromb 4:320–328

    Article  Google Scholar 

  53. Sun X, Jiao X, Ma Y et al (2016) Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochem Biophys Res Commun 1-2:63–70

    Article  Google Scholar 

  54. Sykiotis GP, Bohmann D (2010) Stress-activated cap'n'collar transcription factors in aging and human disease. Sci Signal 112:re3

    Google Scholar 

  55. Tan Y, Sheng Z, Zhou P et al (2019) Plasma trimethylamine N-oxide as a novel biomarker for plaque rupture in patients with ST-segment-elevation myocardial infarction. Circ Cardiovasc Interv 1:e007281

    Article  Google Scholar 

  56. Tang WH, Wang Z, Levison BS et al (2013) Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 17:1575–1584

    Article  Google Scholar 

  57. Tang WHW, Li DY, Hazen SL (2019) Dietary metabolism, the gut microbiome, and heart failure. Nat Rev Cardiol 3:137–154

    Article  Google Scholar 

  58. Wang Z, Klipfell E, Bennett BJ et al (2011) Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 7341:57–63

    Article  Google Scholar 

  59. Wang Z, Roberts AB, Buffa JA et al (2015) Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 7:1585–1595

    Article  Google Scholar 

  60. Wu P, Chen J, Chen J et al (2020) Trimethylamine N-oxide promotes apoE(-/-) mice atherosclerosis by inducing vascular endothelial cell pyroptosis via the SDHB/ROS pathway. J Cell Physiol 10:6582–6591

    Article  Google Scholar 

  61. Wu Y, Song F, Li Y et al (2021) Acacetin exerts antioxidant potential against atherosclerosis through Nrf2 pathway in apoE(-/-) Mice. J Cell Mol Med 1:521–534

    Article  Google Scholar 

  62. Yang S, Dai H, Lu Y et al (2022) Trimethylamine N-oxide promotes cell proliferation and angiogenesis in colorectal cancer. Immunol Res:7043856

  63. Zhang Y, Fu Y, Jia L et al (2022) TMT-based quantitative proteomic profiling of human monocyte-derived macrophages and foam cells. Proteome Sci 1:1

    Article  Google Scholar 

  64. Zheng L, Zheng J, Xie Y et al (2019) Serum gut microbe-dependent trimethylamine N-oxide improves the prediction of future cardiovascular disease in a community-based general population. Atherosclerosis:126–131

  65. Zhu W, Gregory JC, Org E et al (2016) Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 1:111–124

    Article  Google Scholar 

Download references

Funding

This study was supported by the grants from the National Natural Science Foundation of China (No. 81672084).

Author information

Authors and Affiliations

Authors

Contributions

First author: do experiments, write articles, participate in part of the experimental design. Second, third, and fourth authors: assist in completing the experiment. Corresponding author: participate in article revision, experimental design and supervision, financial support. The authors declare that all data were generated in-house and that no paper mill was used.

Corresponding author

Correspondence to XiuRu Guan.

Ethics declarations

Research involving human participants and/or animals

This study does not involve human participants and/or animals.

Informed consent

All authors were informed and agreed to sign.

Conflict of interest

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Key Points

•The proatherogenic effect of high plasma TMAO level might be associated with the differential expression of NFE2L2 decreased by meta-analysis and bioinformatic analysis.

•TMAO promotes oxidative stress by inhibiting Nrf2/ARE signaling axis in macrophage foam cell.

•TMAO induces lipid accumulation by inhibiting cholesterol efflux protein expression in foam cell which partially alleviated by upregulation of Nrf2.

Supplementary information

ESM 1

Supplementary Table 1 Characteristics of the Included Studies. Abbreviations: M, male; CVD, cardiovascular disease; STEMI, ST-segment elevation myocardial infarction; ACS, Acute coronary syndromes. Data of TMAO Levels are expressed as medians (interquartile ranges) or means ± standard deviations. (DOCX 61 kb)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luo, Z., Yu, X., Wang, C. et al. Trimethylamine N-oxide promotes oxidative stress and lipid accumulation in macrophage foam cells via the Nrf2/ABCA1 pathway. J Physiol Biochem 80, 67–79 (2024). https://doi.org/10.1007/s13105-023-00984-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13105-023-00984-y

Keywords

Navigation