Search
2024 Volume 3
Article Contents
REVIEW   Open Access    

Review of the toxic effects and health functions of arecoline on multiple organ systems

More Information
  • Arecoline, the principal active alkaloid in the areca nut, is known for its ability to induce euphoric sensations. Since ancient times, arecoline has garnered attention for its therapeutic potential in addressing psychiatric disorders and alleviating gastrointestinal ailments. However, in 2020, the International Agency for Research on Cancer has classified arecoline as 'probably carcinogenic to humans' (Group 2B carcinogen), supported by compelling mechanistic evidence. The mechanism of action of arecoline has been extensively studied, but the results of these studies are scattered and lack systematic integration and generalization. In this paper, we have systematically summarized the mechanism of arecoline within the oral cavity, central nervous system, cardiovascular system, and digestion system, in terms of both health functions and toxic effects. In addition, we found some concentration-effect relationship between arecoline in the central nervous system and digestive system, i.e., low doses are beneficial and high doses are harmful. By summarizing the mechanisms of arecoline, this review is poised to provide in-depth and valuable insights into the clinical practice and targeted therapy of arecoline in the future.
  • 加载中
  • [1]

    Peng W, Liu YJ, Wu N, Sun T, He XY, et al. 2015. Areca catechu L.(Arecaceae):a review of its traditional uses, botany, phytochemistry,pharmacology and toxicology. Journal of ethnopharmacology164:340−56

    [2]

    Volgin AD, Bashirzade A, Amstislavskaya TG, Yakovlev OA, Demin KA, et al. 2019. DARK Classics in Chemical Neuroscience: Arecoline. ACS Chemical Neuroscience 10:2176−85

    doi: 10.1021/acschemneuro.8b00711

    CrossRef   Google Scholar

    [3]

    Mehrtash H, Duncan K, Parascandola M, David A, Gritz ER, et al. 2017. Defining a global research and policy agenda for betel quid and areca nut. The Lancet Oncology 18:E767−E775

    doi: 10.1016/S1470-2045(17)30460-6

    CrossRef   Google Scholar

    [4]

    Pasupuleti RR, Lee CH, Osborne PG, Wu MT, Ponnusamy VK. 2022. Rapid green analytical methodology for simultaneous biomonitoring of five toxic areca nut alkaloids using UHPLC-MS/MS for predicting health hazardous risks. Journal of Hazardous Materials 422:126923

    doi: 10.1016/j.jhazmat.2021.126923

    CrossRef   Google Scholar

    [5]

    Oliveira NG, Ramos DL, Dinis-Oliveira RJ. 2021. Genetic toxicology and toxicokinetics of arecoline and related areca nut compounds: an updated review. Archives of Toxicology 95:375−93

    doi: 10.1007/s00204-020-02926-9

    CrossRef   Google Scholar

    [6]

    Huizinga JD, Chen JH, Zhu YF, Pawelka A, McGinn RJ, et al. 2014. The origin of segmentation motor activity in the intestine. Nature communications 5:3326

    doi: 10.1038/ncomms4326

    CrossRef   Google Scholar

    [7]

    Chen Q, Jiang Z, Zhang J, Cao L, Chen Z. 2021. Arecoline hydrobromide enhances jejunum smooth muscle contractility via voltage-dependent potassium channels in W/Wv mice. Physiological Research 70:437−46

    doi: 10.33549/physiolres.934557

    CrossRef   Google Scholar

    [8]

    Xu Z, Adilijiang A, Wang W, You P, Lin D, et al. 2019. Arecoline attenuates memory impairment and demyelination in a cuprizone-induced mouse model of schizophrenia. Neuroreport 30:134−38

    doi: 10.1097/WNR.0000000000001172

    CrossRef   Google Scholar

    [9]

    Tariot PN, Cohen RM, Welkowitz JA, Sunderland T, Newhouse PA, et al. 1988. Multiple-dose arecoline infusions in Alzheimer's disease. Archives of General Psychiatry 45:901−5

    doi: 10.1001/archpsyc.1988.01800340023003

    CrossRef   Google Scholar

    [10]

    Marques MM, Beland FA, Lachenmeier DW, Phillips DH, Chung FL, et al. 2021. Carcinogenicity of acrolein, crotonaldehyde, and arecoline. The Lancet Oncology 22:19−20

    doi: 10.1016/S1470-2045(20)30727-0

    CrossRef   Google Scholar

    [11]

    Guha N, Warnakulasuriya S, Vlaanderen J, Straif K. 2014. Betel quid chewing and the risk of oral and oropharyngeal cancers: a meta-analysis with implications for cancer control. International Journal of Cancer 135:1433−43

    doi: 10.1002/ijc.28643

    CrossRef   Google Scholar

    [12]

    Lee CH, Ko YC, Huang HL, Chao YY, Tsai CC, et al. 2003. The precancer risk of betel quid chewing, tobacco use and alcohol consumption in oral leukoplakia and oral submucous fibrosis in southern Taiwan. British Journal of Cancer 88:366−72

    doi: 10.1038/sj.bjc.6600727

    CrossRef   Google Scholar

    [13]

    Shih YH, Chiu KC, Wang TH, Lan WC, Tsai BH, et al. 2021. Effects of melatonin to arecoline-induced reactive oxygen species production and DNA damage in oral squamous cell carcinoma. Journal of the Formosan Medical Association 120:668−78

    doi: 10.1016/j.jfma.2020.07.037

    CrossRef   Google Scholar

    [14]

    Chuerduangphui J, Ekalaksananan T, Chaiyarit P, Patarapadungkit N, Chotiyano A, et al. 2018. Effects of arecoline on proliferation of oral squamous cell carcinoma cells by dysregulating c-Myc and miR-22, directly targeting oncostatin M. PLoS One 13:e0192009

    doi: 10.1371/journal.pone.0192009

    CrossRef   Google Scholar

    [15]

    Hsu HJ, Chang KL, Yang YH, Shieh TY. 2001. The effects of arecoline on the release of cytokines using cultured peripheral blood mononuclear cells from patients with oral mucous diseases. The Kaohsiung Journal of Medical Sciences 17:175−82

    Google Scholar

    [16]

    Shih YT, Chen PS, Wu CH, Tseng YT, Wu YC, et al. 2010. Arecoline, a major alkaloid of the areca nut, causes neurotoxicity through enhancement of oxidative stress and suppression of the antioxidant protective system. Free Radical Biology & Medicine 49:1471−79

    doi: 10.1016/j.freeradbiomed.2010.07.017

    CrossRef   Google Scholar

    [17]

    Lin WY, Tsai BCK, Day CH, Chiu PL, Chen RJ, et al. 2021. Arecoline induces heart injure via Fas/Fas ligand apoptotic pathway in heart of Sprague-Dawley rat. Environmental toxicology 36:1567−75

    doi: 10.1002/tox.23153

    CrossRef   Google Scholar

    [18]

    Wang TN, Huang MS, Lin MC, Duh TH, Lee CH, et al. 2014. Betel chewing and arecoline affects eotaxin-1, asthma and lung function. PLoS One 9:e91889

    doi: 10.1371/journal.pone.0091889

    CrossRef   Google Scholar

    [19]

    Zhou J, Sun Q, Yang Z, Zhang J. 2014. The hepatotoxicity and testicular toxicity induced by arecoline in mice and protective effects of vitamins C and E. The Korean Journal of Physiology & Pharmacology 18:143−48

    doi: 10.4196/kjpp.2014.18.2.143

    CrossRef   Google Scholar

    [20]

    Cox S, Vickers ER, Ghu S, Zoellner H. 2010. Salivary arecoline levels during areca nut chewing in human volunteers. Journal of Oral Pathology & Medicine 39:465−9

    doi: 10.1111/j.1600-0714.2009.00881.x

    CrossRef   Google Scholar

    [21]

    Venkatesh D, Puranik RS, Vanaki SS, Puranik SR. 2018. Study of salivary arecoline in areca nut chewers. Journal of Oral and Maxillofacial Pathology 22:446

    doi: 10.4103/jomfp.JOMFP_143_18

    CrossRef   Google Scholar

    [22]

    Rajalalitha P, Vali S. 2005. Molecular pathogenesis of oral submucous fibrosis − a collagen metabolic disorder. Journal of Oral Pathology & Medicine 34:321−28

    doi: 10.1111/j.1600-0714.2005.00325.x

    CrossRef   Google Scholar

    [23]

    Jeng JH, Wang YJ, Chiang BL, Lee PH, Chan CP, et al. 2003. Roles of keratinocyte inflammation in oral cancer: regulating the prostaglandin E2, interleukin-6 and TNF-α production of oral epithelial cells by areca nut extract and arecoline. Carcinogenesis 24:1301−15

    doi: 10.1093/carcin/bgg083

    CrossRef   Google Scholar

    [24]

