AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (2.2 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Open Access

Molecular mechanisms of aflatoxin neurotoxicity and potential neuroprotective agents

Chongshan Daia,b( )Erjie TiancHui LidSubhajit Das GuptaeZhihui HaoaZhanhui WangaTony VelkovfJianzhong Shena,b( )
National Key Laboratory of Veterinary Public Health and Safety, Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
Beijing Key Laboratory of Diagnostic and Traceability Technologies for Food Poisoning, Beijing Center for Disease Prevention and Control, Beijing 100013, China
Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas 75230, USA
Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Victoria 3800, Australia

Peer review under responsibility of Tsinghua University Press.

Show Author Information

Abstract

Aflatoxins (AFTs) represent one of the most notorious classes of deadly mycotoxins produced by certain fungi that are found on agricultural crops. Aflatoxins are highly toxic to mammals and are known to cause a series of detrimental effects, including neuro-, hepato-, nephron-, and immuno-toxicity. In this original review we summarize the mechanisms of aflatoxin-induced neurotoxicity and the clinical potential of novel neuroprotective agents. Aflatoxin B1 (AFB1) is the most toxic congener among the 21 identified AFTs. Recent studies have shown that food borne exposure to AFB1 and/or its metabolites often leads to fatal neurotoxicity in animals and humans. Animal studies indicated that AFB1 exposure could induce abnormal behavioral changes, including anxiety, lethargy disorders, depression-like behavior, cognitive, learning and memory defects, and decreased feeding behavior. Mechanistically, AFB1 exposure has been associated with lipid peroxidation, ablation of non-enzymatic and enzymatic antioxidant defense systems and decreased neurotransmitter levels. AFB1 exposure has also been shown to induce DNA damage, apoptosis, pyroptosis, and mitochondrial dysfunction in the brain tissue. Several signaling pathways, including gasdermin D, toll like receptor 2 (TLR2), TLR4, Akt, NF-κB, ERK/MAPK, protein kinase C (PKC), and mitochondrial apoptotic pathways have been shown to participate in AFB1-induced neuronal or astrocyte cell death. Targeting these pathways by small molecules (e.g., quercetin, curcumin, and gallic acid, and dimethyl fumarate), Chinese herbal extracts (e.g., Artichoke leaf extract, Chelidonium majus ethanolic extract, pumpkin extract, and Crocus sativus L. tea), and probiotic supplements could effectively improve AFB1-induced neurobehavioral abnormalities and neurotoxicity. To date, the precise molecular mechanisms of AFB1-induced neurotoxicity and potential neuroprotective agents remain unclear. In the present review, the clinical manifestations, molecular mechanisms, and potential neuroprotective agents of AFB1-induced neurotoxicity are summarized in the broadest overview. It is most hopeful that this broad reaching review provides valuable insights and stimulates broader discussion to develop the effective neuroprotective agents against aflatoxins.

References

[1]

C. Dai, E. Tian, Z. Hao, et al., Aflatoxin B1 toxicity and protective effects of curcumin: molecular mechanisms and clinical implications, Antioxidants (Basel) 11 (2022). https://doi.org/10.3390/antiox11102031.

[2]

S. Adunphatcharaphon, C.T. Elliott, T. Sooksimuang, et al., The evolution of multiplex detection of mycotoxins using immunoassay platform technologies, J. Hazard. Mater. 432 (2022) 128706. https://doi.org/10.1016/j.jhazmat.2022.128706.

[3]

C.P. Wild, Y.Y. Gong, Mycotoxins and human disease: a largely ignored global health issue, Carcinogenesis 31 (2010) 71-82. https://doi.org/10.1093/carcin/bgp264.

[4]

B. Ge, K. Zhao, W. Wang, et al., Determination of 14 mycotoxins in Chinese herbs by liquid chromatography-tandem mass spectrometry with immunoaffinity purification, Se Pu 29 (2011) 495-500. https://doi.org/10.3724/sp.j.1123.2011.00495.

[5]

A.E. Desjardins, T.M. Hohn, S.P. McCormick, Trichothecene biosynthesis in Fusarium species: chemistry, genetics, and significance, Microbiol. Rev. 57 (1993) 595-604.

