284
Views
11
CrossRef citations to date
0
Altmetric
Review Article

Biomonitoring of mycotoxin exposure using urinary biomarker approaches: a review

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 383-403 | Received 18 Dec 2018, Accepted 11 May 2019, Published online: 12 Jul 2019

References

  • Abia, W.A., et al., 2013. Bio-monitoring of mycotoxin exposure in Cameroon using a urinary multi-biomarker approach. Food and chemical toxicology, 62, 927–934.
  • Abrunhosa, L., Calado, T., and Venancio, A., 2011. Incidence of fumonisin B(2) production by Aspergillus niger in Portuguese wine regions. Journal of agricultural and food chemistry, 59 (13), 7514–7518.
  • Akdemir, C., et al., 2010. Estimation of ochratoxin A in some Turkish populations: an analysis in urine as a simple, sensitive and reliable biomarker. Food and chemical toxicology, 48 (3), 877–882.
  • Alassane-Kpembi, I., et al., 2017. Mycotoxins co-contamination: methodological aspects and biological relevance of combined toxicity studies. Critical reviews in food science and nutrition, 57 (16), 3489–3507.
  • Ali, N., Blaszkewicz, M., and Degen, G.H., 2016a. Assessment of deoxynivalenol exposure among Bangladeshi and German adults by a biomarker-based approach. Toxicology letters, 258, 20–28.
  • Ali, N., et al., 2015a. Deoxynivalenol exposure assessment for pregnant women in Bangladesh. Toxins, 7 (10), 3845–3857.
  • Ali, N., et al., 2015b. First results on citrinin biomarkers in urines from rural and urban cohorts in Bangladesh. Mycotoxin research, 31 (1), 9–16.
  • Ali, N., et al., 2016b. Urinary biomarkers of ochratoxin A and citrinin exposure in two Bangladeshi cohorts : follow-up study on regional and seasonal influences. Archives of toxicology, 90 (11), 2683–2697.
  • Ali, N., et al., 2016c. Occurrence of aflatoxin M1 in urines from rural and urban adult cohorts in Bangladesh. Archives of toxicology, 90 (7), 1749–1755.
  • Ali, N., et al., 2017a. Determination of aflatoxin M1 in urine samples indicates frequent dietary exposure to aflatoxin B1 in the Bangladeshi population. International journal of hygiene and environmental health, 220 (2), 271–281.
  • Ali, N., Muñoz, K., and Degen, G.H., 2017b. Ochratoxin A and its metabolites in urines of German adults – An assessment of variables in biomarker analysis. Toxicology letters, 275, 19–26.
  • Amirahmadi, M., et al., 2018. Simultaneous analysis of mycotoxins in corn flour using LC/MS-MS combined with a modified QuEChERS procedure. Toxin reviews, 37 (3), 187–195.
  • Amoriello, T. et al. 2018. Behaviour of durum wheat cultivars towards deoxynivalenol content: A multi-year assay in Italy. Italian Journal of Agronomy, 13(1), 12–20.
  • Assunção, R., et al., 2015. Single-compound and cumulative risk assessment of mycotoxins present in breakfast cereals consumed by children from Lisbon region, Portugal. Food and chemical toxicology, 86, 274–281.
  • Assunção, R., Silva, M.J., and Alvito, P., 2016. Challenges in risk assessment of multiple mycotoxins in food. World mycotoxin journal, 9 (5), 791–811.
  • Ayelign, A., et al., 2017. Assessment of aflatoxin exposure among young children in Ethiopia using urinary biomarkers urinary biomarkers. Food additives and contaminants: part A, 34 (9), 1606–1616.
  • Belhassen, H., et al., 2015. Zearalenone and its metabolites in urine and breast cancer risk: a case-control study in Tunisia. Chemosphere, 128, 1–6.
  • Bernhoft, A., et al., 2001. Placental transfer of the estrogenic mycotoxin zearalenone in rats. Reproductive toxicology, 15 (5), 545–550.
  • Berthiller, F., et al., 2009. Formation, determination and significance of masked and other conjugated mycotoxins. Analytical and bioanalytical chemistry, 395 (5), 1243–1252.
  • Biehl, M.L., et al., 1993. Biliary excretion and enterohepatic cycling of zearalenone in immature pigs. Toxicology and applied pharmacology, 121 (1), 152–159.
  • Borchers, A., et al., 2010. Food Safety. Clinical reviews in allergy and immunology, 39 (2), 95–141.