    Kale AD, Mane DR, Shukla D. 2013. Expression of transforming growth factor β and its correlation with lipodystrophy in oral submucous fibrosis: an immunohistochemical study. Medicina oral, patologia oral y cirugia bucal 18:e12−e18

    doi: 10.4317/medoral.18226

    CrossRef   Google Scholar

    [25]

    Vindevoghel L, Kon A, Lechleider RJ, Uitto J, Roberts AB, et al. 1998. Smad-dependent transcriptional activation of human type VII collagen gene (COL7A1) promoter by transforming growth factor-β. The Journal of Biological Chemistry 273:13053−57

    doi: 10.1074/jbc.273.21.13053

    CrossRef   Google Scholar

    [26]

    Prockop DJ, Kivirikko KI. 1995. Collagens: molecular biology, diseases, and potentials for therapy. Annual Review of Biochemistry 64:403−34

    doi: 10.1146/annurev.bi.64.070195.002155

    CrossRef   Google Scholar

    [27]

    Feres-Filho EJ, Choi YJ, Han X, Takala TE, Trackman PC. 1995. Pre- and post-translational regulation of lysyl oxidase by transforming growth factor-β1 in osteoblastic MC3T3-E1 cells. The Journal of Biological Chemistry 270:30797−803

    doi: 10.1074/jbc.270.51.30797

    CrossRef   Google Scholar

    [28]

    Deng YT, Chen HM, Cheng SJ, Chiang CP, Kuo MYP. 2009. Arecoline-stimulated connective tissue growth factor production in human buccal mucosal fibroblasts: Modulation by curcumin. Oral Oncology 45:e99−e105

    doi: 10.1016/j.oraloncology.2009.04.004

    CrossRef   Google Scholar

    [29]

    Li X, Ling TY, Gao YJ, Tang DS, Li WH. 2009. Arecoline and oral keratinocytes may affect the collagen metabolism of fibroblasts. Journal of Oral Pathology & Medicine 38:422−26

    doi: 10.1111/j.1600-0714.2009.00758.x

    CrossRef   Google Scholar

    [30]

    Chang YC, Tsai CH, Lai YL, Yu CC, Chi WY, et al. 2014. Arecoline-induced myofibroblast transdifferentiation from human buccal mucosal fibroblasts is mediated by ZEB1. Journal of cellular and molecular medicine 18:698−708

    doi: 10.1111/jcmm.12219

    CrossRef   Google Scholar

    [31]

    Lee YH, Yang LC, Hu FW, Peng CY, Yu CH, et al. 2016. Elevation of Twist expression by arecoline contributes to the pathogenesis of oral submucous fibrosis. Journal of the Formosan Medical Association 115:311−17

    doi: 10.1016/j.jfma.2015.05.009

    CrossRef   Google Scholar

    [32]

    Warnakulasuriya S, Kujan O, Aguirre-Urizar JM, Bagan JV, González-Moles MÁ, et al. 2021. Oral potentially malignant disorders: A consensus report from an international seminar on nomenclature and classification, convened by the WHO Collaborating Centre for Oral Cancer. Oral Diseases 27:1862−80

    doi: 10.1111/odi.13704

    CrossRef   Google Scholar

    [33]

    Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, et al. 2021. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians 71:209−49

    doi: 10.3322/caac.21660

    CrossRef   Google Scholar

    [34]

    Chen YK, Huang HC, Lin LM, Lin CC. 1999. Primary oral squamous cell carcinoma: an analysis of 703 cases in southern Taiwan. Oral Oncology 35:173−79

    doi: 10.1016/S1368-8375(98)00101-8

    CrossRef   Google Scholar

    [35]

    Chang NW, Pei RJ, Tseng HC, Yeh KT, Chan HC, et al. 2010. Co-treating with arecoline and 4-nitroquinoline 1-oxide to establish a mouse model mimicking oral tumorigenesis. Chemico-Biological Interactions 183:231−37

    doi: 10.1016/j.cbi.2009.10.005

    CrossRef   Google Scholar

    [36]

    Tu HF, Chen MY, Lai JCY, Chen YL, Wong YW, et al. 2019. Arecoline-regulated ataxia telangiectasia mutated expression level in oral cancer progression. Head & Neck 41:2525−37

    doi: 10.1002/hed.25718

    CrossRef   Google Scholar

    [37]

    Helton ES, Chen X. 2007. p53 modulation of the DNA damage response. Journal of Cellular Biochemistry 100:883−96

    doi: 10.1002/jcb.21091

    CrossRef   Google Scholar

    [38]

    Tsai YS, Lee KW, Huang JL, Liu YS, Juo SHH, et al. 2008. Arecoline, a major alkaloid of areca nut, inhibits p53, represses DNA repair, and triggers DNA damage response in human epithelial cells. Toxicology 249:230−37

    doi: 10.1016/j.tox.2008.05.007

    CrossRef   Google Scholar

    [39]

    Lee PH, Chang MC, Chang WH, Wang TM, Wang YJ, et al. 2006. Prolonged exposure to arecoline arrested human KB epithelial cell growth: regulatory mechanisms of cell cycle and apoptosis. Toxicology 220:81−89

    doi: 10.1016/j.tox.2005.07.026

    CrossRef   Google Scholar

    [40]

    Li J, Chen S, Liao Y, Wang H, Zhou D, et al. 2022. Arecoline Is Associated With Inhibition of Cuproptosis and Proliferation of Cancer-Associated Fibroblasts in Oral Squamous Cell Carcinoma: A Potential Mechanism for Tumor Metastasis. Frontiers in oncology 12:925743

    doi: 10.3389/fonc.2022.925743

    CrossRef   Google Scholar

    [41]

    Lu TX, Rothenberg ME. 2018. MicroRNA. The Journal of Allergy and Clinical Immunology 141:1202−7

    doi: 10.1016/j.jaci.2017.08.034

    CrossRef   Google Scholar

    [42]

    Zhang Y, Wang X, Han S, Wang Y, Liu R, et al. 2021. Suppression of miR-886-3p mediated by arecoline (ARE) contributes to the progression of oral squamous cell carcinoma. Journal of Investigative Medicine 69:377−81

    doi: 10.1136/jim-2020-001405

    CrossRef   Google Scholar

    [43]

    Shiah SG, Hsiao JR, Chang WM, Chen YW, Jin YT, et al. 2014. Downregulated miR329 and miR410 promote the proliferation and invasion of oral squamous cell carcinoma by targeting Wnt-7b. Cancer Research 74:7560−72

    doi: 10.1158/0008-5472.CAN-14-0978

    CrossRef   Google Scholar

    [44]

    Shiah SG, Hsiao JR, Chang HJ, Hsu YM, Wu GH, et al. 2020. MiR-30a and miR-379 modulate retinoic acid pathway by targeting DNA methyltransferase 3B in oral cancer. Journal of Biomedical Science 27:46

    doi: 10.1186/s12929-020-00644-z

    CrossRef   Google Scholar

    [45]

    Hsieh YP, Chen KC, Chen MY, Huang LY, Su AY, et al. 2022. Epigenetic deregulation of protein tyrosine kinase 6 promotes carcinogenesis of oral squamous cell carcinoma. International Journal of Molecular Sciences 23:4495

    doi: 10.3390/ijms23094495

    CrossRef   Google Scholar

    [46]

    Lee CH, Chang JSM, Syu SH, Wong TS, Chan JYW, et al. 2015. IL-1β promotes malignant transformation and tumor aggressiveness in oral cancer. Journal of Cellular Physiology 230:875−84

    doi: 10.1002/jcp.24816

    CrossRef   Google Scholar

    [47]

    Li X, Chen W, Gao Y, Song J, Gu Y, et al. 2022. Fat mass and obesity-associated protein regulates arecoline-exposed oral cancer immune response through programmed cell death-ligand 1. Cancer Science 113:2962−73

    doi: 10.1111/cas.15332

    CrossRef   Google Scholar

    [48]

    Zhou B, Zhu W, Ren C. 2019. First steps to regulate advertising of areca nut in China. The Lancet Oncology 20:615−16

    doi: 10.1016/S1470-2045(19)30231-1

    CrossRef   Google Scholar

    [49]

    Siregar P, Audira G, Feng LY, Lee JH, Santoso F, et al. 2021. Pharmaceutical assessment suggests locomotion hyperactivity in zebrafish triggered by arecoline might be associated with multiple muscarinic acetylcholine receptors activation. Toxins 13:259

    doi: 10.3390/toxins13040259

    CrossRef   Google Scholar

    [50]

