1
|
Pires RC, da Costa Calumby J, Rosim RE, Pires RD, Borowsky AM, Ali S, de Paiva EL, Silva R, Pimentel TC, da Cruz AG, de Oliveira CAF, Corassin CH. Evaluation of Ability of Inactivated Biomasses of Lacticaseibacillus rhamnosus and Saccharomyces cerevisiae to Adsorb Aflatoxin B 1 In Vitro. Foods 2024; 13:3299. [PMID: 39456361 PMCID: PMC11506918 DOI: 10.3390/foods13203299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Biological decontamination strategies using microorganisms to adsorb aflatoxins have shown promising results for reducing the dietary exposure to these contaminants. In this study, the ability of inactivated biomasses of Lacticaseibacillus rhamnosus (LRB) and Saccharomyces cerevisiae (SCB) incorporated alone or in combination into functional yogurts (FY) at 0.5-4.0% (w/w) to adsorb aflatoxin B1 (AFB1) was evaluated in vitro. Higher adsorption percentages (86.9-91.2%) were observed in FY containing 1.0% LR + SC or 2.0% SC (w/w). The survival of mouse embryonic fibroblasts increased after exposure to yogurts containing LC + SC at 1.0-4.0% (w/w). No significant differences were noted in the physicochemical and sensory characteristics between aflatoxin-free FY and control yogurts (no biomass) after 30 days of storage. The incorporation of combined LRB and SCB into yogurts as vehicles for these inactivated biomasses is a promising alternative for reducing the exposure to dietary AFB1. The results of this trial support further studies to develop practical applications aiming at the scalability of using the biomasses evaluated in functional foods to mitigate aflatoxin exposure.
Collapse
Affiliation(s)
- Rogério Cury Pires
- Departamento de Zootecnia, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Av. Pádua Dias, 11, Piracicaba 13418-900, SP, Brazil; (R.C.P.); (R.D.P.); (A.M.B.)
| | - Julia da Costa Calumby
- Departamento de Engenharia de Alimentos, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (J.d.C.C.); (R.E.R.); (S.A.); (E.L.d.P.); (C.H.C.)
| | - Roice Eliana Rosim
- Departamento de Engenharia de Alimentos, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (J.d.C.C.); (R.E.R.); (S.A.); (E.L.d.P.); (C.H.C.)
| | - Rogério D’Antonio Pires
- Departamento de Zootecnia, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Av. Pádua Dias, 11, Piracicaba 13418-900, SP, Brazil; (R.C.P.); (R.D.P.); (A.M.B.)
| | - Aline Moreira Borowsky
- Departamento de Zootecnia, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Av. Pádua Dias, 11, Piracicaba 13418-900, SP, Brazil; (R.C.P.); (R.D.P.); (A.M.B.)
| | - Sher Ali
- Departamento de Engenharia de Alimentos, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (J.d.C.C.); (R.E.R.); (S.A.); (E.L.d.P.); (C.H.C.)
| | - Esther Lima de Paiva
- Departamento de Engenharia de Alimentos, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (J.d.C.C.); (R.E.R.); (S.A.); (E.L.d.P.); (C.H.C.)
| | - Ramon Silva
- Instituto Federal do Paraná, R. Felipe Tequinha Street, 1400, Paranavaí 87703-536, PR, Brazil; (R.S.); (T.C.P.)
| | - Tatiana Colombo Pimentel
- Instituto Federal do Paraná, R. Felipe Tequinha Street, 1400, Paranavaí 87703-536, PR, Brazil; (R.S.); (T.C.P.)
| | - Adriano Gomes da Cruz
- Departamento de Alimentos, Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro, R. Sen. Furtado, 121/125, Rio de Janeiro 20270-021, RJ, Brazil;
| | - Carlos Augusto Fernandes de Oliveira
- Departamento de Engenharia de Alimentos, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (J.d.C.C.); (R.E.R.); (S.A.); (E.L.d.P.); (C.H.C.)
| | - Carlos Humberto Corassin
- Departamento de Engenharia de Alimentos, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Av. Duque de Caxias Norte, 225, Pirassununga 13635-900, SP, Brazil; (J.d.C.C.); (R.E.R.); (S.A.); (E.L.d.P.); (C.H.C.)
| |
Collapse
|
2
|
Ali O, Szabó A. Fumonisin distorts the cellular membrane lipid profile: A mechanistic insight. Toxicology 2024; 506:153860. [PMID: 38871209 DOI: 10.1016/j.tox.2024.153860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Monitoring modifications in membrane lipids in association with external stimuli/agents, including fumonisins (FUMs), is a widely employed approach to assess cellular metabolic response/status. FUMs are prevalent fusariotoxins worldwide that have diverse structures with varying toxicity across species; nevertheless, they can induce metabolic disturbances and disease, including cancer. The capacity of FUMs to disrupt membrane lipids, demonstrated across numerous species and organs/tissues, is ascribed to a multitude of factors/events, which range from direct to indirect effects. Certain events are well established, whereas the potential consequences of others remain speculative. The most notable effect is their resemblance to sphingoid bases, which impacts the synthesis of ceramides leading to numerous changes in lipids' composition that are not limited to sphingolipids' composition of the membranes. The next plausible scenario involves the induction of oxidative stress, which is considered an indirect/secondary effect of FUMs. Additional modes of action include modifications of enzyme activities and nuclear signals related to lipid metabolism, although these are likely not yet fully comprehended. This review provides in-depth insight into the current state of these events and their potential mechanistic actions in modifying membrane lipids, with a focus on long-chain fatty acids. This paper also presents a detailed description of the reported modifications to membrane lipids by FUMs.