  • Borutova, R., et al., 2012. Co-prevalence and statistical correlations between mycotoxins in feedstuffs collected in the Asia-Oceania in 2010. Animal feed science and technology, 178 (3–4), 190–197.
  • Bottalico A., Perrone G., 2002. Toxigenic Fusarium species and mycotoxins associated with head blight in small-grain cereals in Europe. In: Logrieco A., Bailey J.A., Corazza L., Cooke B.M. (eds) Mycotoxins in Plant Disease, 611–624.
  • Bryden, W.L., 2007. Mycotoxins in the food chain: human health implications. Asia pacific journal of clinical nutrition, 16 (1), 95–101.
  • Campagnollo, F.B., et al., 2016. The prevalence and effect of unit operations for dairy products processing on the fate of aflatoxin M1: a review. Food control, 68, 310–329.
  • Cano-Sancho, G., et al., 2010. Biomonitoring of Fusarium spp. Mycotoxins: perspectives for an individual exposure assessment tool. Food science and technology international, 16 (3), 266–276.
  • Coronel, M.B., et al., 2011. Ochratoxin A and its metabolite ochratoxin alpha in urine and assessment of the exposure of inhabitants of Lleida, Spain. Food and chemical toxicology, 49 (6), 1436–1442.
  • Delongchamp, R.R., and Young, J.F., 2001. Tissue sphinganine as a biomarker of fumonisin-induced apoptosis. Food additives and contaminants, 18 (3), 255–261.
  • Deng, C., et al., 2018. Risk assessment of deoxynivalenol in high-risk area of China by human biomonitoring using an improved high throughput UPLC-MS/MS method. Scientific reports, 8 (1), 3901.
  • Diaz, D. E., 2005. The mycotoxin blue book. Nottingham, UK: Nottingham University Press.
  • Dietrich, D.R., Heussner, A.H., and O’Brien, E. 2005. Ochratoxin A: comparative pharmacokinetics and toxicological implications (experimental and domestic animals and humans). Food additives and contaminants, 22 (sup1), 45–52.
  • Domijan, A., et al., 2009. Urine ochratoxin A and sphinganine/sphingosine ratio in residents of the endemic nephropathy area in Croatia. Arhiv za higijenu rada i toksikologiju, 60 (4), 387–393.
  • Duarte, S., et al., 2010. Monitoring of ochratoxin A exposure of the Portuguese population through a nationwide urine survey-Winter 2007. The science of the total environment, 408 (5), 1195–1198.
  • Duarte, S.C., et al., 2009. Ochratoxin A exposure assessment of the inhabitants of Lisbon during winter 2007/2008 through bread and urine analysis. Food additives and contaminants, 26 (10), 1411–1420.
  • Duarte, S.C., et al., 2012. Determinants of ochratoxin A exposure – A one year follow-up study of urine levels. International journal of hygiene and environmental health, 215 (3), 360–367.
  • Ediage, E.N., et al., 2012. A direct assessment of mycotoxin biomarkers in human urine samples by liquid chromatography tandem mass spectrometry. Analytica chimica acta, 741, 58–69.
  • Ediage, E.N., et al., 2013. Multimycotoxin analysis in urines to assess infant exposure: a case study in Cameroon. Environment international, 57–58, 50–59.
  • European Commission, 2000. Opinion of the scientific committee on food on Fusarium- toxins part 2: Zearalenone (ZEA). Scientific Committee on Food, 22 june, 1–12.
  • European Commission, 2006. Commission Regulation (EC) No 1881/2006 of 19 December 2006 setting maximum levels for certain contaminants in foodstuffs. Oj L364, 5–24.
  • European Food Safety Authority, 2016. Appropriateness to set a group health-based guidance value for zearalenone and its modified forms. EFSA journal, 14 (4), 4425, 17–36.
  • Ezekiel, C.N., et al., 2014. Mycotoxin exposure in rural residents in northern Nigeria: a pilot study using multi-urinary biomarkers. Environment international, 66, 138–145.
  • Fakhri, Y., et al., 2019a. Concentration and prevalence of aflatoxin M1 in human breast milk in Iran: systematic review, meta-analysis, and carcinogenic risk assessment: a review. Journal of food protection, 82 (5), 785–795.
  • Fakhri, Y., et al., 2019b. Aflatoxin M1 in human breast milk: a global systematic review, meta-analysis, and risk assessment study (Monte Carlo simulation). Trends in food science and technology, 88, 333–342.