    Sun YP, Liu Q, Luo J, Guo P, Chen F, et al. 2010. Systemic administration of arecoline reduces ethanol-induced sleeping through activation of central muscarinic receptor in mice. Alcoholism, Clinical and Experimental Research 34:150−7

    doi: 10.1111/j.1530-0277.2009.01076.x

    CrossRef   Google Scholar

    [51]

    Xiao BM, Xiao NQ, Peng MJ, Gong LM, Li C, et al. 2013. Research on effects of arecoline on refreshing and acute toxicity test. China Modern Medicine 20:14−16

    doi: 10.3969/j.issn.1674-4721.2013.20.005

    CrossRef   Google Scholar

    [52]

    Ono M, Minamoto Y, Shibata S, Watanabe S. 1995. Attenuating effect of arecoline and physostigmine on an impairment of mealtime-associated activity rhythm in old rats. Physiology & behavior 57:189−91

    doi: 10.1016/0031-9384(94)00248-4

    CrossRef   Google Scholar

    [53]

    Ghelardini C, Galeotti N, Lelli C, Bartolini A. 2001. M1 receptor activation is a requirement for arecoline analgesia. Il Farmaco 56:383−85

    doi: 10.1016/S0014-827X(01)01091-6

    CrossRef   Google Scholar

    [54]

    Chandra JN, Malviya M, Sadashiva CT, Subhash MN, Rangappa KS. 2008. Effect of novel arecoline thiazolidinones as muscarinic receptor 1 agonist in Alzheimer's dementia models. Neurochemistry international 52:376−83

    doi: 10.1016/j.neuint.2007.07.006

    CrossRef   Google Scholar

    [55]

    Serikuly N, Alpyshov ET, Wang D, Wang J, Yang L, et al. 2021. Effects of acute and chronic arecoline in adult zebrafish: Anxiolytic-like activity, elevated brain monoamines and the potential role of microglia. Progress in Neuro-Psychopharmacology & Biological Psychiatry 104:109977

    doi: 10.1016/j.pnpbp.2020.109977

    CrossRef   Google Scholar

    [56]

    Molinengo L, Cassone MC, Orsetti M. 1986. Action of arecoline on the levels of acetylcholine, norepinephrine and dopamine in the mouse central nervous system. Pharmacology Biochemistry and Behavior 24:1801−3

    doi: 10.1016/0091-3057(86)90525-3

    CrossRef   Google Scholar

    [57]

    Aviello G, Knaus UG. 2018. NADPH oxidases and ROS signaling in the gastrointestinal tract. Mucosal Immunology 11:1011−23

    doi: 10.1038/s41385-018-0021-8

    CrossRef   Google Scholar

    [58]

    Jiang JM, Wang L, Gu HF, Wu K, Xiao F, et al. 2016. Arecoline Induces Neurotoxicity to PC12 Cells: Involvement in ER Stress and Disturbance of Endogenous H2S Generation. Neurochemical research 41:2140−48

    doi: 10.1007/s11064-016-1929-6

    CrossRef   Google Scholar

    [59]

    Ko AMS, Lee CH, Ko AMJ, Ko YC. 2020. Betel quid dependence mechanism and potential cessation therapy. Progress in Neuro-Psychopharmacology & Biological Psychiatry 103:109982

    doi: 10.1016/j.pnpbp.2020.109982

    CrossRef   Google Scholar

    [60]

    Chen PH, Tu HP, Wang SJ, Ko AMS, Lee CP, et al. 2012. Monoamine oxidase A variants are associated with heavy betel quid use. Addiction Biology 17:786−97

    doi: 10.1111/j.1369-1600.2011.00331.x

    CrossRef   Google Scholar

    [61]

    Papke RL, Horenstein NA, Stokes C. 2015. Nicotinic Activity of Arecoline, the Psychoactive Element of "Betel Nuts", Suggests a Basis for Habitual Use and Anti-Inflammatory Activity. PLoS One 10:e0140907

    doi: 10.1371/journal.pone.0140907

    CrossRef   Google Scholar

    [62]

    So EC, Huang YM, Hsing CH, Liao YK, Wu SN. 2015. Arecoline inhibits intermediate-conductance calcium-activated potassium channels in human glioblastoma cell lines. European journal of pharmacology 758:177−87

    doi: 10.1016/j.ejphar.2015.03.065

    CrossRef   Google Scholar

    [63]

    Demissie S, Rogers CF, Hiramoto NS, Ghanta VK, Hiramoto RN. 1995. Arecoline a muscarinic cholinergic agent conditions central pathways that modulate natural killer cell activity. Journal of neuroimmunology 59:57−63

    doi: 10.1016/0165-5728(95)00025-W

    CrossRef   Google Scholar

    [64]

    Molinengo L, Orsetti M, Pastorello B, Scordo I, Ghi P. 1995. The action of arecoline on retrieval and memory storage evaluated in the staircase maze. Neurobiology of Learning and Memory 63:167−73

    doi: 10.1006/nlme.1995.1017

    CrossRef   Google Scholar

    [65]

    Soncrant TT, Raffaele KC, Asthana S, Berardi A, Pearse Morris P, et al. 1993. Memory improvement without toxicity during chronic, low dose intravenous arecoline in Alzheimer's disease. Psychopharmacology (Berl) 112:421−27

    doi: 10.1007/BF02244889

    CrossRef   Google Scholar

    [66]

    Raffaele KC, Asthana S, Berardi A, Haxby JV, Morris PP, et al. 1996. Differential response to the cholinergic agonist arecoline among different cognitive modalities in Alzheimer's disease. Neuropsychopharmacology 15:163−70

    doi: 10.1016/0893-133X(95)00179-H

    CrossRef   Google Scholar

    [67]

    Shakya B, Siddique YH. 2018. Exploring the neurotoxicity and changes in life cycle parameters of Drosophila melanogaster exposed to arecoline. The Journal of Basic Applied Zoology 79:1

    doi: 10.1186/s41936-018-0014-x

    CrossRef   Google Scholar

    [68]

    Yan W, Zhang T, Li S, Wang Y, Zhu L, et al. 2023. oxidative stress and endoplasmic reticulum stress contributes to arecoline and its secondary metabolites-induced dyskinesia in zebrafish embryo. International Journal of Molecular Sciences 24:6327

    doi: 10.3390/ijms24076327

    CrossRef   Google Scholar

    [69]

    García-Algar O, Vall O, Alameda F, Puig C, Pellegrini M, et al. 2005. Prenatal exposure to arecoline (areca nut alkaloid) and birth outcomes. Archives of disease in childhood − Fetal and neonatal edition 90:F276−F277

    doi: 10.1136/adc.2004.061325

    CrossRef   Google Scholar

    [70]

    The Writing Committee of the Report on Cardiovascular Health and Diseases in China. 2022. Report on cardiovascular health and diseases in China 2021: an updated summary. Biomedical and Environmental Sciences 35:573−603

    doi: 10.3967/bes2022.079

    CrossRef   Google Scholar

    [71]

    Choudhury MD, Chetia P, Choudhury KD, Talukdar AD, Datta-Choudhari M. 2012. Atherogenic effect of Arecoline: A computational study. Bioinformation 8:229−32

    doi: 10.6026/97320630008229

    CrossRef   Google Scholar

    [72]

    Hung DZ, Deng JF. 1998. Acute myocardial infarction temporally related to betel nut chewing. Veterinary and Human Toxicology 40:25−28

    Google Scholar

    [73]

    Chen YC, Lee HC, Lee HH, Su HM, Lin TH, Hsu PC. 2016. Areca nut chewing complicated with non-obstructive and obstructive ST elevation myocardial infarction. Acta Cardiologica Sinica 32:103−7

    doi: 10.6515/acs20141225a

    CrossRef   Google Scholar

    [74]

    Tseng SK, Chang MC, Hsu M-L, Su CY, Chi LY, et al. 2014. Arecoline inhibits endothelial cell growth and migration and the attachment to mononuclear cells. Journal of Dental Sciences 9:258−64

    doi: 10.1016/j.jds.2012.12.015

    CrossRef   Google Scholar

    [75]

    Goto H, Tanaka N, Tanigawa K, Shimada Y, Itoh T, et al. 1997. Endothelium-dependent vasodilator effect of extract prepared from the seeds of Areca catechu on isolated rat aorta. Phytotherapy Research 11:457−59

    doi: 10.1002/(SICI)1099-1573(199709)11:6<457::AID-PTR123>3.0.CO;2-J

    CrossRef   Google Scholar

    [76]

    Beil ME, Goodman FR, Shlevin HH, Smith EF III. 1986. Evaluation of the cardiovascular effects of arecoline in the anesthetized dog. Drug Development Research 9:203−12

    doi: 10.1002/ddr.430090304

    CrossRef   Google Scholar

    [77]

    Krstić MK. 1973. The origin of the hypertensive effect of arecoline in rats. Pharmacology 9:16−26

    doi: 10.1159/000136362

    CrossRef   Google Scholar

    [78]