Collapse
Affiliation(s)
- Omeralfaroug Ali
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, Kaposvár 7400, Hungary.
| | - András Szabó
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Institute of Physiology and Animal Nutrition, Department of Animal Physiology and Health, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, Kaposvár 7400, Hungary; HUN-REN-MATE Mycotoxins in the Food Chain Research Group, Hungarian University of Agriculture and Life Sciences, Guba Sándor Str. 40, Kaposvár 7400, Hungary
| |
Collapse
|
3
|
Gao Z, Luo K, Zhu Q, Peng J, Liu C, Wang X, Li S, Zhang H. The natural occurrence, toxicity mechanisms and management strategies of Fumonisin B1:A review. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 320:121065. [PMID: 36639041 DOI: 10.1016/j.envpol.2023.121065] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 06/17/2023]
Abstract
Fumonisin B1 (FB1) contaminates various crops, causing huge losses to agriculture and livestock worldwide. This review summarizes the occurrence regularity, toxicity, toxic mechanisms and management strategies of FB1. Specifically, FB1 contamination is particularly serious in developing countries, humid and hot regions. FB1 exposure can produce different toxic effects on the nervous system, respiratory system, digestive system and reproductive system. Furthermore, FB1 can also cause systemic immunotoxicity. The mechanism of toxic effects of FB1 is to interfere with the normal pathway of sphingolipid de novo biosynthesis by acting as a competitive inhibitor of ceramide synthase. Meanwhile, the toxic products of sphingolipid metabolic disorders can cause oxidative stress and apoptosis. FB1 also often causes feed contamination by mixing with other mycotoxins, and then exerts combined toxicity. For detection, lateral flow dipstick technology and enzyme linked immunosorbent assay are widely used in the detection of FB1 in commercial feeds, while mainstream detection methods such as high performance liquid chromatography and liquid chromatography-mass spectrometry are widely used in the laboratory theoretical study of FB1. For purification means of FB1, some natural plant extracts (such as Zingiber officinale and Litsea Cubeba essential oil) and their active compounds have been proved to inhibit the toxic effects of FB1 and protect livestock due to their antifungal and antioxidant effects. Natural plant extract has the advantages of high efficiency, low cost and no contamination residue. This review can provide information for comprehensive understanding of FB1, and provide reference for formulating reasonable treatment and management strategies in livestock production.
Collapse
Affiliation(s)
- Zhicheng Gao
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, 510642, People's Republic of China
| | - Kangxin Luo
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, 510642, People's Republic of China
| | - Qiuxiang Zhu
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, 510642, People's Republic of China
| | - Jinghui Peng
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, 510642, People's Republic of China
| | - Chang Liu
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, 510642, People's Republic of China
| | - Xiaoyue Wang
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, 510642, People's Republic of China
| | - Shoujun Li
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, 510642, People's Republic of China
| | - Haiyang Zhang
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, 510642, People's Republic of China.
| |
Collapse
|
4
|
Li K, Li H, Song X, Kuang X, Liu S, Zhu S, Li D. The preventive effect of mussel oil on gestational diabetes mellitus in pregnant mice fed by a high-fat and high-sucrose diet. Food Funct 2023; 14:1198-1208. [PMID: 36602239 DOI: 10.1039/d2fo03320h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The present study aimed to investigate the preventive effect of mussel oil (MO) on gestational diabetes mellitus (GDM) in mice fed by a high-fat and high-sucrose (HFHS) diet. Pregnant mice were allocated to four groups: normal diet + corn oil (CO), HFHS + CO, HFHS + fish oil (FO), and HFHS + MO. The total n-3 polyunsaturated fatty acids (PUFAs) in MO (51.30%) and FO (48.25%) were comparable (mainly C22:6n-3 and C20:5n-3). HFHS + MO and HHFS + FO had a significantly lower area under the curve (AUC) for the oral glucose tolerance test (OGTT) than the HFHS + CO group. The HFHS + MO group but not HFHS + FO group had a significantly lower AUC for the insulin tolerance test (ITT) than the HFHS + CO group. The HFHS + MO group had significantly lower homeostasis model assessment-insulin resistance (HOMA-IR) and fasting serum insulin than the HHFS + FO and HFHS + CO groups. Liver sphingosine kinase 1 (SphK1) was significantly higher, while SphK2, Akt, and P-Akt were significantly lower in the HFHS + CO group compared with the normal diet + CO group. The HFHS + MO group but not the HFHS + FO group had significantly higher SphK2, Akt, and P-Akt than the HFHS + CO group. SphK2 had a strong negative correlation with the AUC for the OGTT (r = -0.759, p = 0.001) and insulin tolerance test (ITT) (r = -0.637; p = 0.008), fasting serum insulin (r = -0.594, p = 0.015), fasting blood glucose (r = -0.587, p = 0.017) and HOMA-IR (r = -0.629, p = 0.009) and a strong positive correlation with Akt (r = 0.594, p = 0.015) and P-Akt (r = 0.676, p = 0.004). In conclusion, mussel oil improved glucose intolerance and insulin resistance during mice pregnancy, which was superior to the effects of fish oil.
Collapse
Affiliation(s)
- Kelei Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.,School of Public Health, Qingdao University, Qingdao, China.
| | - Huiying Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.,School of Public Health, Qingdao University, Qingdao, China.
| | - Xiaolei Song
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.,School of Public Health, Qingdao University, Qingdao, China.
| | - Xiaotong Kuang
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.,School of Public Health, Qingdao University, Qingdao, China.
| | - Shiyi Liu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.,School of Public Health, Qingdao University, Qingdao, China.
| | - Suqing Zhu
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.,School of Public Health, Qingdao University, Qingdao, China.