  • Flajs, D., and Peraica, M., 2009. Toxicological properties of citrinin. Archives of industrial hygiene and toxicology, 60 (4), 457–464.
  • Fleck, S.C., et al., 2016. Urine and serum biomonitoring of exposure to environmental estrogens II: soy isoflavones and zearalenone in pregnant women. Food and chemical toxicology, 95, 19–27.
  • Föllmann, W., et al., 2016. Biomonitoring of mycotoxins in urine: pilot study in mill workers. Journal of toxicology and environmental health – part A: current issues, 79 (22-23), 1015–1025.
  • Food and Agriculture Organization of the United Nations – FAO; World Health Organization – WHO, 2011a. Evaluation of certain food additives and contaminants: seventy-fourth report of the Joint FAO/WHO Expert Committee on Food Additives. Evaluation of certain food additives and contaminantes. WHO Technical Report Series, 966, 55–70.
  • Food and Agriculture Organization of the United Nations – FAO; World Health Organization – WHO, 2011b. Evaluation of certain contaminants in food: seventy-second report of the Joint FAO/WHO Expert Committee on Food Additives. Evaluation of certain contaminants in food. WHO Technical Report Series, 959, 1–105.
  • Franco, L.T., et al., 2019. Assessment of mycotoxin exposure and risk characterization using occurrence data in foods and urinary biomarkers in Brazil. Food and chemical toxicology, 128, 21–34.
  • Fredlund, E. et al. 2013. Deoxynivalenol and other selected Fusarium toxins in Swedish oats — Occurrence and correlation to specific Fusarium species. International Journal of Food Microbiology, 167 (2), 276–283.
  • Gambacorta, S., et al., 2013. Validation study on urinary biomarkers of exposure for aflatoxin B1, ochratoxin A, fumonisin B1, deoxynivalenol and zearalenone in piglets. World mycotoxin journal, 6 (3), 299–308.
  • Gerding, J., Cramer, B., and Humpf, H.U., 2014. Determination of mycotoxin exposure in Germany using an LC-MS/MS multibiomarker approach. Molecular nutrition and food research, 58 (12), 2358–2368.
  • Gerding, J., et al., 2015. A comparative study of the human urinary mycotoxin excretion patterns in Bangladesh, Germany, and Haiti using a rapid and sensitive LC-MS/MS approach. Mycotoxin research, 31 (3), 127–136.
  • Gonçalves, B.L., et al., 2018. Mycotoxins in fruits and fruit-based products: occurrence and methods for decontamination. Toxin reviews, 38. DOI:https://doi.org/10.1080/15569543.2018.1457056
  • Goyarts, T., and Dänicke, S., 2006. Bioavailability of the Fusarium toxin deoxynivalenol (DON) from naturally contaminated wheat for the pig. Toxicology letters, 163 (3), 171–182.
  • Gratz, S.W., Duncan, G., and Richardson, A.J., 2013. The human fecal microbiota metabolizes deoxynivalenol and deoxynivalenol-3-glucoside and may be responsible for urinary deepoxy-deoxynivalenol. Applied and environmental microbiology, 79 (6), 1821–1825.
  • Grenier, B., and Applegate, T.J., 2013. Modulation of intestinal functions following mycotoxin ingestion: meta-analysis of published experiments in animals. Toxins, 5 (2), 396–430.
  • Groopman, J.D., and Kensler, T.W., 1999. The light at the end of the tunnel for chemical-specific biomarkers: daylight or headlight? Carcinogenesis, 20 (1), 1–11.
  • Groopman, J.D., et al., 1992a. Molecular dosimetry of aflatoxin-N7-guanine in human urine obtained in The Gambia, West Africa. Cancer epidemiology, biomarkers and prevention, 1, 221–227.
  • Groopman, J.D., et al., 1992b. Molecular dosimetry in rat urine of aflatoxin- N7-guanine and other aflatoxin metabolites by multiple monoclonal antibody affinity chromatography and immunoaffinity/high performance liquid chromatography. Cancer research, 52, 267–274.
  • Groopman, J.D., et al., 1992c. Molecular dosimetry of urinary aflatoxin-DNA adducts in people living in Guangxi autonomous region, People’s Republic of China. Cancer research, 52, 45–52.
  • Groves, C.E., Morales, M., and Wright, S.H., 1998. Peritubular transport of ochratoxin A in rabbit renal proximal tubules. Journal of pharmacology and experimental therapeutics, 284, 943–948.