[6]

M.A. Haque, Y. Wang, Z. Shen, et al., Mycotoxin contamination and control strategy in human, domestic animal and poultry: a review, Microb. Pathog. 142 (2020) 104095. https://doi.org/10.1016/j.micpath.2020.104095.

[7]

T. Chen, J. Liu, Y. Li, et al., Burden of disease associated with dietary exposure to aflatoxins in China in 2020, Nutrients 14 (2022) 1027. https://doi.org/10.3390/nu14051027.

[8]

J. Martínez, M. Hernández-Rodríguez, A. Méndez-Albores, et al., Computational studies of aflatoxin B1 (AFB1): a review, Toxins 15 (2023) 135.

[9]

S. Subramaniam, M.R. Sabran, J. Stanslas, et al., Effect of aflatoxin B1 exposure on the progression of depressive-like behavior in rats, Front. Nutr. 9 (2022) 1032810. https://doi.org/10.3389/fnut.2022.1032810.

[10]

A. Alsayyah, R. ElMazoudy, M. Al-Namshan, et al., Chronic neurodegeneration by aflatoxin B1 depends on alterations of brain enzyme activity and immunoexpression of astrocyte in male rats, Ecotoxicol. Environ. Saf. 182 (2019) 109407. https://doi.org/10.1016/j.ecoenv.2019.109407.

[11]

O.A. Oyelami, S.M. Maxwell, K.A. Adelusola, et al., Aflatoxins in the lungs of children with kwashiorkor and children with miscellaneous diseases in Nigeria, J. Toxicol. Environ. Health 51 (1997) 623-628. https://doi.org/10.1080/00984109708984048.

[12]

B. Huang, Q. Chen, L. Wang, et al., Aflatoxin B1 Induces neurotoxicity through reactive oxygen species generation, DNA damage, apoptosis, and S-phase cell cycle arrest, Int. J. Mol. Sci. 21 (2020) 6517. https://doi.org/10.3390/ijms21186517.

[13]

H. Qureshi, S.S. Hamid, S.S. Ali, et al., Cytotoxic effects of aflatoxin B1 on human brain microvascular endothelial cells of the blood-brain barrier, Med. Mycol. 53 (2015) 409-416. https://doi.org/10.1093/mmy/myv010.

[14]

M.D. Baldissera, C.F. Souza, C.C. Zeppenfeld, et al., Aflatoxin B(1)-contaminated diet disrupts the blood-brain barrier and affects fish behavior: involvement of neurotransmitters in brain synaptosomes, Environ. Toxicol. Pharmacol. 60 (2018) 45-51. https://doi.org/10.1016/j.etap.2018.04.003.

[15]

Z.I. Linardaki, F.N. Lamari, M. Margarity, Saffron (Crocus sativus L.) tea intake prevents learning/memory defects and neurobiochemical alterations induced by aflatoxin B(1) exposure in adult mice, Neurochem. Res. 42 (2017) 2743-2754. https://doi.org/10.1007/s11064-017-2283-z.

[16]

T.S. Wu, Y.C. Cheng, P.J. Chen, et al., Exposure to aflatoxin B(1) interferes with locomotion and neural development in zebrafish embryos and larvae, Chemosphere 217 (2019) 905-913. https://doi.org/10.1016/j.chemosphere.2018.11.058.

[17]

M.M.M. Makhlouf, Histological and ultrastructural study of aflatoxin B1 induced neurotoxicity in Sciatic nerve of adult male albino rats, Ultrastruct. Pathol. 44 (2020) 52-60. https://doi.org/10.1080/01913123.2019.1709933.

[18]

S. Park, J.Y. Lee, S. You, et al., Neurotoxic effects of aflatoxin B1 on human astrocytes in vitro and on glial cell development in zebrafish in vivo, J. Hazard. Mater. 386 (2020) 121639. https://doi.org/10.1016/j.jhazmat.2019.121639.

[19]

J. Zhang, D. Su, Q. Liu, et al., Gasdermin D-mediated microglial pyroptosis exacerbates neurotoxicity of aflatoxins B1 and M1 in mouse primary microglia and neuronal cultures, Neurotoxicology 91 (2022) 305-320. https://doi.org/10.1016/j.neuro.2022.06.003.