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, China.,School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
5
|
Alvito P, Assunção RM, Bajard L, Martins C, Mengelers MJB, Mol H, Namorado S, van den Brand AD, Vasco E, Viegas S, Silva MJ. Current Advances, Research Needs and Gaps in Mycotoxins Biomonitoring under the HBM4EU-Lessons Learned and Future Trends. Toxins (Basel) 2022; 14:826. [PMID: 36548723 PMCID: PMC9783896 DOI: 10.3390/toxins14120826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022] Open
Abstract
Mycotoxins are natural metabolites produced by fungi that contaminate food and feed worldwide. They can pose a threat to human and animal health, mainly causing chronic effects, e.g., immunotoxic and carcinogenic. Due to climate change, an increase in European population exposure to mycotoxins is expected to occur, raising public health concerns. This urges us to assess the current human exposure to mycotoxins in Europe to allow monitoring exposure and prevent future health impacts. The mycotoxins deoxynivalenol (DON) and fumonisin B1 (FB1) were considered as priority substances to be studied within the European Human Biomonitoring Initiative (HBM4EU) to generate knowledge on internal exposure and their potential health impacts. Several policy questions were addressed concerning hazard characterization, exposure and risk assessment. The present article presents the current advances attained under the HBM4EU, research needs and gaps. Overall, the knowledge on the European population risk from exposure to DON was improved by using new harmonised data and a newly derived reference value. In addition, mechanistic information on FB1 was, for the first time, organized into an adverse outcome pathway for a congenital anomaly. It is expected that this knowledge will support policy making and contribute to driving new Human Biomonitoring (HBM) studies on mycotoxin exposure in Europe.
Collapse
Affiliation(s)
- Paula Alvito
- National Institute of Health Dr. Ricardo Jorge (INSA), 1649-016 Lisboa, Portugal
- Centre for Environmental and Marine Studies (CESAM), University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Ricardo Manuel Assunção
- National Institute of Health Dr. Ricardo Jorge (INSA), 1649-016 Lisboa, Portugal
- Centre for Environmental and Marine Studies (CESAM), University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
- IUEM, Instituto Universitário Egas Moniz, Egas Moniz-Cooperativa de Ensino Superior, CRL, Campus Universitário—Quinta da Granja, Monte da Caparica, 2829-511 Caparica, Portugal
| | - Lola Bajard
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, 611 37 Brno, Czech Republic
| | - Carla Martins
- National Institute of Health Dr. Ricardo Jorge (INSA), 1649-016 Lisboa, Portugal
- NOVA National School of Public Health, NOVA University of Lisbon, 1600-560 Lisbon, Portugal
- Comprehensive Health Research Center, CHRC, 1600-560 Lisbon, Portugal
| | - Marcel J. B. Mengelers
- National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, The Netherlands
| | - Hans Mol
- Wageningen Food Safety Research (WFSR), Part of Wageningen University & Research, 6708 WB Wageningen, The Netherlands
| | - Sónia Namorado
- National Institute of Health Dr. Ricardo Jorge (INSA), 1649-016 Lisboa, Portugal
- Comprehensive Health Research Center, CHRC, 1600-560 Lisbon, Portugal
| | - Annick D. van den Brand
- National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, The Netherlands
| | - Elsa Vasco
- National Institute of Health Dr. Ricardo Jorge (INSA), 1649-016 Lisboa, Portugal
| | - Susana Viegas
- NOVA National School of Public Health, NOVA University of Lisbon, 1600-560 Lisbon, Portugal
- Comprehensive Health Research Center, CHRC, 1600-560 Lisbon, Portugal
| | - Maria João Silva
- National Institute of Health Dr. Ricardo Jorge (INSA), 1649-016 Lisboa, Portugal
- ToxOmics—NOVA Medical School, NOVA University of Lisbon, 1150-082 Lisboa, Portugal
| |
Collapse
|
6
|
van den Brand AD, Bajard L, Steffensen IL, Brantsæter AL, Dirven HAAM, Louisse J, Peijnenburg A, Ndaw S, Mantovani A, De Santis B, Mengelers MJB. Providing Biological Plausibility for Exposure-Health Relationships for the Mycotoxins Deoxynivalenol (DON) and Fumonisin B1 (FB1) in Humans Using the AOP Framework. Toxins (Basel) 2022; 14:279. [PMID: 35448888 PMCID: PMC9030459 DOI: 10.3390/toxins14040279] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/04/2022] [Accepted: 04/10/2022] [Indexed: 02/07/2023] Open
Abstract
Humans are chronically exposed to the mycotoxins deoxynivalenol (DON) and fumonisin B1 (FB1), as indicated by their widespread presence in foods and occasional exposure in the workplace. This exposure is confirmed by human biomonitoring (HBM) studies on (metabolites of) these mycotoxins in human matrices. We evaluated the exposure-health relationship of the mycotoxins in humans by reviewing the available literature. Since human studies did not allow the identification of unequivocal chronic health effects upon exposure to DON and FB1, the adverse outcome pathway (AOP) framework was used to structure additional mechanistic evidence from in vitro and animal studies on the identified adverse effects. In addition to a preliminary AOP for DON resulting in the adverse outcome (AO) 'reduced body weight gain', we developed a more elaborated AOP for FB1, from the molecular initiating event (MIE) 'inhibition of ceramide synthases' leading to the AO 'neural tube defects'. The mechanistic evidence from AOPs can be used to support the limited evidence from human studies, to focus FB1- and DON-related research in humans to identify related early biomarkers of effect. In order to establish additional human exposure-health relationships in the future, recommendations are given to maximize the information that can be obtained from HBM.