  • Hagelberg, S., Hult, K., and Fuchs, R., 1989. Toxicokinetics of ochratoxin A in several species and its plasma-binding properties. Journal of applied toxicology, 9 (2), 91–96.
  • Haschek, W.M., et al., 1992. Characterization of fumonisin toxicity in orally and intravenously dosed swine. Mycopathologia, 117 (1-2), 83–96.
  • Heshmati, A., et al., 2017. Co-occurrence of aflatoxins and ochratoxin A in dried fruits in Iran: dietary exposure risk assessment. Food and chemical toxicology: an international journal published for the British industrial biological research association, 106 (Pt A), 202–208.
  • Heshmati, A., et al., 2019. Changes in aflatoxins content during processing of pekmez as a traditional product of grape. Lebensmittel-wissenschaft and technologie, 103, 178–185.
  • Heyndrickx, E., et al., 2015. Human biomonitoring of multiple mycotoxins in the Belgian population: results of the BIOMYCO study. Environment international, 84, 82–89.
  • Hsieh, D.P., and Atkinson, D.N., 1991. Bisfuranoid mycotoxins: their genotoxicity and carcinogenicity. Advances in experimental medicine and biology, 283, 525–532.
  • Hussein, H., and Brasel, J., 2001. Toxicity, metabolism and impact of mycotoxins on human and animals. Toxicology, 167 (2), 101–134.
  • Huybrechts, B., et al., 2015. Fast and sensitive LC – MS/MS method measuring human mycotoxin exposure using biomarkers in urine. Archives of toxicology, 89 (11), 1993–2005.
  • International Agency for Research on Cancer. 2002. IARC Monograph on the Evaluation of Carcinogenic Risk to Humans. 82, 171. Available at: http://monographs.iarc.fr/ENG/Monographs/vol82/mono82.pdf. Accessed 17 October 18.
  • Jager, A.V., et al., 2016. Assessment of aflatoxin exposure using serum and urinary biomarkers in São Paulo, Brazil: a pilot study. International journal of hygiene and environmental health, 219 (3), 294–300.
  • Johnson, N.M., et al., 2010. Aflatoxin and PAH exposure biomarkers in a U.S. population with a high incidence of hepatocellular carcinoma. Science of the total environment, 408 (23), 6027–6031.
  • Jørgensen, K., 1998. Survey of pork, poultry, coffee, beer and pulses for ochratoxin A. Food additives and contaminants, 15 (5), 550–554.
  • Karbancioglu-Güler, F., and Heperkan, D., 2009. Natural occurrence of fumonisin B1 in dried figs as an unexpected hazard. Food and chemical toxicology : an international journal published for the British industrial biological research association, 47 (2), 289–292.
  • Khaldi, N., and Wolfe, K.H., 2011. Evolutionary origins of the fumonisin secondary metabolite gene cluster in Fusarium verticillioides and Aspergillus niger. International journal of evolutionary biology, 2011, 1.
  • Khaneghah, A M., et al., 2018a. Prevalence and concentration of ochratoxin A, zearalenone, deoxynivalenol and total aflatoxin in cereal-based products: a systematic review and meta-analysis. Food chemical and toxicology, 118, 830–848.
  • Khaneghah, A.M., et al., 2018b. Impact of unit operations during processing of cereal-based products on the levels of deoxynivalenol, total aflatoxin, ochratoxin A, and zearalenone: a systematic review and meta-analysis. Food chemistry, 268, 611–624.
  • Khaneghah, A.M., et al., 2018c. Deoxynivalenol and its masked forms: Characteristics, incidence, control and fate during wheat and wheat based products processing – A review. Trends in food science and technology, 71, 13–24.
  • Kiessling, K.H., and Pettersson, H., 1978. Metabolism of zearalenone in rat liver. Acta pharmacologica et toxicologica, 43 (4), 285–290.
  • Klapec, T., et al., 2012. Urinary ochratoxin A and ochratoxin alpha in pregnant women. Food and chemical toxicology : an international journal published for the British industrial biological research association, 50 (12), 4487–4492.
  • Kongkapan, J., et al., 2015. A Brief overview of our current understanding of nivalenol: a growing potential danger vet to be fully investigated. Israel journal of veterinary medicine, 71, 1–7.