[20]

N.S. Souto, A. Claudia Monteiro Braga, M. Lutchemeyer de Freitas, et al., Aflatoxin B1 reduces non-enzymatic antioxidant defenses and increases protein kinase C activation in the cerebral cortex of young rats, Nutr. Neurosci. 21 (2018) 268-275. https://doi.org/10.1080/1028415x.2017.1278837.

[21]

J. Mehrzad, A.M. Malvandi, M. Alipour, et al., Environmentally relevant level of aflatoxin B(1) elicits toxic pro-inflammatory response in murine CNS-derived cells, Toxicol. Lett. 279 (2017) 96-106. https://doi.org/10.1016/j.toxlet.2017.07.902.

[22]

E. Madrigal-Santillán, J.A. Morales-González, N. Vargas-Mendoza, et al., Antigenotoxic studies of different substances to reduce the DNA damage induced by aflatoxin B(1) and ochratoxin A, Toxins (Basel) 2 (2010) 738-757. https://doi.org/10.3390/toxins2040738.

[23]

E. Gugliandolo, A.F. Peritore, R. D’Amico, et al., Evaluation of neuroprotective effects of quercetin against aflatoxin B1-intoxicated mice, Animals (Basel) 10 (2020) 898. https://doi.org/10.3390/ani10050898.

[24]

M. Mazaheri, A.A. Moosavi-Movahedi, A.A. Saboury, et al., Curcumin protects β-lactoglobulin fibril formation and fibril-induced neurotoxicity in PC12 Cells, PLoS One 10 (2015) e0133206. https://doi.org/10.1371/journal.pone.0133206.

[25]

S.C. Chalivendra, C. DeRobertis, P.K. Chang, et al., Cyclopiazonic acid is a pathogenicity factor for aspergillus flavus and a promising target for screening germplasm for ear rot resistance, Mol. Plant Microbe. Interact. 30 (2017) 361-373. https://doi.org/10.1094/mpmi-02-17-0026-r.

[26]

I.A. Adedara, S.E. Owumi, A.K. Oyelere, et al., Neuroprotective role of gallic acid in aflatoxin B(1)-induced behavioral abnormalities in rats, J. Biochem. Mol. Toxicol. 35 (2021) e22684. https://doi.org/10.1002/jbt.22684.

[27]

G. Aytekin Sahin, D. Karabulut, G. Unal, et al., Effects of probiotic supplementation on very low dose AFB1-induced neurotoxicity in adult male rats, Life Sci. 306 (2022) 120798. https://doi.org/10.1016/j.lfs.2022.120798.

[28]

M. Frangiamone, M. Alonso-Garrido, G. Font, et al., Pumpkin extract and fermented whey individually and in combination alleviated AFB1- and OTA-induced alterations on neuronal differentiation in vitro, Food Chem. Toxicol. 164 (2022) 113011. https://doi.org/10.1016/j.fct.2022.113011.

[29]

E.A. Ibrahim, M.I. Yousef, D.A. Ghareeb, et al., Artichoke leaf extract-mediated neuroprotection against effects of aflatoxin in male rats, Biomed. Res. Int. 2022 (2022) 4421828. https://doi.org/10.1155/2022/4421828.

[30]

E.M. Mateo, A. Tarazona, M. Jiménez, et al., Lactic acid bacteria as potential agents for biocontrol of aflatoxigenic and ochratoxigenic fungi, Toxins (Basel) 14 (2022) 807. https://doi.org/10.3390/toxins14110807.

[31]

N. Ra Kasem, F.A. Mannaa, K.G. Abdel-Wahhab, et al., Preventive efficiency of chelidonium majus ethanolic extract against aflatoxin B(1) induced neurochemical deteriorations in rats, Pak. J. Biol. Sci. 25 (2022) 234-244. https://doi.org/10.3923/pjbs.2022.234.244.

[32]

F. Trebak, A. Alaoui, D. Alexandre, et al., Impact of aflatoxin B1 on hypothalamic neuropeptides regulating feeding behavior, Neurotoxicology 49 (2015) 165-173. https://doi.org/10.1016/j.neuro.2015.06.008.