Collapse
Affiliation(s)
| | - Lola Bajard
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, 611 37 Brno, Czech Republic;
| | - Inger-Lise Steffensen
- Norwegian Institute of Public Health (NIPH), 0213 Oslo, Norway; (I.-L.S.); (A.L.B.); (H.A.A.M.D.)
| | - Anne Lise Brantsæter
- Norwegian Institute of Public Health (NIPH), 0213 Oslo, Norway; (I.-L.S.); (A.L.B.); (H.A.A.M.D.)
| | - Hubert A. A. M. Dirven
- Norwegian Institute of Public Health (NIPH), 0213 Oslo, Norway; (I.-L.S.); (A.L.B.); (H.A.A.M.D.)
| | - Jochem Louisse
- Wageningen Food Safety Research (WFSR), 6708 WB Wageningen, The Netherlands; (J.L.); (A.P.)
| | - Ad Peijnenburg
- Wageningen Food Safety Research (WFSR), 6708 WB Wageningen, The Netherlands; (J.L.); (A.P.)
| | - Sophie Ndaw
- Institut National de Recherche et de Sécurité (INRS), 54500 Vandoeuvre-Lés-Nancy, France;
| | - Alberto Mantovani
- Istituto Superiore di Sanità (ISS), 00161 Rome, Italy; (A.M.); (B.D.S.)
| | - Barbara De Santis
- Istituto Superiore di Sanità (ISS), 00161 Rome, Italy; (A.M.); (B.D.S.)
| | - Marcel J. B. Mengelers
- Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, The Netherlands;
| |
Collapse
|
7
|
Qu L, Wang L, Ji H, Fang Y, Lei P, Zhang X, Jin L, Sun D, Dong H. Toxic Mechanism and Biological Detoxification of Fumonisins. Toxins (Basel) 2022; 14:182. [PMID: 35324679 PMCID: PMC8954241 DOI: 10.3390/toxins14030182] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/14/2022] [Accepted: 02/26/2022] [Indexed: 11/16/2022] Open
Abstract
Food safety is related to the national economy and people's livelihood. Fumonisins are widely found in animal feed, feed raw materials, and human food. This can not only cause economic losses in animal husbandry but can also have carcinogenicity or teratogenicity and can be left in animal meat, eggs, and milk which may enter the human body and pose a serious threat to human health. Although there are many strategies to prevent fumonisins from entering the food chain, the traditional physical and chemical methods of mycotoxin removal have some disadvantages, such as an unstable effect, large nutrient loss, impact on the palatability of feed, and difficulty in mass production. As a safe, efficient, and environmentally friendly detoxification technology, biological detoxification attracts more and more attention from researchers and is gradually becoming an accepted technique. This work summarizes the toxic mechanism of fumonisins and highlights the advances of fumonisins in the detoxification of biological antioxidants, antagonistic microorganisms, and degradation mechanisms. Finally, the future challenges and focus of the biological control and degradation of fumonisins are discussed.
Collapse
Affiliation(s)
- Linkai Qu
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China;
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Lei Wang
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Hao Ji
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Yimeng Fang
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Pengyu Lei
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Xingxing Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China;
| | - Libo Jin
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Da Sun
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| | - Hao Dong
- College of Life Sciences, Jilin Agricultural University, Changchun 130118, China;
- Biomedical Collaborative Innovation Center of Zhejiang Province, Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China; (L.W.); (H.J.); (Y.F.); (P.L.); (L.J.)
| |
Collapse
|
8
|
Awuchi CG, Ondari EN, Nwozo S, Odongo GA, Eseoghene IJ, Twinomuhwezi H, Ogbonna CU, Upadhyay AK, Adeleye AO, Okpala COR. Mycotoxins’ Toxicological Mechanisms Involving Humans, Livestock and Their Associated Health Concerns: A Review. Toxins (Basel) 2022; 14:toxins14030167. [PMID: 35324664 PMCID: PMC8949390 DOI: 10.3390/toxins14030167] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/16/2022] [Accepted: 01/25/2022] [Indexed: 12/21/2022] Open
Abstract
Mycotoxins are well established toxic metabolic entities produced when fungi invade agricultural/farm produce, and this happens especially when the conditions are favourable. Exposure to mycotoxins can directly take place via the consumption of infected foods and feeds; humans can also be indirectly exposed from consuming animals fed with infected feeds. Among the hundreds of mycotoxins known to humans, around a handful have drawn the most concern because of their occurrence in food and severe effects on human health. The increasing public health importance of mycotoxins across human and livestock environments mandates the continued review of the relevant literature, especially with regard to understanding their toxicological mechanisms. In particular, our analysis of recently conducted reviews showed that the toxicological mechanisms of mycotoxins deserve additional attention to help provide enhanced understanding regarding this subject matter. For this reason, this current work reviewed the mycotoxins’ toxicological mechanisms involving humans, livestock, and their associated health concerns. In particular, we have deepened our understanding about how the mycotoxins’ toxicological mechanisms impact on the human cellular genome. Along with the significance of mycotoxin toxicities and their toxicological mechanisms, there are associated health concerns arising from exposures to these toxins, including DNA damage, kidney damage, DNA/RNA mutations, growth impairment in children, gene modifications, and immune impairment. More needs to be done to enhance the understanding regards the mechanisms underscoring the environmental implications of mycotoxins, which can be actualized via risk assessment studies into the conditions/factors facilitating mycotoxins’ toxicities.
Collapse
Affiliation(s)
- Chinaza Godseill Awuchi
- Department of Biochemistry, Kampala International University, Bushenyi P.O. Box 20000, Uganda; (E.N.O.); (S.N.); (G.A.O.); (I.J.E.)