  • Kostelanska, M. et al. 2009. Occurrence of deoxynivalenol and its major conjugate, deoxynivalenol-3-glucoside, in beer and some brewing intermediates. Journal of Agricultural and Food Chemistry, 57, 3187–3194.
  • Kuiper-Goodman, T., Scott, P.M., and Watanabe, H., 1987. Risk assessment of the mycotoxin zearalenone. Regulatory toxicology and pharmacology, 7, 253–306.
  • Lei, Y., et al., 2013. Estimation of urinary concentration of aflatoxin M1 in Chinese pregnant women. Journal of food science78 (11), T1835–T1838.
  • Magan, N., and Olsen, M., 2006. Mycotoxins in food, Boca Raton: CRC Press.
  • Mahmood Fashandi, H., Abbasi, R., and Mousavi Khaneghah, A., 2018. The detoxification of aflatoxin M1 by Lactobacillus acidophilus and Bifidobacterium spp.: a review. Journal of food processing and preservation, 42 (9), e13704.
  • Marasas, W.F., 2001. Discovery and occurrence of the fumonisins: a historical perspective. Environmental health perspectives, 2, 239–243.
  • Marasas, W.F., et al., 1988. Leukoencephalomalacia in a horse induced by fumonisin B1 isolated from Fusarium moniliforme. Onderstepoort journal of veterinary research, 55, 197–203.
  • Martinez-Larranaga, M.R., et al., 1999. Toxicokinetics and oral bioavailability of fumonisin B1. Veterinary and human toxicology, 41 (6), 357–362.
  • Mason, S., et al., 2015. A survey on relationship between diet and urinary excretion of aflatoxin M1 : a screening pilot study on Iranian population. Journal of food quality and hazards control, 2, 66–70.
  • Maul, R., et al., 2015. In vitro glucuronidation kinetics of deoxynivalenol by human and animal microsomes and recombinant human UGT enzymes. Archives of toxicology, 89 (6), 949–960.
  • Meky, F.A., et al., 2001. Deoxynivalenol-induced immunomodulation of human lymphocyte proliferation and cytokine production. Food and chemical toxicology, 39 (8), 827–836.
  • Meky, F.A., et. al., 2003. Development of a urinary biomarker of human exposure to deoxynivalenol. Food and chemical toxicology, 41 (2), 265–273.
  • Minervini, F., et. al., 2004. Toxicity and apoptosis induced by the mycotoxins nivalenol, deoxynivalenol and fumonisin B1 in a human erythroleukemia cell line. Toxicology in vitro, 18 (1), 21–28.
  • Missmer, S.A., et al., 2006. Exposure to fumonisins and the occurrence of neural tube defects along the Texas-Mexico border. Environmental health perspectives, 114 (2), 237–241.
  • Mousavi Khaneghah, A., D Chaves, R., and Akbarirad, H., 2017. Detoxification of aflatoxin M1 (AFM1) in dairy base beverages (acidophilus milk) by using different types of lactic acid bacteria – mini review. Current nutrition and food science, 13 (2), 78–81.
  • Mousavi Khaneghah, A., et al., 2018. Aflatoxins in cereals: state of the art. Journal of food safety, 38 (6), e12532.
  • Muñoz, K., Blaszkewicz, M., and Degen, H., 2010. Simultaneous analysis of ochratoxin A and its major metabolite ochratoxin alpha in plasma and urine for an advanced biomonitoring of the mycotoxin. Journal of chromatography b, 878 (27), 2623–2629.
  • Olsen, M., Pettersson, H., and Kiessling, K.H., 1981. Reduction of zearalenone to zearalenol in female rat liver by 3 α-hydroxy-steroid dehydrogenase. Acta pharmacologica et toxicologica, 48 (2), 157–161.
  • Olsen, M.E., et al., 1985. Quantitative liquid chromatographic method using fluorescence detection for determining zearalenone and its metabolites in blood plasma and urine. Journal association of official analytical chemists, 68, 632–635.
  • Ostry, V., et al., 2017. Mycotoxins as human carcinogens – The IARC monographs classification. Mycotoxin research, 33 (1), 65–73.
  • Oteiza, J.M., et al., 2017. Influence of production on the presence of patulin and ochratoxin A in fruit juices and wines of Argentina. LWT-Food Science and Technology, 80, 200–207.
  • Papageorgiou, M., et al., 2018a. Assessment of urinary deoxynivalenol biomarkers in UK children and adolescents. Toxins, 10 (2), 1–13.