    Ku CW, Day CH, Ou HC, Ho TJ, Chen RJ, et al. 2021. The molecular mechanisms underlying arecoline-induced cardiac fibrosis in rats. Open Life Sciences 16:1182−92

    doi: 10.1515/biol-2021-0116

    CrossRef   Google Scholar

    [79]

    Ho TJ, Chi-Kang Tsai B, Kuo CH, Luk HN, Day CH, et al. 2022. Arecoline induces cardiotoxicity by upregulating and activating cardiac hypertrophy-related pathways in Sprague-Dawley rats. Chemico-Biological Interactions 354:109810

    doi: 10.1016/j.cbi.2022.109810

    CrossRef   Google Scholar

    [80]

    Xie DP, Chen LB, Liu CY, Zhang CL, Liu KJ, et al. 2004. Arecoline excites the colonic smooth muscle motility via M3 receptor in rabbits. The Chinese Journal of Physiology 47:89−94

    Google Scholar

    [81]

    Li CB, Yang X, Tang WB, Liu CY, Xie DP. 2010. Arecoline excites the contraction of distal colonic smooth muscle strips in rats via the M3 receptor−extracellular Ca2+ influx−Ca2+ store release pathway. Canadian Journal of Physiology and Pharmacology 88:439−47

    doi: 10.1139/Y10-024

    CrossRef   Google Scholar

    [82]

    Gemmell MA. 1958. Arecoline hydrobromide as a taentafuge in dogs, with special reference to its use in controlling hydatid disease. Australian Veterinary Journal 34:207−12

    doi: 10.1111/j.1751-0813.1958.tb05879.x

    CrossRef   Google Scholar

    [83]

    Nikitin V, Yakovlev N, Kochetov V. 1963. The efficacy of arecoline against cestodes in dogs. Veterinariya 40:53−54

    Google Scholar

    [84]

    Zhu L, Li D, Yang X. 2023. Gut metabolomics and 16S rRNA sequencing analysis of the effects of arecoline on non-alcoholic fatty liver disease in rats. Frontiers in Pharmacology 14:1132026

    doi: 10.3389/fphar.2023.1132026

    CrossRef   Google Scholar

    [85]

    Xu M, Su S, Jiang S, Li W, Zhang Z, et al. 2023. Short-term arecoline exposure affected the systemic health state of mice, in which gut microbes played an important role. Ecotoxicology and Environmental Safety 259:115055

    doi: 10.1016/j.ecoenv.2023.115055

    CrossRef   Google Scholar

    [86]

    Zhao H, Ding T, Chen Y, Yang W, Rao J, et al. 2023. Arecoline aggravates acute ulcerative colitis in mice by affecting intestinal microbiota and serum metabolites. Frontiers in Immunology 14:1197922

    doi: 10.3389/fimmu.2023.1197922

    CrossRef   Google Scholar

    [87]

    Dasgupta R, Saha I, Pal S, Bhattacharyya A, Sa G, et al. 2006. Immunosuppression, hepatotoxicity and depression of antioxidant status by arecoline in albino mice. Toxicology 227:94−104

    doi: 10.1016/j.tox.2006.07.016

    CrossRef   Google Scholar

    [88]

    Liu ST, Young GC, Lee YC, Chang YF. 2011. A preliminary report on the toxicity of arecoline on early pregnancy in mice. Food and Chemical Toxicology 49:144−48

    doi: 10.1016/j.fct.2010.10.009

    CrossRef   Google Scholar

    [89]

    Taylor RFH, Al-Jarad N, John LME, Barnes NC, Conroy DM. 1992. Betel-nut chewing and asthma. Lancet 339:1134−36

    doi: 10.1016/0140-6736(92)90732-I

    CrossRef   Google Scholar

    [90]

    Xiao RM, Wang JJ, Chen JY, Sun LJ, Chen Y. 2014. Effects of arecoline on hepatic cytochrome P450 activity and oxidative stress. The Journal of Toxicological Sciences 39:609−14

    doi: 10.2131/jts.39.609

    CrossRef   Google Scholar

    [91]

    Cheng HL, Su SJ, Huang LW, Hsieh BS, Hu YC, et al. 2010. Arecoline induces HA22T/VGH hepatoma cells to undergo anoikis - involvement of STAT3 and RhoA activation. Molecular cancer 9:126

    doi: 10.1186/1476-4598-9-126

    CrossRef   Google Scholar

    [92]

    Chan YH, Liu TC, Liao CK, Cheng YF, Tsai CH, et al. 2019. Consumption of betel quid contributes to sensorineural hearing impairment through arecoline-induced oxidative stress. Scientific Reports 9:14554

    doi: 10.1038/s41598-019-49815-5

    CrossRef   Google Scholar

    [93]

    Chang BE, Liao MH, Kuo MYP, Chen CH. 2004. Developmental toxicity of arecoline, the major alkaloid in betel nuts, in zebrafish embryos. Birth Defects Research Part A, Clinical and molecular teratology 70:28−36

    doi: 10.1002/bdra.10136

    CrossRef   Google Scholar

    [94]

    Li WD, Zang CJ, Yin S, Shen W, Sun QY, Zhao M. 2020. Metformin protects against mouse oocyte apoptosis defects induced by arecoline. Cell Proliferation 53:e12809

    doi: 10.1111/cpr.12809

    CrossRef   Google Scholar

    [95]

    Wang SW, Hwang GS, Chen TJ, Wang PS. 2008. Effects of arecoline on testosterone release in rats. American journal of physiology − Endocrinology and metabolism 295:E497−E504

    doi: 10.1152/ajpendo.00045.2008

    CrossRef   Google Scholar

    [96]

    Selvan RS, Venkateswaran KS, Rao AR. 1989. Influence of arecoline on immune system: I. Short term effects on general parameters and on the adrenal and lymphoid organs. Immunopharmacology and immunotoxicology 11:347−77

    doi: 10.3109/08923978909005375

    CrossRef   Google Scholar

    [97]

    Dasgupta R, Chatterji U, Nag TC, Chaudhuri-Sengupta S, Nag D, et al. 2010. Ultrastructural and hormonal modulations of the thyroid gland following arecoline treatment in albino mice. Molecular and Cellular Endocrinology 319:1−7

    doi: 10.1016/j.mce.2010.01.005

    CrossRef   Google Scholar

    [98]

    Wen XM, Zhang YL, Liu XM, Guo SX, Wang H. 2006. Immune responses in mice to arecoline mediated by lymphocyte muscarinic acetylcholine receptor. Cell Biology International 30:1048−53

    doi: 10.1016/j.cellbi.2006.09.015

    CrossRef   Google Scholar

    [99]

    Hsu HF, Tsou TC, Chao HR, Shy CG, Kuo YT, et al. 2010. Effects of arecoline on adipogenesis, lipolysis, and glucose uptake of adipocytes − A possible role of betel-quid chewing in metabolic syndrome. Toxicology and Applied Pharmacology 245:370−77

    doi: 10.1016/j.taap.2010.04.008

    CrossRef   Google Scholar

    [100]

    Tian ZH, Weng JT, Shih LJ, Siao AC, Chan TY, et al. 2018. Arecoline inhibits the growth of 3T3-L1 preadipocytes via AMP-activated protein kinase and reactive oxygen species pathways. PLoS One 13:e0200508

    doi: 10.1371/journal.pone.0200508

    CrossRef   Google Scholar

  • Cite this article

    Liu H, Zheng H, Zhang J, Chen F, Hu X, et al. 2024. Review of the toxic effects and health functions of arecoline on multiple organ systems. Food Innovation and Advances 3(1): 31−41 doi: 10.48130/fia-0024-0005
    Liu H, Zheng H, Zhang J, Chen F, Hu X, et al. 2024. Review of the toxic effects and health functions of arecoline on multiple organ systems. Food Innovation and Advances 3(1): 31−41 doi: 10.48130/fia-0024-0005

Figures(3)  /  Tables(2)

Article Metrics

Article views(530) PDF downloads(65)

REVIEW   Open Access    

Review of the toxic effects and health functions of arecoline on multiple organ systems

Food Innovation and Advances  3 2024, 3(1): 31-41  |  Cite this article

Abstract: Arecoline, the principal active alkaloid in the areca nut, is known for its ability to induce euphoric sensations. Since ancient times, arecoline has garnered attention for its therapeutic potential in addressing psychiatric disorders and alleviating gastrointestinal ailments. However, in 2020, the International Agency for Research on Cancer has classified arecoline as 'probably carcinogenic to humans' (Group 2B carcinogen), supported by compelling mechanistic evidence. The mechanism of action of arecoline has been extensively studied, but the results of these studies are scattered and lack systematic integration and generalization. In this paper, we have systematically summarized the mechanism of arecoline within the oral cavity, central nervous system, cardiovascular system, and digestion system, in terms of both health functions and toxic effects. In addition, we found some concentration-effect relationship between arecoline in the central nervous system and digestive system, i.e., low doses are beneficial and high doses are harmful. By summarizing the mechanisms of arecoline, this review is poised to provide in-depth and valuable insights into the clinical practice and targeted therapy of arecoline in the future.