- Correspondence: (C.G.A.); (C.O.R.O.)
| | - Erick Nyakundi Ondari
- Department of Biochemistry, Kampala International University, Bushenyi P.O. Box 20000, Uganda; (E.N.O.); (S.N.); (G.A.O.); (I.J.E.)
| | - Sarah Nwozo
- Department of Biochemistry, Kampala International University, Bushenyi P.O. Box 20000, Uganda; (E.N.O.); (S.N.); (G.A.O.); (I.J.E.)
| | - Grace Akinyi Odongo
- Department of Biochemistry, Kampala International University, Bushenyi P.O. Box 20000, Uganda; (E.N.O.); (S.N.); (G.A.O.); (I.J.E.)
| | - Ifie Josiah Eseoghene
- Department of Biochemistry, Kampala International University, Bushenyi P.O. Box 20000, Uganda; (E.N.O.); (S.N.); (G.A.O.); (I.J.E.)
| | | | - Chukwuka U. Ogbonna
- Department of Biochemistry, Federal University of Agriculture, P.M.B. 2240, Abeokuta 110124, Ogun State, Nigeria;
| | - Anjani K. Upadhyay
- Heredity Healthcare & Lifesciences, 206-KIIT TBI, Patia, Bhubaneswar 751024, Odisha, India;
| | - Ademiku O. Adeleye
- Faith Heroic Generation, No. 36 Temidire Street, Azure 340251, Ondo State, Nigeria;
| | - Charles Odilichukwu R. Okpala
- Department of Functional Foods Product Development, Wrocław University of Environmental and Life Sciences, 51-630 Wrocław, Poland
- Correspondence: (C.G.A.); (C.O.R.O.)
| |
Collapse
|
9
|
Silva GD, Brochers-Lacchini FC, Leopoldino AM. How do sphingolipids play a role in epigenetic mechanisms and gene expression? Epigenomics 2021; 14:219-222. [PMID: 34905958 DOI: 10.2217/epi-2021-0425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Gabriel da Silva
- Depto de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernanda Coeli Brochers-Lacchini
- Depto de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Andréia Machado Leopoldino
- Depto de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
10
|
Molina-Pintor I, Rojas-García A, Medina-Díaz I, Barrón-Vivanco B, Bernal-Hernández Y, Ortega-Cervantes L, Ramos A, Herrera-Moreno J, González-Arias C. An update on genotoxic and epigenetic studies of fumonisin B1. WORLD MYCOTOXIN J 2021. [DOI: 10.3920/wmj2021.2720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Fumonisins (FBs), a widespread group of mycotoxins produced by Fusarium spp., are natural contaminants in cereals and foodstuffs. Fumonisin B1 (FB1) is the most toxic and prevalent mycotoxin of this group, and it has been reported that FB1 accounts for 70-80% of FBs produced by the mycotoxigenic strains. The mode of action of FB1 depends on the structural similarity with sphinganine/sphingosine N-acyltransferase. This fact causes an accumulation of sphingoid bases and blocks the sphingolipid biosynthesis or the function of sphingolipids. Diverse toxic effects and diseases such as hepatocarcinogenicity, hepatotoxicity, nephrotoxicity, and cytotoxicity have been reported, and diseases like leukoencephalomalacia in horses and pulmonary oedema in horses and swine have been described. In humans, FBs have been associated with oesophageal cancer, liver cancer, neural tube defects, and infantile growth delay. However, despite the International Agency for Research on Cancer designated FB1 as a possibly carcinogenic to humans, its genotoxicity and epigenetic properties have not been clearly elucidated. This review aims to summarise the progress in research about the genotoxic and epigenetics effects of FB1.
Collapse
Affiliation(s)
- I.B. Molina-Pintor
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Los fresnos s/n. Tepic, Nayarit C.P. 63155, México
- Posgrado en Ciencias Biológico Agropecuarias, Unidad Académica de Agricultura, Km. 9 Carretera Tepic-Compostela, Xalisco, Nayarit, Mexico
| | - A.E. Rojas-García
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Los fresnos s/n. Tepic, Nayarit C.P. 63155, México
| | - I.M. Medina-Díaz
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Los fresnos s/n. Tepic, Nayarit C.P. 63155, México
| | - B.S. Barrón-Vivanco
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Los fresnos s/n. Tepic, Nayarit C.P. 63155, México
| | - Y.Y. Bernal-Hernández
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Los fresnos s/n. Tepic, Nayarit C.P. 63155, México
| | - L. Ortega-Cervantes
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Los fresnos s/n. Tepic, Nayarit C.P. 63155, México
| | - A.J. Ramos
- Food Technology Department, Lleida University, UTPV-XaRTA, Agrotecnio Center, Av. Rovira Roure 191, Lleida, 25198, Spain
| | - J.F. Herrera-Moreno
- Laboratory of Precision Environmental Health Sciences, Mailman School of Public Health, Columbia University, 630 west 168th Street, P&S Building Room 16-416, New York, NY, USA
| | - C.A. González-Arias
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Los fresnos s/n. Tepic, Nayarit C.P. 63155, México
| |
Collapse
|
11
|
Yu J, Yang M, Han J, Pang X. Fungal and mycotoxin occurrence, affecting factors, and prevention in herbal medicines: a review. TOXIN REV 2021. [DOI: 10.1080/15569543.2021.1925696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Jingsheng Yu
- Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing, China
| | - Meihua Yang
- Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianping Han
- Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing, China
| | - Xiaohui Pang
- Key Lab of Chinese Medicine Resources Conservation, State Administration of Traditional Chinese Medicine of the People’s Republic of China, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Engineering Research Center of Chinese Medicine Resource, Ministry of Education, Beijing, China
| |
Collapse
|
12
|
Zhu L, Yuhan J, Huang K, He X, Liang Z, Xu W. Multidimensional analysis of the epigenetic alterations in toxicities induced by mycotoxins. Food Chem Toxicol 2021; 153:112251. [PMID: 33961929 DOI: 10.1016/j.fct.2021.112251] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/30/2021] [Accepted: 04/30/2021] [Indexed: 02/07/2023]
Abstract
Mycotoxins contaminate all types of food and feed, threatening human and animal health through food chain accumulation, producing various toxic effects. Increasing attention is being focused on the molecular mechanism of mycotoxin-induced toxicity in all kinds of in vivo and in vitro models. Epigenetic alterations, including DNA methylation, non-coding RNAs (ncRNAs), and protein post-translational modifications (PTMs), were identified as being involved in various types of mycotoxin-induced toxicity. In this review, the emphasis was on summarizing the epigenetic alterations induced by mycotoxin, including aflatoxin B1 (AFB1), ochratoxin A (OTA), zearalenone (ZEA), fumonisin B1 (FB1), and deoxynivalenol (DON). This review summarized and analyzed the roles of DNA methylation, ncRNAs, and protein PTMs after mycotoxin exposure based on recently published papers. Moreover, the main research methods and their deficiencies were determined, while some remedial suggestions are proposed. In summary, this review helps to understand better the epigenetic alterations induced by the non-genotoxic effects of mycotoxin.