[33]

Q. Zhang, Z. Feng, J. Lu, et al., Aflatoxin B1 inhibited autophagy flux by inducing lysosomal alkalinization in HepG2 cells, Toxicol. Mech. Methods 31 (2021) 450-456. https://doi.org/10.1080/15376516.2021.1909196.

[34]

R.A. Coulombe Jr, R.P. Sharma, Effect of repeated dietary exposure of aflatoxin B1 on brain biogenic amines and metabolites in the rat, Toxicol. Appl. Pharmacol. 80 (1985) 496-501. https://doi.org/10.1016/0041-008x(85)90394-1.

[35]

M. Gregorio, K. Bordin, P. Souto, et al., Comparative biotransformation of aflatoxin B1 in swine, domestic fowls, and humans, Toxin Rev. 34 (2015) 142-150. https://doi.org/10.3109/15569543.2015.1091979.

[36]

G.J. Diaz, H.W. Murcia, S.M. Cepeda, Cytochrome P450 enzymes involved in the metabolism of aflatoxin B1 in chickens and quail, Poult. Sci. 89 (2010) 2461-2469. https://doi.org/10.3382/ps.2010-00864.

[37]

F.P. Guengerich, W.W. Johnson, T. Shimada, et al., Activation and detoxication of aflatoxin B1, Mutat. Res. 402 (1998) 121-128. https://doi.org/10.1016/s0027-5107(97)00289-3.

[38]

F. Ihara, M. Nishimura, Y. Muroi, et al., Changes in neurotransmitter levels and expression of immediate early genes in brain of mice infected with Neospora caninum, Sci. Rep. 6 (2016) 23052. https://doi.org/10.1038/srep23052.

[39]

A. Jawaid, E. Pawlowicz, P.E. Schulz, Do acetylcholinesterase inhibitors increase anxiety and depression in elderly adults with dementia? J.A.G.S. 63 (2015) 1702-1704. https://doi.org/10.1111/jgs.13567.

[40]

E.M. Al Olayan, A.S. Aloufi, O.D. AlAmri, et al., Protocatechuic acid mitigates cadmium-induced neurotoxicity in rats: role of oxidative stress, inflammation and apoptosis, Sci. Total Environ. 723 (2020) 137969. https://doi.org/10.1016/j.scitotenv.2020.137969.

[41]

T.O. Ajiboye, Colistin sulphate induced neurotoxicity: studies on cholinergic, monoaminergic, purinergic and oxidative stress biomarkers, Biomed. Pharmacother. 103 (2018) 1701-1707. https://doi.org/10.1016/j.biopha.2018.04.189.

[42]

B. Cheng, F. Jiang, M. Su, et al., Effects of lincomycin hydrochloride on the neurotoxicity of zebrafish, Ecotoxicol. Environ. Saf. 201 (2020) 110725. https://doi.org/10.1016/j.ecoenv.2020.110725

[43]

J. de Almeida, R. Dolezal, O. Krejcar, et al., Molecular modeling studies on the interactions of aflatoxin B1 and its metabolites with human acetylcholinesterase. part Ⅱ: interactions with the catalytic anionic site (CAS), Toxins (Basel) 10 (2018) 389. https://doi.org/10.3390/toxins10100389.

[44]

J. de Almeida, S.F.A. Cavalcante, R. Dolezal, et al., Surface screening, molecular modeling and in vitro studies on the interactions of aflatoxin M1 and human enzymes acetyl- and butyrylcholinesterase, Chem. Biol. Interact. 308 (2019) 113-119. https://doi.org/10.1016/j.cbi.2019.05.022.

[45]

K. Sheng, X. Lu, J. Yue, et al., Role of neurotransmitters 5-hydroxytryptamine and substance P in anorexia induction following oral exposure to the trichothecene T-2 toxin, Food Chem. Toxicol. 123 (2019) 1-8. https://doi.org/10.1016/j.fct.2018.10.041.

[46]

H. Sies, Oxidative stress: a concept in redox biology and medicine, Redox Biol. 4 (2015) 180-183. https://doi.org/10.1016/j.redox.2015.01.002.