    • Areca nut, derived from the seeds of the Areca catechu L. palm, stands as a traditional commodity deeply rooted in the cultures of Asia, East Africa, and the Western Pacific[1]. Chewing areca nut is an ancient custom followed by the people living in these areas to obtain relaxation, better concentration, and euphoria, and statistically, adult chewing rates range from 2.3% in China to 47.8% in Indonesia[2,3]. The constituents within the areca nut encompass diverse compounds, including polysaccharides, flavonoids, fatty acids, and alkaloids[4]. Among these components, alkaloids stand out as the primary active constituents, and arecoline constitutes a significant proportion of 0.3%−0.6%[1,5]. Approximately 600 million individuals worldwide consume areca nut, making arecoline the most commonly used substance by humans after alcohol, caffeine, and nicotine[2].

      Historically, areca nut has served as a medicinal plant with ancient roots. Areca nut occupies an essential position in traditional Chinese medicine classics such as the Compendium of Materia Medica and is often used to treat gastrointestinal disorders such as dysentery, bloating, and constipation[1]. Modern studies have shown that arecoline, the main active ingredient in areca nut, stimulates intestinal smooth muscle contraction and promotes intestinal peristalsis by stimulating muscarinic acetylcholine receptor (mAchR) and voltage-gated potassium channels, thus improving intestinal health[6,7]. In addition, as a psychoactive substance, arecoline can alleviate spatial working memory deficits in neurodivergent mice and cognitive deficits in Alzheimer's patients under specific conditions, demonstrating therapeutic potential for neurological disorders[8,9].

      However, in 2020, the International Agency for Research on Cancer classified arecoline as 'probably carcinogenic to humans' (Group 2B carcinogen) based on compelling mechanistic evidence[10]. Approximately half of oral cancers reported are attributed to areca nut chewing in the Indian subcontinent and Taiwan[11]. When chewing areca nut, the oral cells are rubbed by areca nut fibers and infiltrated by arecoline, prone to inflammatory reactions and collagen disorders, forming oral mucosal fibrosis, a type of oral precancerous lesion[12]. In addition, increased oxidative stress, epigenetic dysregulation, and immune dysfunction due to arecoline may also be an important cause of oral cancer[1315]. Arecoline can affect virtually every organ in the body, including but not limited to neurotoxicity[16], cardiotoxicity[17], causing asthma[18], and decreasing embryonic viability[19]. Given the widespread use of arecoline, it is particularly urgent to clarify the pharmacologic and toxicologic effects and mechanisms of arecoline on various organs.

      In this review, we briefly discuss the multifaceted actions of arecoline on various organs, considering pharmacological and toxicological perspectives, and offering a nuanced understanding of how arecoline affects different physiological systems. We delve into the health functions of arecoline on vital systems, including its influence on neurotransmitter modulation, smooth muscle contraction, and the notable antiparasitic properties of arecoline. We also underpin the toxic effects of arecoline on critical organ systems, encompassing factors like fibrosis, oxidative stress, immune dysfunction, and epigenetic alterations. Given the extensive discussions surrounding arecoline, we aim to advance the understanding of its intricate pharmacological and toxicological profiles, ultimately paving the way for developing therapeutic strategies.

    • Arecoline has been observed to accumulate in the oral cavity after entry, posing a significant risk to oral health. Salivary concentrations of arecoline in volunteers chewing 0.5 grams of areca nut ranged from 5.66 to 97.39 µg/mL[20]. Even after a brief chewing period, 100 ng/mL residual concentrations are common[21]. In vitro, arecoline can stimulate cultured cells at concentrations as low as 0.1 µg/mL and is cytotoxic at 10 µg/mL[20]. Epidemiological studies have established a correlation between regular consumption of areca nut and potentially malignant oral diseases, such as oral submucous fibrosis (OSF) and oral squamous cell carcinoma (OSCC) (Fig. 1).

      Figure 1. 

      Possible mechanisms of oral submucous fibrosis (OSF) and oral oral squamous cell carcinoma (OSCC) induced by arecoline.

    • OSF is a kind of oral potentially malignant disorder, mainly caused by areca nut chewing[22]. The physical friction of arecoline coarse fiber and the chemical irritation of arecoline can cause damage and inflammation to oral tissue, and long-term chewing habits can lead to abnormal and persistent tissue inflammation, which is a critical factor in developing cancer and tissue fibrosis[23]. Various inflammatory mediators play pivotal roles in these pathogenic processes. Arecoline is implicated in stimulating the cellular expression of pro-inflammatory and pro-fibrotic cytokines, including prostaglandin E2 (PGE2), interleukin-6 (IL-6), tumor necrosis factor α (TNF-α), transforming growth factor-β (TGF-β) and cyclooxygenase 2 (Cox-2)[23,24]. Among those inflammatory factors, arecoline enhances collagen synthesis[25], increases procollagenase levels[26], and upregulates lysyl oxidase activity (a key enzyme in collagen fiber processing)[27] by inducing TGF-β signaling[24]. Arecoline also induces the expression of connective tissue growth factor (CTGF), a downstream target of TGF-β, by activating mitogen-activated protein kinase (MAPK) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)[28]. Additionally, in oral keratinocytes-fibroblasts, tissue inhibitor of metalloproteinases 1 (TMP1), an inhibitor of enzymes involved in extracellular matrix (ECM) degradation, exhibited increased production following arecoline pre-treatment[29].

      Another possible OSF route is epithelial-mesenchymal transition (EMT). Zinc finger E-box binding homeobox 1, a transcription factor that instigates EMT, experiences increased expression under the influence of arecoline. This upregulation drives the expression of α-smooth muscle actin (α-SMA) via activation of the α-SMA promoter, thus prompting the differentiated metastasis of myofibroblasts in buccal mucosal fibroblasts, subsequently contributing to ECM accumulation and participating in OSF pathogenesis[30]. Moreover, Twist is another EMT transcription factor that plays a role in arecoline-associated OSF by regulating collagen contraction and wound healing capacity in OSF[31]. Overall, stimulation of the immune system, TMP1, and EMT, leading to disturbances in collagen homeostasis, are possible mechanisms by which arecoline causes OSF, as detailed in Fig. 1.

    • OSF is a precancerous lesion that may precede the OSCC diagnosis[32]. According to the Global Cancer Observatory, in 2020, there were 377,713 cases (2.0%) of lip and oral cancer and 177,757 deaths (1.8%)[33]. OSCC accounts for more than 90% of oral cancer cases, with a five-year survival rate of 40%−50%[34]. Approximately half of oral cancers reported are attributed to areca nut chewing in the Indian subcontinent and Taiwan[11]. In mouse models, a standard method for simulating oral tumors is a combined treatment with arecoline and 4-nitroquinoline-1-oxide[35]. These findings support that OSCC has a significant association with arecoline. Existing studies demonstrate that arecoline may cause OSCC by inducing increased oxidative stress, epigenetic dysregulation, and immune dysfunction.

      Arecoline induced the production of reactive oxygen species (ROS) and reduced expression of antioxidant enzymes, leading to chromosomal damage and gene mutations[13], one of the pathogenic mechanisms of OSCC. Typically, the body's ROS is in equilibrium with the antioxidant system, but sometimes, this equilibrium can be disturbed, such as after arecoline ingestion. When 50−200 μg/mL arecoline was used to treat gingival epithelial Smulow-Glickman cells and oral epithelial cell lines OEC-M1 and SAS, it induced ROS production and inhibited catalase expression, caused DNA double-strand breaks and activated the DNA repair response, which was characterized by down-regulation of ATM mRNA expression and an increase in p-ATM and γ-H2AX expression in the cells[13,36]. The tumor suppressor gene p53 regulates almost all DNA repair pathways[37]. Tsai et al. found that arecoline can inhibit the p53-regulated p21WAF1 promoter and p53 protein expression in KB and HEp-2 cells, inducing cell cycle arrest in S and G2/M phases[38]. Apart from these changes, arecoline also increases the phosphorylation levels of Wee1 kinase and Cdc2Tyr15 associated with S and G2/M phase arrest in KB epithelial cells[39], as well as regulating the expression of cell cycle protein D1, cell cycle protein A, cell cycle protein E, CDK4 and CDK2 in HaCaT keratinocytes[40].