Collapse
Affiliation(s)
- Liye Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Jieyu Yuhan
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Kunlun Huang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Xiaoyun He
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Zhihong Liang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Wentao Xu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
13
|
Arumugam T, Ghazi T, Chuturgoon AA. Molecular and epigenetic modes of Fumonisin B 1 mediated toxicity and carcinogenesis and detoxification strategies. Crit Rev Toxicol 2021; 51:76-94. [PMID: 33605189 DOI: 10.1080/10408444.2021.1881040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fumonisin B1 (FB1) is a natural contaminant of agricultural commodities that has displayed a myriad of toxicities in animals. Moreover, it is known to be a hepatorenal carcinogen in rodents and may be associated with oesophageal and hepatocellular carcinomas in humans. The most well elucidated mode of FB1-mediated toxicity is its disruption of sphingolipid metabolism; however, enhanced oxidative stress, endoplasmic reticulum stress, autophagy, and alterations in immune response may also play a role in its toxicity and carcinogenicity. Alterations to the host epigenome may impact on the toxic and carcinogenic response to FB1. Seeing that the contamination of FB1 in food poses a considerable risk to human and animal health, a great deal of research has focused on new methods to prevent and attenuate FB1-induced toxic consequences. The focus of the present review is on the molecular and epigenetic interactions of FB1 as well as recent research involving FB1 detoxification.
Collapse
Affiliation(s)
- Thilona Arumugam
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Terisha Ghazi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
14
|
Wangia-Dixon RN, Nishimwe K. Molecular toxicology and carcinogenesis of fumonisins: a review. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:44-67. [PMID: 33554724 DOI: 10.1080/26896583.2020.1867449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Fumonisins, discovered in 1988 are a group of naturally occurring toxins produced by fusarium pathogenic fungi. Besides their presence in animal feeds, contamination of human foods such as corn, millet, oats, rye, barley, wheat and their products are widespread. Exposure to fumonisins results in species and organ specific toxicities including neurological disorders among equids, pulmonary edema in swine, esophageal cancer in humans and both kidney and liver related toxicities in rodents. This review seeks to consolidate groundbreaking research on the science of fumonisins toxicity, highlight recent progress on fumonisins research, and provide an overview of plausible mechanistic biomarkers for fumonisins exposure assessment.
Collapse
Affiliation(s)
- Ruth Nabwire Wangia-Dixon
- Department of Environmental Health Science, University of Georgia, Athens, Georgia, USA
- KAVI Institute of Clinical Research, University of Nairobi, Nairobi, Kenya
| | - Kizito Nishimwe
- Department of Food Science and Human Nutrition, Iowa State University, Ames, Iowa, USA
- School of Agriculture and Food Science, University of Rwanda, Kigali, Rwanda
| |
Collapse
|
15
|
Taruscio D, Mantovani A. Multifactorial Rare Diseases: Can Uncertainty Analysis Bring Added Value to the Search for Risk Factors and Etiopathogenesis? ACTA ACUST UNITED AC 2021; 57:medicina57020119. [PMID: 33525390 PMCID: PMC7911455 DOI: 10.3390/medicina57020119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/23/2021] [Accepted: 01/24/2021] [Indexed: 11/16/2022]
Abstract
Uncertainty analysis is the process of identifying limitations in knowledge and evaluating their implications for scientific conclusions. Uncertainty analysis is a stable component of risk assessment and is increasingly used in decision making on complex health issues. Uncertainties should be identified in a structured way and prioritized according to their likely impact on the outcome of scientific conclusions. Uncertainty is inherent to the rare diseases (RD) area, where research and healthcare have to cope with knowledge gaps due to the rarity of the conditions; yet a systematic approach toward uncertainties is not usually undertaken. The uncertainty issue is particularly relevant to multifactorial RD, whose etiopathogenesis involves environmental factors and genetic predisposition. Three case studies are presented: the newly recognized acute multisystem inflammatory syndrome in children and adolescents associated with SARS-CoV-2 infection; the assessment of risk factors for neural tube defects; and the genotype-phenotype correlation in familial Mediterranean fever. Each case study proposes the initial identification of the main epistemic and sampling uncertainties and their impacts. Uncertainty analysis in RD may present aspects similar to those encountered when conducting risk assessment in data-poor scenarios; therefore, approaches such as expert knowledge elicitation may be considered. The RD community has a main strength in managing uncertainty, as it proactively develops stakeholder involvement, data sharing and open science. The open science approaches can be profitably integrated by structured uncertainty analysis, especially when dealing with multifactorial RD involving environmental and genetic risk factors.