[47]

X. Zhang, Y. Wang, T. Velkov, et al., T-2 toxin-induced toxicity in neuroblastoma-2a cells involves the generation of reactive oxygen, mitochondrial dysfunction and inhibition of Nrf2/HO-1 pathway, Food Chem. Toxicol. 114 (2018) 88-97. https://doi.org/10.1016/j.fct.2018.02.010.

[48]

C. Dai, X. Xiao, F. Sun, et al., T-2 toxin neurotoxicity: role of oxidative stress and mitochondrial dysfunction, Arch. Toxicol. 93 (2019) 3041-3056. https://doi.org/10.1007/s00204-019-02577-5.

[49]

T. Sun, Q. Zhang, M. Li, et al., T-2 Toxin induces apoptotic cell death and protective autophagy in mouse microglia BV2 cells, J. Fungi (Basel) 8 (2022) 761. https://doi.org/10.3390/jof8080761.

[50]

C. Dai, X. Xiao, J. Li, et al., Molecular mechanisms of neurotoxicity induced by polymyxins and chemoprevention, ACS Chem. Neurosci. 10 (2019) 120-131. https://doi.org/10.1021/acschemneuro.8b00300.

[51]

M. Gulec, E. Oral, O.B. Dursun, et al., Mirtazapine protects against cisplatin-induced oxidative stress and DNA damage in the rat brain, Psychiatry. Clin. Neurosci. 67 (2013) 50-58. https://doi.org/10.1111/j.1440-1819.2012.02395.x.

[52]

N.S. Chandel, Mitochondria: back to the future, Nat. Rev. Mol. Cell Biol. 19 (2018) 76. https://doi.org/10.1038/nrm.2017.133.

[53]

D.A. Drechsel, M. Patel, Role of reactive oxygen species in the neurotoxicity of environmental agents implicated in Parkinson’s disease, Free Radic. Biol. Med. 45 (2008) 1045. https://doi.org/10.1016/j.freeadbiomed.2008.07.005.

[54]

C. Cid-Castro, D. Hernandez-Espinosa, J. Moran, ROS as regulators of mitochondrial dynamics in neurons, Cell. Mol. Neurobiol. 38 (2018) 995-1007. https://doi.org/10.1007/s10571-018-0584-7.

[55]

W.X. Zhang, P. Kubes, NOX2: is the best defense a good offense? Blood 139 (2022) 2851-2853. https://doi.org/10.1182/blood.2022016192.

[56]

W. Cao, J. Gao, Y. Zhang, et al., Autophagy up-regulated by MEK/ERK promotes the repair of DNA damage caused by aflatoxin B1, Toxicol. Mech. Methods 32 (2022) 87-96. https://doi.org/10.1080/15376516.2021.1968985.

[57]

W. Kallenborn-Gerhardt, S.W. Hohmann, K.M. Syhr, et al., Nox2-dependent signaling between macrophages and sensory neurons contributes to neuropathic pain hypersensitivity, Pain 155 (2014) 2161-2170. https://doi.org/10.1016/j.pain.2014.08.013.

[58]

J. Deng, L. Zhao, N.Y. Zhang, et al., Aflatoxin B(1) metabolism: regulation by phase Ⅰ and Ⅱ metabolizing enzymes and chemoprotective agents, Mutat. Res. Rev. 778 (2018) 79-89. https://doi.org/10.1016/j.mrrev.2018.10.002.

[59]

J.W. Shin, K.S. Chun, D.H. Kim, et al., Curcumin induces stabilization of Nrf2 protein through Keap1 cysteine modification, Biochem. Pharmacol. 173 (2020) 113820. https://doi.org/10.1016/j.bcp.2020.113820.

[60]

M.C. Martinez, R. Andriantsitohaina, Reactive nitrogen species: molecular mechanisms and potential significance in health and disease, Antioxid. Redox Signal. 11 (2009) 669-702. https://doi.org/10.1089/ars.2007.1993.

[61]

C.S. Dai, B. Li, Y. Zhou, et al., Curcumin attenuates quinocetone induced apoptosis and inflammation via the opposite modulation of Nrf2/HO-1 and NF-kB pathway in human hepatocyte L02 cells, Food Chemic. Toxicol. 95 (2016) 52-63. https://doi.org/10.1016/j.fct.2016.06.025.