      Arecoline also induces epigenetic changes in oral cells, including alterations in non-coding RNA and DNA methylation, which can lead to the development of OSCC. MicroRNA is an endogenous non-coding single-stranded RNA molecule[41]. In the ORL-48(T) squamous cell carcinoma cell line, 0.025 μg/mL arecoline decreased miR-22 expression in cells, leading to reduced inhibition of oncostatin M, an IL-6-family inflammatory cytokine, which then promotes OSCC proliferation[14]. Similarly, in OSCC cell lines, arecoline could also participate in OSCC proliferation and metastasis by inhibiting miR-886-3p[42]. In OSCC patients and mice, arecoline promotes OSCC proliferation by reducing the expression of miR-329 and miR-410 genes, inducing the expression of Wnt-7b and β-catenin proteins[43]. Abnormal DNA methylation is another common epigenetic change in OSCC. Under arecoline exposure, miR-30a and miR-379 levels were reduced in OSCC cells, targeting increased DNMT3B expression, which mediated the downregulation and methylation of ADHEF1 and ALH1A2 involved in retinoid metabolism to promote the progression of OSCC[44]. In addition, arecoline exposure resulted in hypomethylation of PTK6 with increased PTK6 expression, which increased the proliferation rate, migration, and invasion of OSCC cells, as demonstrated in mice[45].

      As the body's defense barrier, the immune system is important in recognizing and rejecting tumors. As early as 2001, it was noted that areca nut/arecoline reduced IL-2, TNF-α, TGF-β, and interferon-gamma (IFN-γ) levels in mononuclear cells of normal subjects and patients with squamous cell carcinoma[15]. In OSCC cell lines, arecoline increased the production of the pro-inflammatory factor IL-β partly through inflammasome, and IL-β induced angiogenesis and EMT, thereby promoting OSCC invasiveness[46]. Likewise, arecoline stimulated the production of PGE2, inhibited the expression of CD69 on CD4+ and CD8+ T cells in cellular KB oral cancer cells[23]. Furthermore, arecoline also increased the expression of obesity-associated protein, which regulates the expression of programmed cell death-ligand 1 via m6A modification and myc, as a means to increase the resistance of OSCC cells to CD8 T cells, thereby conferring the ability to immune escape from OSCC cells[47]. Based on the results of the current studies, long-term use of arecoline may increase the risk of OSCC. Therefore, reducing or avoiding exposure to arecoline is one of the most critical steps to prevent the risk of OSCC. In conclusion, both in vivo and in vitro studies support that arecoline impairs oral health.

    • There is a proverb in Hunan Province, China—areca nut and smoke; mana is boundless; areca nut and wine, get everything you want; areca nut, smoke, and wine, live to 99[48]. As a fat-soluble tertiary amine, arecoline crosses the blood-brain barrier well to enter and modulate the CNS, delivering a wide range of bodily effects, including euphoria, cognitive modulation, and addiction (Table 1)[2].

      Table 1.  Effect of arecoline on the CNS.

      EffectAnimal/cellSpecific effectPathway/mediatorsDoseRef.
      Beneficial effectsXenopus laevis oocytesAnti-inflammatory activityAs a silent agonist of α7 nAChR, targeting and regulating intracellular signaling against inflammation and pain/[61]
      Glioblastoma cell lines (U373 and U87MG)Interfere with the aggressiveness of malignant gliomasInhibition of intermediate conductance Ca2+-activated K+ channels10 and 30 μM[62]
      ZebrafishCluster disruption and increased social interactionIncreased norepinephrine, serotonin, and DOPAC levels decreased 5-hydroxyindoleacetic acid/serotonin level, and homovanillic acid/dopamine ratios10 mg/L[55]
      ZebrafishMotor hyperactivityBinds with multiple mAChRs (M1−M4) to induce hyperactivity0.001, 0.01, 0.1,
      and 1 ppm
      [49]
      Male Swiss albino miceAntinociceptionBy activation of central muscarinic receptors0.3−1 mg/kg ip[53]
      RatAttenuated a time perception impairment induced by daily scheduled feedingBy modulating central cholinergic10 mg/kg/d[52]
      RatAnti-phenobarbital sodium-induced sleep timeNot mentioned0.5 mg[51]
      Male ICR miceShortened the duration of
      ethanol-induced sleep
      Acts as a muscarinic agonist to relieve ethanol-induced central depression and intoxication0.125−1.0 mg/kg, s.c.[50]
      CPZ miceAttenuating memory impairment and demyelinationActs as a muscarinic receptor 1 cholinergic agonist to improve cognition and promote myelination processes in the frontal cortex2.5 or 5 mg/kg/d[8]
      Female BALB/c miceIncreased the activity of preactivated NK cellsBy stimulating the secretion of corticotropin-releasing hormone and adrenocorticotropic hormone1.5 mg/kg[63]
      Male albino ratsImproved retrieval and memory storage in the stair mazeNot mentioned0.5 mg/kg[64]
      Human (Alzheimer)Low-dose arecoline improved cognitive performance, highest-dose impaired psychomotor activationBy modulating central cholinergic1, 2, or 4 mg/h infusions 2 h[9]
      Human (Alzheimer)Improved memoryAs a cholinergic agonist, maintaining patients’ cholinergic steady-state0.042−1.7 mg/h Infusion for 11−16 d[65]
      Human (Alzheimer)Improved cognitionAs a muscarinic receptor agonist, regulating patients’ cholinergic system0.5, 1, 2, 4, 8, 16, 22,
      28, 34, and 40 mg/d
      [66]
      NeurotoxicityPrimary cortical neuronInduction of neuronal cell deathBy attenuating antioxidant defense and enhancing oxidative stress50−200 μM[16]
      PC12 CellsApoptosisBy inducing endoplasmic reticulum stress, attenuating H2S levels, CBS and 3-MST protein expression0.5−2 mM[58]
      Drosophila melanogasterNeurotoxic agent and affected the life cycle parametersBy reducing acetylcholinesterase and MAO, increasing caspase-3, caspase-9 activity, and oxidative stress20, 40 and 80 μM[67]
      ZebrafishDyskinesiaBy increasing ROS, endoplasmic reticulum stress, apoptotic p53 signaling pathway.10 μM[68]
      Male albino ratsDecreased correct responses and accelerated spontaneous decay of memoryNot mentioned3.5 and 8 mg/kg[64]
      Addiction-relatedXenopus laevis oocytesHabitual useBy activating addiction-related nAChR activity, receptors containing α4, β2, α6 and β3 subunits/[61]
      Xenopus laevis oocytesAddictionBy activating α4 nAChR100 μM[62]
      ZebrafishWithdrawal syndrome-like responsesNot mentioned1 mg/L[55]
      Pregnant womenExceptional adverse birth outcomeNot mentionedNot mentioned[69]
    • The effects of arecoline on the CNS are complex; at some doses, arecoline can cause excitability and enhance cognitive performance. In zebrafish (Danio rerio) larvae, arecoline increases locomotor activity even at concentrations as low as 0.001 ppm[49]. In mice, arecoline shortens ethanol-induced sleep time (0.125 to 1.0 mg/kg)[50]. Arecoline also increases anti-1.5 × 10−4 phenobarbital sodium-induced sleep time by up to 38 min (0.5 mg)[51]. These phenotypes suggest that arecoline has significant excitatory effects. In addition, spatial memory impairment and brain demyelination were well alleviated in schizophrenic mice treated with 5 mg/kg/d arecoline[8]. Daily arecoline injections of 10 mg/kg attenuated the impairment of mealtime-associated activity on the fasting day in old rats[52]. In Chinese medicine, herbs with areca nut as the main ingredient can manage palpitations, insomnia, and mental irregularities[1]. In clinical practice, a low dose of arecoline can improve cognitive impairment, emotional capacity, and psychomotor activity in Alzheimer's patients[9]. It was found that arecoline is an agonist of mAChRs, which may promote body excitability and antinociception effects and improve learning and memory by activating the M1 muscarinic receptor subtype[53,54]. Additionally, arecoline exposure increased dopamine levels in the brains of mice and zebrafish, which may also be a reason for arecoline's ability to promote excitation in the organism[55,56]. These findings indicate that arecoline enhances cognitive performance and induces organic excitability, possibly by modulating neurotransmitter homeostasis in the brain.