  • Papageorgiou, M., et al., 2018b. Occurrence of deoxynivalenol in an elderly cohort in the UK: a biomonitoring approach approach. Food additives and contaminants: part A, 49, 2032–2044.
  • Paustenbach, D., and Galbraith, D., 2006. Biomonitoring and bio-markers: exposure assessment will never be the same. Environmental health perspectives, 114 (8), 1143–1149.
  • Pestka, J.J., and Smolinski, A.T., 2005. Deoxynivalenol: toxicology and potential effects on humans. Journal of toxicology and environmental health – part B critical reviews, 8 (1), 39–69.
  • Pestka, J.J., et al., 2017. Sex is a determinant for deoxynivalenol metabolism and elimination in the mouse. Toxins (Basel), 9, 1–12.
  • Petzinger, E., and Ziegler, K., 2000. Ochratoxin A from a toxicological perspective. Journal of veterinary pharmacology and therapeutics, 23, 91–98.
  • Piekkola, S., et al., 2012. Characterisation of aflatoxin and deoxynivalenol exposure among pregnant Egyptian women. Food additives and contaminants: part A chemistry, 29 (6), 962–971.
  • Polychronaki, N., et al., 2008. Urinary biomarkers of aflatoxin exposure in young children from Egypt and Guinea. Food and chemical toxicology : an international journal published for the British industrial biological research association, 46 (2), 519–526.
  • Poór, M., et al., 2017. Interaction of mycotoxin zearalenone with human serum albumin. Journal of photochemistry and photobiology B, biology, 170, 16–24.
  • Qian, G.-S., et al., 1994. A follow-up study of urinary markers of aflatoxin exposure and liver cancer risk in Shanghai, People’s Republic of China. Cancer epidemiology, biomarkers and prevention, 3, 3–10.
  • Rahmani, J., et al., 2018. The prevalence of aflatoxin M1 in milk of Middle East region: a systematic review, meta-analysis and probabilistic health risk assessment. Food and chemical toxicology, 118, 653–666.
  • Rastegar, H., et al., 2017. Removal of aflatoxin B1 by roasting with lemon juice and/or citric acid in contaminated pistachio nuts. Food control, 71, 279–284.
  • Redzwan, S.M., et al., 2012. Association between aflatoxin M1 excreted in human urine samples with the consumption of milk and dairy products. Bulletin of environmental contamination and toxicology, 89 (6), 1115–1119.
  • Riley, R. T., and Voss, K. A., 2011. Developing mechanism-based and exposure biomarkers for mycotoxins in animals. In: S. De Saeger, ed. Determining mycotoxins and mycotoxigenic fungi in food and feed. Cambridge, UK: Woodhead Publishing Limited, 245–275.
  • Riley, R.T., et al., 2012. The kinetics of urinary fumonisin B1 excretion in humans consuming maize-based diets. Molecular nutrition and food research, 56, 1445–1455.
  • Ringot, D., et al., 2006. Toxicokinetics and toxicodynamics of ochratoxin A, an update. Chemico-biological interactions, 159 (1), 18–46.
  • Rizzo, A., Eskola, M., and Atroshi, F., 2002. Ochratoxin A in cereals, foodstuffs and human plasma. European journal of plant pathology, 108 (7), 631–637.
  • Rodríguez-Carrasco, Y., et al., 2014. Exposure assessment approach through mycotoxin/creatinine ratio in urine by GC-MS/MS. Food and chemical toxicology, 72, 69–75.
  • Romero, A.C., et al., 2010. Occurrence of AFM1 in urine samples of a Brazilian population and association with food consumption. Food control, 21 (4), 554–558.
  • Sabbioni, G., 1990. Chemical and physical properties of the major serum albumin adduct of aflatoxin B1 and their implications for the quantification in biological samples. Chemico-biological interactions, 75 (1), 1–15.
  • Sarkanj, B., et al., 2013. Urinary analysis reveals high deoxynivalenol exposure in pregnant women from Croatia. Food and chemical toxicology journal, 62, 231–237.
  • Sarkanj, B., et al., 2018. Ultra-sensitive, stable isotope assisted quantification of multiple urinary mycotoxin exposure biomarkers. Analytica chimica acta, 1019, 84–92.
  • Sass, D.C., et al., 2013. Methods for chemical preparation of aflatoxin B1 adducts, AFB1-N7-guanine and AFB1-lysine. Toxin reviews, 32, 68–74.