[62]

R. Medzhitov, Origin and physiological roles of inflammation, Nature 454 (2008) 428-435. https://doi.org/10.1038/nature07201.

[63]

H. Wake, A.J. Moorhouse, J. Nabekura, Functions of microglia in the central nervous system-beyond the immune response, Neuron. Glia. Biol. 7 (2011) 47-53. https://doi.org/10.1017/S1740925X12000063.

[64]

D. Matias, J. Balça-Silva, G.C. da Graça, et al., Microglia/astrocytes-glioblastoma crosstalk: crucial molecular mechanisms and microenvironmental factors, Front. Cell. Neurosci. 12 (2018) 235. https://doi.org/10.3389/fncel.2018.00235.

[65]

K. He, X. Liang, T. Wei, et al., DNA damage in BV-2 cells: an important supplement to the neurotoxicity of CdTe quantum dots, J. Appl. Toxicol. 39 (2019) 525-539. https://doi.org/10.1002/jat.3745.

[66]

Z. Liu, Q. Zhu, E. Song, et al., Polybrominated diphenyl ethers quinone exhibits neurotoxicity by inducing DNA damage, cell cycle arrest, apoptosis and p53-driven adaptive response in microglia BV2 cells, Toxicology 457 (2021) 152807. https://doi.org/10.1016/j.tox.2021.152807.

[67]

J.P. Novo, B. Martins, R.S. Raposo, et al., Cellular and molecular mechanisms mediating methylmercury neurotoxicity and neuroinflammation, Int. J. Mol. Sci. 22 (2021) 3101. https://doi.org/10.3390/ijms22063101.

[68]

N.G. Bahey, H.O.A. Elaziz, K.K.E.S. Gadalla, Toxic effect of aflatoxin B1 and the role of recovery on the rat cerebral cortex and hippocampus, Tissue Cell 47 (2015) 559-566. https://doi.org/10.1016/j.tice.2015.09.001.

[69]

Y. Zhou, S. Wang, H. Luo, et al., Aflatoxin B1 induces microglia cells apoptosis mediated by oxidative stress through NF-κB signaling pathway in mice spinal cords, Environ. Toxicol. Pharmacol. 90 (2022) 103794. https://doi.org/10.1016/j.etap.2021.103794

[70]

N. Karunaweera, R. Raju, E. Gyengesi, et al., Plant polyphenols as inhibitors of NF-κB induced cytokine production a potential anti-inflammatory treatment for Alzheimer’s disease? Front. Mol. Neurosci. 8 (2015) 24. https://doi.org/10.3389/fnmol.2015.00024.

[71]

A. Kumar, Y. Takada, A.M. Boriek, et al., Nuclear factor-κB: its role in health and disease, J. Mol. Med. 82 (2004) 434-448. https://doi.org/10.1007/s00109-004-0555-y.

[72]

C. Jobin, C.A. Bradham, M.P. Russo, et al., Curcumin blocks cytokine-mediated NF-κB activation and proinflammatory gene expression by inhibiting inhibitory factor Ⅰ-κB kinase activity, J. Immunol. 163 (1999) 3474-3483.

[73]

K.V. Swanson, M. Deng, J.P. Ting, The NLRP3 inflammasome: molecular activation and regulation to therapeutics, Nat. Rev. Immunol. 19 (2019) 477-489. https://doi.org/10.1038/s41577-019-0165-0.

[74]

A.M. Malvandi, J. Mehrzad, M. Saleh-moghaddam, Biologically relevant doses of mixed aflatoxins B and G up-regulate MyD88, TLR2, TLR4 and CD14 transcripts in human PBMCs, Immunopharmacol. Immunotoxicol. 35 (2013) 528-532. https://doi.org/10.3109/08923973.2013.803572.

[75]

J. Mehrzad, G. Klein, J. Kamphues, et al., In vitro effects of very low levels of aflatoxin B1 on free radicals production and bactericidal activity of bovine blood neutrophils, Vet. Immunol. Immunopathol. 141 (2011) 16-25. https://doi.org/10.1016/j.vetimm.2011.01.010.