    • Surprisingly, as the concentration increases, arecoline begins to disrupt the oxidative and antioxidant balance in the body, inducing neurotoxicity and apoptosis. NADPH oxidase (NOX) is a key enzyme for redox signaling and a significant source of ROS cluster in vivo[57]. Cellular experiments indicated that arecoline at concentrations of 50−200 μM can increase ROS by upregulating NOX2 levels and decrease glutathione (GSH) and superoxide dismutase (SOD) levels, causing redox imbalance in neurons. In this state, the expression of pro-apoptotic proteins (cytochrome c, Bax, caspase-9, and caspase-3) was increased, and the manifestation of anti-apoptotic protein Bcl-2 was diminished, which finally induced neuronal cell death[16]. Jiang et al. suggested that arecoline can induce neurotoxicity by causing endoplasmic reticulum stress in neuronal cells and interfering with endogenous H2S synthesis[58]. Moreover, zebrafish showed elevated expression of the protooncogenes c-fos and c-jun in the brain after 10 mg/L arecoline treatment, which was associated with cancer transformation and progression[55].

    • After long-term consumption of areca nut, users may experience dependence such as tolerance, loss of control, craving, and salience, with surveys indicating that a high percentage of current users are dependent, accounting for 40% to 80% of the total[59]. Once discontinued, users may experience withdrawal symptoms, including mood swings, anxiety, irritability, and insomnia[2]. Dependency mechanisms are usually associated with the brain's dopamine system. As early as the 1980s, researchers showed that arecoline increased dopamine levels in the mouse cortex[56], and this was validated by the results that zebrafish exposed to acute arecoline increased brain levels of norepinephrine, serotonin, and the dopamine metabolite 3,4-Dihydroxyphenylacetic acid (DOPAC)[55]. Chen et al. emphasized that this increased dopamine may be partially derived from Monoamine oxidase A (MAO-A) inhibition, and MAO-A activity was indeed inhibited in neuroblastoma SH-SY5Y cells and rats after arecoline treatment, and individuals carrying the at-risk MAO-A allele were more likely to exhibit a dependent response in the population survey[60]. Furthermore, the α4β2 nicotinic acetylcholine receptor (nAChR) in the brain is a crucial regulator of the limbic dopamine system in the midbrain, and arecoline may mediate the rewarding effects behind habitual arecoline use through activation of the α4β2 nAChR[61].

      The studies above have shown that the effects of arecoline on the CNS are complex. At lower doses, arecoline stimulates acetylcholine receptors, improving cognition and euphoria. However, higher doses of arecoline can induce neurotoxicity, apoptosis, and cancer transformation in the CNS. Prolonged intake and abuse of arecoline can lead to addiction, tolerance, and dependence through the release of dopamine in the brain. The positive cognitive enhancement effects of arecoline can be utilized to provide a new therapeutic idea for related diseases. However, it is important to be aware of the potential neurological problems that may arise from long-term use and abuse of arecoline to safeguard our neurological health.

    • The China Cardiovascular Health and Disease Report 2021 projects that cardiovascular disease now affects 330 million people and is currently the leading cause of death for the population, accounting for more than 40%[70]. Areca nut chewing may increase the risk of cardiovascular diseases[71], for instance, following areca nut chewing, one patient with coronary artery disease experienced an acute myocardial infarction[72], while two other patients experienced ST-elevation myocardial infarction[73]. Tseng et al. pointed out that when arecoline concentration was higher than 0.2 mM, it caused contraction, rounding, and shedding of EAHY cells, weakened the wound closure activity of EAHY cells, and promoted the adhesion of monocytes to EAHY cells, which is an early step in atherosclerosis[74]. Furthermore, arecoline causes atherosclerosis by interfering with low-density lipoprotein (LDL) receptors to inhibit endocytosis of LDL cholesterol and interfering with high-density lipoprotein (HDL) receptors to prevent hepatic uptake of HDL cholesterol[71].

      In addition to atherosclerosis, Goto et al. found that a subfraction isolated from areca nut (concentration above 3 × 10−7 μg/mL) had vasodilatory effects and relaxed rat aorta with intact endothelium-containing[75]. In anesthetized dogs, only 10 ng/kg of arecoline was able to reduce arterial blood pressure. When the dose was increased to 30 or 100 μg/kg, the experimental animals even showed a sustained interruption of cardiac activity[76]. Mice also showed a decrease in blood pressure after arecoline injection[77], suggesting arecoline may have similar, evolutionarily conserved cardiovascular system effects in animals such as humans, canines, and mice.

    • Studies have established an association between arecoline and cardiovascular disease, but the underlying mechanisms regarding arecoline-induced cardiotoxicity are poorly understood. Similar to the induction of oral cell fibrosis, arecoline induces cardiomyocyte fibrosis by disrupting the balance of the extracellular matrix by affecting immunity (TGF-β) and the enzymes that synthesize and degrade the extracellular matrix. Specifically, when mice were fed arecoline above 5 mg/kg/d, significant fibrosis occurred in the heart; on the one hand, arecoline was able to increase the expression of TGF-β1 and its downstream molecule p-Smad2/3, CTGF and its transcription factor SP1 were in turn regulated by the p-Smad2/3 pathway to increase their expression, thus participating in cardiac fibrosis; on the other hand, arecoline increased plasminogen activator and plasminogen activator expression, which in turn is involved in cardiac fibrosis by inducing the expression of matrix metalloproteinases-9[78]. In a study by Lin et al., arecoline at 5 mg/kg/d and above-induced protein expression of Fas-ligand receptors (Fas) ligand, Fas and Fas-associated protein with death domain in Sprague-Dawley rat cardiomyocytes, followed by activation of caspase 8 and caspase 3, causing apoptosis[17]. When the concentration was increased to 50 mg/kg/d, the authors found that the expression levels of apoptotic factors (tBid, Bak, cytochrome c) were increased and survival biomarkers (Bcl2, BclxL) were decreased, and the mitochondrial apoptotic pathway was activated in rat cardiomyocytes[17]. In addition, recent experiments have shown that arecoline-treated mice develop signs of cardiac hypertrophy, which are associated with expression of the MEK5/ERK5 and JAK2/STAT3 signaling pathways, the MAPK signaling cascade, as well as calcium-regulated neurophosphatase and NFATc3[79].

      Together, these findings support that arecoline may influence the course of cardiovascular diseases in a dose-response relationship (Fig. 2). However, given the complexity of the cardiovascular system and the large number of causal factors of such diseases, there is no exact mechanism to elucidate the relationship between arecoline and cardiovascular system diseases.

      Figure 2. 

      Possible mechanisms of cardiovascular diseases and cardiotoxicity.

    • When peristalsis occurs in the gastrointestinal tract, it moves and mixes the food to facilitate the absorption of nutrients and water, which is essential for life[6]. Modern research has shown that arecoline, the main active ingredient in areca nut, stimulates the contraction of the gastrointestinal tract muscles in several ways. In the jejunum, arecoline hydrobromide causes smooth muscle contraction by inhibiting voltage-gated potassium channels and inducing smooth muscle cell depolarization[7]. Similarly, in mice and rabbits, arecoline induced colonic smooth muscle motility in a dose-dependent manner, stimulated by the muscarinic (M3) receptor − extracellular Ca2+ influx − Ca2+ store release pathway[80,81]. In addition, as a known antiparasitic with low toxicity, arecoline relieved symptoms of gastrointestinal disorders such as vomiting, diarrhea, and intestinal obstruction caused by the parasites through paralyzing the parasites[82,83].

    • Human health is shaped and influenced by the body's commensal microbiota, especially the gut microbiota. For example, one of the mechanisms involved in the pathogenesis of non-alcoholic fatty liver disease (NAFLD) is the alteration of gut microbiota. Zhu et al. found that the abundance of intestinal flora was increased after 0.5−5 mg/kg arecoline treatment in mice, in which Butyricicoccus, Christensenella, and Coriobacteriaceae reversed NAFLD in mice by modulating the Cox-2/PGE2 pathway as well as by increasing the protective effect of intestinal epithelial cells[84]. Additionally, alterations in gut microbiota may induce changes in immunity and exacerbate intestinal disorders. After exposure to 6 and 30 mg/kg arecoline, inflammation and dysbiosis occurred in the intestines of mice, and the results of correlation analyses indicated that arecoline may accelerate the secretion of lipopolysaccharides by promoting an increase in the abundance of the Muribaculaceae bacterium DSM 103720, which in turn encourages the development of inflammation[85]. On a similar note, treatment with 5 mg/kg arecoline decreased the proportion of probiotics in the mouse gut and increased Odoribacter, Bacteroides, and pro-inflammatory factors (IL-6, IL-1β, and TNF-α) as a means of exacerbating ulcerative enteritis in mice[86].

      In conclusion, although arecoline can treat dyspepsia, constipation, and anti-parasite, as well as improve the intestinal homeostasis of the NAFLD model through the intestinal flora, inappropriate use of arecoline can disturb the homeostasis of intestinal flora, leading to different degrees of intestinal inflammation and ecological dysregulation. Therefore, despite the medicinal value of arecoline, the mechanism of action on the gastrointestinal tract needs to be further explored in the future, aiming to protect the health of the human gastrointestinal tract while enabling the development of new therapeutic agents and targets for gastrointestinal diseases.