Collapse
Affiliation(s)
- Domenica Taruscio
- National Center for Rare Diseases, Italian National Institute of Health (ISS), 00161 Roma, Italy
- Correspondence:
| | - Alberto Mantovani
- Department on Food Safety, Nutrition and Veterinary Public Health, Italian National Institute of Health (ISS), 00161 Roma, Italy;
| |
Collapse
|
16
|
Arumugam T, Ghazi T, Chuturgoon A. Fumonisin B 1 Epigenetically Regulates PTEN Expression and Modulates DNA Damage Checkpoint Regulation in HepG2 Liver Cells. Toxins (Basel) 2020; 12:toxins12100625. [PMID: 33007920 PMCID: PMC7601513 DOI: 10.3390/toxins12100625] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/19/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Fumonisin B1 (FB1), a Fusarium-produced mycotoxin, is found in various foods and feeds. It is a well-known liver carcinogen in experimental animals; however, its role in genotoxicity is controversial. The current study investigated FB1-triggered changes in the epigenetic regulation of PTEN and determined its effect on DNA damage checkpoint regulation in human liver hepatoma G2 (HepG2) cells. Following treatment with FB1 (IC50: 200 µM; 24 h), the expression of miR-30c, KDM5B, PTEN, H3K4me3, PI3K, AKT, p-ser473-AKT, CHK1, and p-ser280-CHK1 was measured using qPCR and/or Western blot. H3K4me3 enrichment at the PTEN promoter region was assayed via a ChIP assay and DNA damage was determined using an ELISA. FB1 induced oxidative DNA damage. Total KDM5B expression was reduced, which subsequently increased the total H3K4me3 and the enrichment of H3K4me3 at PTEN promoters. Increased H3K4me3 induced an increase in PTEN transcript levels. However, miR-30c inhibited PTEN translation. Thus, PI3K/AKT signaling was activated, inhibiting CHK1 activity via phosphorylation of its serine 280 residue preventing the repair of damaged DNA. In conclusion, FB1 epigenetically modulates the PTEN/PI3K/AKT signaling cascade, preventing DNA damage checkpoint regulation, and induces significant DNA damage.
Collapse
|
17
|
Claeys L, Romano C, De Ruyck K, Wilson H, Fervers B, Korenjak M, Zavadil J, Gunter MJ, De Saeger S, De Boevre M, Huybrechts I. Mycotoxin exposure and human cancer risk: A systematic review of epidemiological studies. Compr Rev Food Sci Food Saf 2020; 19:1449-1464. [PMID: 33337079 DOI: 10.1111/1541-4337.12567] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 03/13/2020] [Accepted: 04/03/2020] [Indexed: 01/01/2023]
Abstract
In recent years, there has been an increasing interest in investigating the carcinogenicity of mycotoxins in humans. This systematic review aims to provide an overview of data linking exposure to different mycotoxins with human cancer risk. Publications (2019 and earlier) of case-control or longitudinal cohort studies were identified in PubMed and EMBASE. These articles were then screened by independent reviewers and their quality was assessed according to the Newcastle-Ottawa scale. Animal, cross-sectional, and molecular studies satisfied criteria for exclusion. In total, 14 articles were included: 13 case-control studies and 1 longitudinal cohort study. Included articles focused on associations of mycotoxin exposure with primary liver, breast, and cervical cancer. Overall, a positive association between the consumption of aflatoxin-contaminated foods and primary liver cancer risk was verified. Two case-control studies in Africa investigated the relationship between zearalenone and its metabolites and breast cancer risk, though conflicting results were reported. Two case-control studies investigated the association between hepatocellular carcinoma and fumonisin B1 exposure, but no significant associations were observed. This systematic review incorporates several clear observations of dose-dependent associations between aflatoxins and liver cancer risk, in keeping with IARC Monograph conclusions. Only few human epidemiological studies investigated the associations between mycotoxin exposures and cancer risk. To close this gap, more in-depth research is needed to unravel evidence for other common mycotoxins, such as deoxynivalenol and ochratoxin A. The link between mycotoxin exposures and cancer risk has mainly been established in experimental studies, and needs to be confirmed in human epidemiological studies to support the evidence-based public health strategies.
Collapse
Affiliation(s)
- Liesel Claeys
- Centre of Excellence in Mycotoxicology and Public Health, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Nutritional Epidemiology Group, International Agency for Research on Cancer, Lyon, France
- Molecular Mechanisms and Biomarkers Group, International Agency for Research on Cancer, Lyon, France
- CRIG, Cancer Research Institute Ghent, Ghent, Belgium
| | - Chiara Romano
- Nutritional Epidemiology Group, International Agency for Research on Cancer, Lyon, France
| | - Karl De Ruyck
- Centre of Excellence in Mycotoxicology and Public Health, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- CRIG, Cancer Research Institute Ghent, Ghent, Belgium
| | - Hayley Wilson
- Nutritional Epidemiology Group, International Agency for Research on Cancer, Lyon, France
| | - Beatrice Fervers
- Department of Cancer and Environment, Centre Léon Bérnard, UA08 INSERM Radiation, Defense, Health and Environment, Lyon, France
| | - Michael Korenjak
- Molecular Mechanisms and Biomarkers Group, International Agency for Research on Cancer, Lyon, France
| | - Jiri Zavadil
- Molecular Mechanisms and Biomarkers Group, International Agency for Research on Cancer, Lyon, France
| | - Marc J Gunter
- Nutritional Epidemiology Group, International Agency for Research on Cancer, Lyon, France
| | - Sarah De Saeger
- Centre of Excellence in Mycotoxicology and Public Health, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- CRIG, Cancer Research Institute Ghent, Ghent, Belgium
| | - Marthe De Boevre
- Centre of Excellence in Mycotoxicology and Public Health, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- CRIG, Cancer Research Institute Ghent, Ghent, Belgium
| | - Inge Huybrechts
- Nutritional Epidemiology Group, International Agency for Research on Cancer, Lyon, France
- CRIG, Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
18
|
Carreira AC, Santos TC, Lone MA, Zupančič E, Lloyd-Evans E, de Almeida RFM, Hornemann T, Silva LC. Mammalian sphingoid bases: Biophysical, physiological and pathological properties. Prog Lipid Res 2019:100995. [PMID: 31445071 DOI: 10.1016/j.plipres.2019.100995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022]
Abstract
Sphingoid bases encompass a group of long chain amino alcohols which form the essential structure of sphingolipids. Over the last years, these amphiphilic molecules were moving more and more into the focus of biomedical research due to their role as bioactive molecules. In fact, free sphingoid bases interact with specific receptors and target molecules and have been associated with numerous biological and physiological processes. In addition, they can modulate the biophysical properties of biological membranes. Several human diseases are related to pathological changes in the structure and metabolism of sphingoid bases. Yet, the mechanisms underlying their biological and pathophysiological actions remain elusive. Within this review, we aimed to summarize the current knowledge on the biochemical and biophysical properties of the most common sphingoid bases and to discuss their importance in health and disease.