[76]

J. Mehrzad, A. Bahari, M.R. Bassami, et al., Immunobiologically relevant level of aflatoxin B(1) alters transcription of key functional immune genes, phagocytosis and survival of human dendritic cells, Immunol. Lett. 197 (2018) 44-52. https://doi.org/10.1016/j.imlet.2018.03.008.

[77]

J. Deguine, G.M. Barton, MyD88: a central player in innate immune signaling, F1000Prime Rep. 6 (2014) 97. https://doi.org/10.12703/p6-97.

[78]

P. Poprac, K. Jomova, M. Simunkova, et al., Targeting free radicals in oxidative stress-related human diseases, Trends. Pharmacol. Sci. 38 (2017) 592-607. https://doi.org/10.1016/j.tips.2017.04.005.

[79]

F.I. Ikegwuonu, The neurotoxicity of aflatoxin B1 in the rat, Toxicology 28 (1983) 247-259. https://doi.org/10.1016/0300-483x(83)90121-x.

[80]

X.L. Wan, N. Li, Y.J. Chen, et al., Protective effects of lycopene on mitochondrial oxidative injury and dysfunction in the liver of aflatoxin B(1)-exposed broilers, Poult. Sci. 100 (2021) 101441. https://doi.org/10.1016/j.psj.2021.101441.

[81]

M. Alonso-Garrido, P. Tedeschi, A. Maietti, et al., Mitochondrial transcriptional study of the effect of aflatoxins, enniatins and carotenoids in vitro in a blood brain barrier model, Food Chem. Toxicol. 137 (2020) 111077. https://doi.org/10.1016/j.fct.2019.111077.

[82]

S. Li, R. Liu, S. Xia, et al., Protective role of curcumin on aflatoxin B1-induced TLR4/RIPK pathway mediated-necroptosis and inflammation in chicken liver, Ecotoxicol. Environ. Saf. 233 (2022) 113319. https://doi.org/10.1016/j.ecoenv.2022.113319.

[83]

Y. Wang, F. Liu, M. Liu, et al., Curcumin mitigates aflatoxin B1-induced liver injury via regulating the NLRP3 inflammasome and Nrf2 signaling pathway, Food Chem. Toxicol. 161 (2022) 112823. https://doi.org/10.1016/j.fct.2022.112823.

[84]

Y. Wang, F. Liu, X. Zhou, et al., Alleviation of oral exposure to aflatoxin B1-induced renal dysfunction, oxidative stress, and cell apoptosis in mice kidney by curcumin, Antioxidants (Basel) 11 (2022) 1082. https://doi.org/10.3390/antiox11061082.

[85]

X. Wang, F. Yang, L. Na, et al., Ferulic acid alleviates AFB1-induced duodenal barrier damage in rats via up-regulating tight junction proteins, down-regulating ROCK, competing CYP450 enzyme and activating GST, Ecotoxicol. Environ. Saf. 241 (2022) 113805. https://doi.org/10.1016/j.ecoenv.2022.113805.

[86]

O. Gursoy-Yuzugullu, H. Yuzugullu, M. Yilmaz, et al., Aflatoxin genotoxicity is associated with a defective DNA damage response bypassing p53 activation, Liver Int. 31 (2011) 561-571. https://doi.org/10.1111/j.1478-3231.2011.02474.x.

[87]

K.D. Wierda, R.F. Toonen, H. de Wit, et al., Interdependence of PKC-dependent and PKC-independent pathways for presynaptic plasticity, Neuron 54 (2007) 275-290. https://doi.org/10.1016/j.neuron.2007.04.001.

[88]

S. Jin, H. Yang, Y. Jiao, et al., Dietary curcumin alleviated acute ileum damage of ducks (anas platyrhynchos) induced by AFB1 through regulating Nrf2-ARE and NF-κB signaling pathways, Foods 10 (2021) 1370. https://doi.org/10.3390/foods10061370.

[89]

F. Wan, L. Tang, G. Rao, et al., Curcumin activates the Nrf2 pathway to alleviate AFB1-induced immunosuppression in the spleen of ducklings, Toxicon 209 (2022) 18-27. https://doi.org/10.1016/j.toxicon.2022.01.010.