    • The effects of arecoline are systemic, in addition to the impact on organs mentioned above, arecoline also has effects causing bronchoconstriction, hepatotoxicity, reproductive toxicity (Fig. 3). Wang et al. first linked arecoline, Eotaxin-1, and asthma, and their results showed that concentrations of arecoline were negatively correlated with some indexes of respiratory function in asthmatic patients[18]. Arecoline also exhibits toxic effects on hepatocytes and germ cells. After arecoline treatment, hepatocytes in mice showed damage to nuclei and mitochondria, accumulation of sizeable intracellular lipid droplets, decreased expression of antioxidant substances, and an increase in hepatotoxicity markers in a dose-dependent manner[87]. In addition, the testicular weight, sperm count, and viability of male mice were significantly reduced after arecoline treatment[19]; pregnancy rate in female mice was significantly reduced and embryo growth and implantation were affected after 200 μg of arecoline treatment[88]. Interestingly, the hepatotoxicity of arecoline and reproductive toxicity to males were attenuated when supplemented with vitamins C and E, which may be related to the antioxidant function of vitamins C and E[19].

      Figure 3. 

      Effects and mechanisms of arecoline on different organs and systems.

      The immune system is an important barrier against external threats and is distributed throughout the body's tissues. For example, arecoline, when present in the host, disrupts this barrier and can lead to various diseases. In the oral cavity and cardiomyocytes, arecoline causes ECM dysregulation by promoting cytokine TGF-β expression, ultimately resulting in fibrosis[24,78]. Similarly, arecoline promotes invasion of OSCC and immune evasion by increasing pro-inflammatory factors and resistance to CD8 T cells[46,47]. In the digestive system, however, arecoline indirectly acts on immunity by altering the gut flora, causing or aggravating gut inflammation[86]. For ease of understanding, we list the effects of arecoline on each of the systems mentioned in this section and their possible mechanisms in Table 2.

      Table 2.  Other toxicological and pharmacological effects of arecoline.

      EffectAnimal/cellSpecific effectPathway/mediatorsDoseRef.
      Respiratory systemHuman and dermal and gingival fibroblastCausing lung function impairmentIn pro-inflammatory conditions (IL-4 and
      TNF-α), arecoline can induce eotaxin-1 release and alter the disease process in asthma
      25 and 100 μg/mL[18]
      HumanAsthmaPossibly related to arecoline-induced bronchoconstriction/[89]
      HepatotoxicityHuman liver microsome and Male Wistar ratsHepatotoxicityBy increasing the hepatic CYP2E1 and CYP2B activity, induced oxidative damage, liver cirrhosis, and hepatocellular carcinoma4, 20, and 100 mg/kg/d[90]
      HA22T/VGH hepatoma cellsInducing anoikisBy inhibiting STAT3 and SHP2 phosphorylation, decreasing the levels of anti-apoptotic factors, as well as by promoting the activity of pro-apoptotic factors0−100 μg/mL[91]
      Human and C57BL/6 mice’s organ of Corti and spiral ganglionsSensorineural hearing impairmentReducing cochlear explant cell activity, inducing cell death and ROS production by causing disruption of hair cells in the organ
      of Corti
      0.2, 0.8, 2, and 10 mM[92]
      MiceFatty degeneration and inflammatory infiltrationBy increasing serum alkaline phosphatase, glutamate oxaloacetate transaminase, glutamate-pyruvate transaminase, and decreasing levels of reduced glutathione, glutathione-S-transferase, SOD, and catalase10 mg/kg body weight[19]
      MiceDecreasing nuclear size; the rough endoplasmic reticulum with profusely inflated cisternae and abundance of lipid dropletsBy Upregulating SGOT and SGPT (hepatotoxicity marker enzymes) in serum5, 10, and 20 mg/kg body weight[87]
      ReproductionZebrafish embryosReducing survival of embryos with growth retardation and lower heart rateGeneral cytotoxic effects mainly due to intracellular thiol depletion0.01%−0.04% (wt/vol)[93]
      OocyteApoptosisBy disrupting actin filament dynamics, spindle assembly, and kinetochore-microtubule attachment stability, mitochondrial distribution, and increasing oxidative stress levels180 μg/mL[94]
      ICR mice and blastocystsReduction of early embryos and inhibition of blastocyst growth and expansionBy inducing DNA damage, cell cycle arrest, or apoptosis0−8.47 × 10−2 M[88]
      Male ratsStimulation of testosterone secretionBy activating L-type calcium channels, increasing 17β-hydroxysteroid dehydrogenase activity and StAR expression, thereby stimulating testosterone production1 μg/kg[95]
      Immunity and endocrineSwiss albino miceLymphocyte depletion of the thymic cortex and the B and T lymphocyte areas in the spleen and MLN, Elevated corticosterone, SGOT, and SGPT levels, and decreased white and red blood cell countsNot mentioned20 mg/kg[96]
      Adult male miceThe orientation of nuclei was irregular, follicle degeneration, a decrease in the T3, T4, number, and size of thyroid follicles, and an increase in the TSH levelMAChRs mediate the effect of arecoline on thyroid10 mg/kg[97]
      BALB/c miceReducing the spleen index, hemolysin, IL-2 production, and splenocyte proliferation induced by concanavalin A or lipopolysaccharideMediated via mAChRs2 mg/kg[98]
      FatMouse 3T3-L1 cells and humanAdipocyte dysfunctionInhibiting adipogenic differentiation, inducing adenylate cyclase-dependent lipolysis, and interfering with insulin-induced glucose uptake≥ 300 µM[99]
      3T3-L1 cellsRegulating the growth of preadipocytesInhibiting the CDK family and the CKI pathway by inactivating AMPK activity as
      well as the intracellular ROS pathway
      0−1,000 μM[100]
    • This paper provides an up-to-date review of the pharmacologic and toxicologic mechanisms of arecoline. We focus on the oral cavity, central nervous system, cardiovascular system, and digestive system and establish a framework for the pharmacological and toxicological mechanisms of systemic systems after arecoline interventions, hoping to provide some directions for refining the understanding of the mechanism of action of arecoline and its future development. We have found arecoline to have potential therapeutic effects by promoting excitation and improving learning and memory through modulation of nAChRs and mAChRs, as well as causing smooth muscle contractions, promoting intestinal peristalsis, treating indigestion, and being antiparasitic in a way that paralyzes parasites. However, arecoline has been shown to cause varying degrees of damage to various systems throughout the body, including causing OSF, OSCC, neurotoxicity, addiction, cardiotoxicity, hepatotoxicity, and reproductive toxicity. Furthermore, in the effects of arecoline on the CNS and digestive system, we found a dose-effect relationship between arecoline, pharmacology, and toxicology, i.e., low doses are beneficial while high doses are harmful. Therefore, to rationally utilize the pharmacological properties of arecoline, further studies are needed to understand the pharmacological and toxicological mechanisms of arecoline fully and to clarify the dosage-effect relationship and the long-term effects to ensure that the protection of human health and safety accompanies the development of the drug.

    • The authors confirm contribution to the paper as follows: writing - original draft: Liu H; visualization: Liu H; writing - review & editing: Zheng H, Wang X; supervision: Zheng H, Wang X; resources: Hu X, Chen F, Zheng H, Zhang J; project administration: Hu X, Chen F, Zheng H, Zhang J, Wang X; funding acquisition: Hu X, Chen F, Zheng H, Zhang J, Wang X. All authors reviewed the results and approved the final version of the manuscript.

    • All data generated or analyzed during this study are included in this published article.

      • This work was supported by the National Natural Science Foundation of China Project (32200387) and the Emergency Project for Risk Assessment of Areca Nut (Key Project of the Department of Agriculture and Rural Affairs of Hainan Province & Wanning Municipal People's Government).

      • The authors declare that they have no conflict of interest.

      • Copyright: © 2024 by the author(s). Published by Maximum Academic Press on behalf of China Agricultural University, Zhejiang University and Shenyang Agricultural University. This article is an open access article distributed under Creative Commons Attribution License (CC BY 4.0), visit https://creativecommons.org/licenses/by/4.0/.
    Figure (3)  Table (2) References (100)
  • About this article
    Cite this article
    Liu H, Zheng H, Zhang J, Chen F, Hu X, et al. 2024. Review of the toxic effects and health functions of arecoline on multiple organ systems. Food Innovation and Advances 3(1): 31−41 doi: 10.48130/fia-0024-0005
    Liu H, Zheng H, Zhang J, Chen F, Hu X, et al. 2024. Review of the toxic effects and health functions of arecoline on multiple organ systems. Food Innovation and Advances 3(1): 31−41 doi: 10.48130/fia-0024-0005

Catalog

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return