  • Schlatter, C., StuderRohr, J., and Rasonyi, T., 1996. Carcinogenicity and kinetic aspects of ochratoxin A. Food additives and contaminants, 13, 43–44.
  • Schoevers, E.J., et al., 2012. Transgenerational toxicity of zearalenone in pigs. Reproductive toxicology (Elmsford, N.Y.), 34 (1), 110–119.
  • Schwartzbord, J., Severe, L., and Brown, D., 2017. Detection of trace aflatoxin M1 in human urine using a commercial ELISA followed by HPLC. Biomarkers, 22 (1), 1–4.
  • Scott, P.M., 2005. Biomarkers of human exposure to ochratoxin A. Food additives and contaminants, 22 (sup1), 99–107.
  • Shephard, G.S., et al., 2013. Multiple mycotoxin exposure determined by urinary biomarkers in rural subsistence farmers in the former Transkei, South Africa. Food and chemical toxicology, 62, 217–225.
  • Shephard, G.S., van der Westhuizen, L., and Sewram, V., 2007. Biomarkers of exposure to fumonisin mycotoxins: a review. Food additives and contaminants, 24 (10), 1196–1201.
  • Shirima, C.P., et al., 2013. Dietary exposure to aflatoxin and fumonisin among Tanzanian children as determined using biomarkers. Molecular nutrition and food research, 1 (57), 1874–1881.
  • Shirima, C.P., et al., 2015. A prospective study of growth and biomarkers of exposure to aflatoxin and fumonisin during early childhood in Tanzania. Research children’s health, 123 (2), 173–179.
  • Solfrizzo, M., et al., 2011. Simultaneous LC-MS/MS determination of aflatoxin M1, ochratoxin A, deoxynivalenol, de-epoxydeoxynivalenol, α and β-zearalenols and fumonisin B1 in urine as a multi-biomarker method to assess exposure to mycotoxins. Analytical and bioanalytical chemistry, 401 (9), 2831–2841.
  • Solfrizzo, M., Gambacorta, L., and Visconti, A., 2014. Assessment of multi-mycotoxin exposure in southern Italy by urinary multi-biomarker determination. Toxins, 6 (2), 523–538.
  • Srey, C., et al., 2014. Deoxynivalenol exposure assessment in young children in Tanzania. Molecular nutrition & food research, 58, 1574–1580.
  • Studer-Rohr, I., Schlatter, J., and Dietrich, D.R., 2000. Kinetic parameters and intraindividual fluctuations of ochratoxin A plasma levels in humans. Archives of toxicology, 74 (9), 499–510.
  • Sun, L.H., et al., 2016. Prevention of aflatoxin B1 hepatoxicity by dietary selenium is associated with inhibition of cytochrome P450 isozymes and up-regulation of 6 selenoprotein genes in chick liver. The journal of nutrition, 146 (4), 655–661.
  • Susca, A., et al., 2010. Correlation of mycotoxin fumonisin B2 production and presence of the fumonisin biosynthetic gene fum8 in Aspergillus niger from grape. Journal of agricultural and food chemistry, 58 (16), 9266–9272.
  • Tanaka, T. et al. 1990. A survey of the natural occurrence of Fusarium mycotoxins, deoxynivalenol, nivalenol and zearalenone, in cereals harvested in the Netherlands. Mycopathologia, 110 (1), 19–22.
  • Torres, O., et al., 2014. Urinary fumonisin B1 and estimated fumonisin intake in women from high- and low-exposure communities in Guatemala. Molecular nutrition and food research, 58 (5), 973–983.
  • Tsegaye, W., et al., 2016. Urinary aflatoxin M1 concentrations among pregnant women in Bishoftu, Ethiopia. The FASEB journal, 30, 1149.28.
  • Turner, N.W., Subrahmanyam, S., and Piletsky, S.A., 2009. Analytical methods for determination of mycotoxins: a review. Analytica chimica acta, 632 (2), 168–180.
  • Turner, P.C., et al., 2008. Deoxynivalenol: rationale for development and application of a urinary biomarker. Food additives and contaminants: part A, chemistry, analysis, control, exposure & risk assessment, 25, 864–871.
  • Turner, P.C., et al., 2010a. A comparison of deoxynivalenol intake and urinary deoxynivalenol in UK adults. Biomarkers : biochemical indicators of exposure, response, and susceptibility to chemicals, 15 (6), 553–562.