[90]

S. Li, R. Liu, G. Wei, et al., Curcumin protects against aflatoxin B1-induced liver injury in broilers via the modulation of long non-coding RNA expression, Ecotoxicol. Environ. Saf. 208 (2021) 111725. https://doi.org/10.1016/j.ecoenv.2020.111725.

[91]

S. Jin, H. Yang, Y. Wang, et al., Dietary curcumin alleviated aflatoxin B1-induced acute liver damage in ducks by regulating NLRP3-caspase-1 signaling pathways, Foods 10 (2021) 3086. https://doi.org/10.3390/foods10123086.

[92]

S. Damiano, W. Jarriyawattanachaikul, F. Girolami, et al., Curcumin supplementation protects broiler chickens against the renal oxidative stress induced by the dietary exposure to low levels of aflatoxin B1, Front. Vet. Sci. 8 (2021) 822227. https://doi.org/10.3389/fvets.2021.822227.

[93]

M. Pauletto, M. Giantin, R. Tolosi, et al., Curcumin mitigates AFB1-induced hepatic toxicity by triggering cattle antioxidant and anti-inflammatory pathways: a whole transcriptomic in vitro study, Antioxidants (Basel) 9 (2020) 1059. https://doi.org/10.3390/antiox9111059.

[94]

P. Cheng, M. Ishfaq, H. Yu, et al., Curcumin ameliorates duodenal toxicity of AFB1 in chicken through inducing P-glycoprotein and downregulating cytochrome P450 enzymes, Poult. Sci. 99 (2020) 7035-7045. https://doi.org/10.1016/j.psj.2020.09.055.

[95]

X. Wang, I. Muhammad, X. Sun, et al., Protective role of curcumin in ameliorating AFB(1)-induced apoptosis via mitochondrial pathway in liver cells, Mol. Biol. Rep. 45 (2018) 881-891. https://doi.org/10.1007/s11033-018-4234-4.

[96]

K.B. Soni, M. Lahiri, P. Chackradeo, et al., Protective effect of food additives on aflatoxin-induced mutagenicity and hepatocarcinogenicity, Cancer Lett. 115 (1997) 129-133. https://doi.org/10.1016/s0304-3835(97)04710-1.

[97]

M.A. Nasef, M.I. Yousef, D.A. Ghareeb, et al., Hepatoprotective effects of a chemically-characterized extract from artichoke (Cynara scolymus L.) against AFB(1)-induced toxicity in rats, Drug Chem. Toxicol. (2022) 1-13. https://doi.org/10.1080/01480545.2022.2129672.

[98]

W.P. Liew, M.R. Sabran, L.T. Than, et al., Metagenomic and proteomic approaches in elucidating aflatoxin B(1) detoxification mechanisms of probiotic Lactobacillus casei Shirota towards intestine, Food Chem. Toxicol. 160 (2022) 112808. https://doi.org/10.1016/j.fct.2022.112808.

[99]

W.P. Liew, Z. Nurul-Adilah, L.T.L. Than, et al., The binding efficiency and interaction of Lactobacillus casei shirota toward aflatoxin B1, Front. Microbiol. 9 (2018) 1503. https://doi.org/10.3389/fmicb.2018.01503.

[100]

H.W. Guo, J. Chang, P. Wang, et al., Effects of compound probiotics and aflatoxin-degradation enzyme on alleviating aflatoxin-induced cytotoxicity in chicken embryo primary intestinal epithelium, liver and kidney cells, AMB Express 11 (2021) 35. https://doi.org/10.1186/s13568-021-01196-7.

Food Science and Human Wellness
Pages 2445-2455
Cite this article:
Dai C, Tian E, Li H, et al. Molecular mechanisms of aflatoxin neurotoxicity and potential neuroprotective agents. Food Science and Human Wellness, 2024, 13(5): 2445-2455. https://doi.org/10.26599/FSHW.2022.9250201

1489

Views

171

Downloads

2

Crossref

2

Web of Science

2

Scopus

0

CSCD

Altmetrics

Received: 22 December 2022
Revised: 24 January 2023
Accepted: 04 March 2023
Published: 10 October 2024
© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return