  • Turner, P.C., et al., 2010b. Determinants of urinary deoxynivalenol and de-epoxy deoxynivalenol in male farmers from Normandy, France. Journal of agricultural and food chemistry, 58 (8), 5206–5212.
  • Turner, P.C., et al., 2011. A biomarker survey of urinary deoxynivalenol in China: the Shanghai women’s health study. Food additives and contaminants: part A chemistry, 28, 1220–1223.
  • Turner, P.C., et al., 2012. A pilot survey for Fusarium mycotoxin biomarkers in women from Golestan, northern Iran. World mycotoxin journal, 5 (2), 195–199.
  • Ueno, Y., et al., 1977. Fate and mode of action of zearalenone. Annales de la nutrition et de l'alimentation, 31 (4-6), 935–948.
  • Varga, J., et al., 2010. Fumonisin contamination and fumonisin producing black Aspergilli in dried vine fruits of different origin. International journal of food microbiology, 143 (3), 143–149.
  • Vidal, A., et al., 2018. Humans significantly metabolize and excrete the mycotoxin deoxynivalenol and its modified form deoxynivalenol-3-glucoside within 24 hours. Scientific reports, 8, 1–11.
  • Videmann, B., et al., 2008. Metabolism and transfer of the mycotoxin zearalenone in human intestinal Caco-2 cells. Food and chemical toxicology, 46 (10), 3279–3286.
  • Wallin, S., et al., 2013. Biomonitoring study of deoxynivalenol exposure and association with typical cereal consumption in Swedish adults. World mycotoxin journal, 6 (4), 439–448.
  • Wallin, S., et al., 2015. Biomonitoring of concurrent mycotoxin exposure among adults in Sweden through urinary multi-biomarker analysis. Food and chemical toxicology: an international journal published for the British industrial biological research association, 83, 133–139.
  • Wan Norhasima, W.M., et al., 2009. The health and toxic adverse effects of Fusarium fungal mycotoxin, fumonisins, on human population. American journal of infectious diseases, 4, 273–281.
  • Warth, B., et al., 2012. Development and validation of a rapid multi-biomarker liquid chromatography/tandem mass spectrometry method to assess human exposure to mycotoxins. Rapid communications in mass spectrometry : rcm, 26 (13), 1533–1540.
  • Warth, B., et al., 2013. New insights into the human metabolism of the Fusarium mycotoxins deoxynivalenol and zearalenone. Toxicology letters, 220 (1), 88–94.
  • Warth, B., et al., 2014. Utilising an LC-MS/MS-based multi-biomarker approach to assess mycotoxin exposure in the Bangkok metropolitan area and surrounding provinces. Food additives and contaminants – part A chemistry, analysis, control, exposure and risk assessment, 31 (12), 2040–2046.
  • Wells, L., et al., 2017. Deoxynivalenol biomarkers in the urine of UK vegetarians. Toxins, 9 (7), 1–12.
  • World Health Organization 1993. International programme on chemical safety, biomarkers and risk assessment: concepts and principles, environmental health criteria 155. Geneva, Switzerland: World Health Organization.
  • World Health Organization 2000. Fumonisin B1 – Environmental health criteria 219. International programme on chemical safety. Geneva: World Health Organization.
  • Wu, L., et al., 2015. Growth performance, serum biochemical profile, jejunal morphology, and the expression of nutrients transporter genes in deoxynivalenol (DON)-challenged growing pigs. BMC veterinary research, 11, 144–154.
  • Wu, Q., et al., 2010. Metabolic pathways of trichothecenes. Drug metabolism reviews, 42 (2), 250–267.
  • Xu, L., et al., 2010. Evaluation of fumonisin biomarkers in a cross-sectional study with two high-risk populations in China. Food additives and contaminants. part A, chemistry, analysis, control, exposure and risk assessment, 27 (8), 1161–1169.
  • Zain, M.E., 2011. Impact of mycotoxins on humans and animals. Journal of saudi chemical society, 15 (2), 129–144.
  • Zhu, J., et al., 1987. Correlation of dietary aflatoxin B1 levels with excretion of aflatoxin M1 in human urine. Cancer research, 47 (7), 1848–1852.
  • Zinedine, A., et al., 2007. Review on the toxicity, occurrence, metabolism, detoxification, regulations and intake of zearalenone: an oestrogenic mycotoxin. Food and chemical toxicology : an international journal published for the British industrial biological research association, 45 (1), 1–18.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.