Collapse
Affiliation(s)
- A C Carreira
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal; Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - T C Santos
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN) and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - M A Lone
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - E Zupančič
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - E Lloyd-Evans
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - R F M de Almeida
- Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal
| | - T Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - L C Silva
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN) and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
19
|
Riley RT, Merrill AH. Ceramide synthase inhibition by fumonisins: a perfect storm of perturbed sphingolipid metabolism, signaling, and disease. J Lipid Res 2019; 60:1183-1189. [PMID: 31048407 PMCID: PMC6602133 DOI: 10.1194/jlr.s093815] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/25/2019] [Indexed: 01/18/2023] Open
Abstract
Fumonisins are mycotoxins that cause diseases of plants and, when consumed by animals, can damage liver, kidney, lung, brain, and other organs, alter immune function, and cause developmental defects and cancer. They structurally resemble sphingolipids (SLs), and studies nearly 30 years ago discovered that the most prevalent fumonisin [fumonisin B1 (FB1)] potently inhibits ceramide synthases (CerSs), enzymes that use fatty acyl-CoAs to N-acylate sphinganine (Sa), sphingosine (So), and other sphingoid bases. CerS inhibition by FB1 triggers a "perfect storm" of perturbations in structural and signaling SLs that include: reduced formation of dihydroceramides, ceramides, and complex SLs; elevated Sa and So and their 1-phosphates, novel 1-deoxy-sphingoid bases; and alteration of additional lipid metabolites from interrelated pathways. Moreover, because the initial enzyme of sphingoid base biosynthesis remains active (sometimes with increased activity), the impact is multiplied by the continued production of damaging metabolites. Evidence from many studies, including characterization of knockout mice for specific CerSs and analyses of human blood (which found that FB1 intake is associated with elevated Sa 1-phosphate), has consistently pointed to CerS as the proximate target of FB1 It is also apparent that the changes in multiple bioactive lipids and related biologic processes account for the ensuing spectrum of animal and plant disease. Thus, the diseases caused by fumonisins can be categorized as "sphingolipidoses" (in these cases, due to defective SL biosynthesis), and the lessons learned about the consequences of CerS inhibition should be borne in mind when contemplating other naturally occurring and synthetic compounds (and genetic manipulations) that interfere with SL metabolism.
Collapse
Affiliation(s)
- Ronald T Riley
- College of Public Health, Department of Environmental Health Science, University of Georgia, Athens, GA 30602
| | - Alfred H Merrill
- School of Biological Sciences and the Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|
20
|
Carreira AC, Santos TC, Lone MA, Zupančič E, Lloyd-Evans E, de Almeida RFM, Hornemann T, Silva LC. Mammalian sphingoid bases: Biophysical, physiological and pathological properties. Prog Lipid Res 2019; 75:100988. [PMID: 31132366 DOI: 10.1016/j.plipres.2019.100988] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/11/2022]
Abstract
Sphingoid bases encompass a group of long chain amino alcohols which form the essential structure of sphingolipids. Over the last years, these amphiphilic molecules were moving more and more into the focus of biomedical research due to their role as bioactive molecules. In fact, free sphingoid bases interact with specific receptors and target molecules, and have been associated with numerous biological and physiological processes. In addition, they can modulate the biophysical properties of biological membranes. Several human diseases are related to pathological changes in the structure and metabolism of sphingoid bases. Yet, the mechanisms underlying their biological and pathophysiological actions remain elusive. Within this review, we aimed to summarize the current knowledge on the biochemical and biophysical properties of the most common sphingoid bases and to discuss their importance in health and disease.
Collapse
Affiliation(s)
- A C Carreira
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, Lisboa 1749-016, Portugal; Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, UK
| | - T C Santos
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN), IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - M A Lone
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - E Zupančič
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal
| | - E Lloyd-Evans
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, UK
| | - R F M de Almeida
- Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, Lisboa 1749-016, Portugal
| | - T Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - L C Silva
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN), IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
21
|
Use of Primary Mouse Embryonic Fibroblasts in Developmental Toxicity Assessments. Methods Mol Biol 2019. [PMID: 31069665 DOI: 10.1007/978-1-4939-9182-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Mouse embryonic fibroblasts (MEFs) are commonly collected as a means to maintain the culture and growth of embryonic stem cells (ESCs). However, their utility can extend well beyond their use exclusively in ESC culture. With collection from various transgenic mouse models, use of MEFs may serve as a more simplistic means to reconstitute in vivo/in utero toxicological assessments in an in vitro format for evaluation of function of specific proteins during toxic insults. The ease of collection, rapid growth kinetics, and large-scale expansion to perform multiple, high-throughput experiments are just some of the advantages of MEF use. Here, we describe procedures for successful MEF isolation and culture. As an example of MEF utility, we use MEFs collected form wild-type (WT) and Nrf2 knockout mice. After collection, MEFs were pretreated with the Nrf2 activator, dithiol-3-thione (D3T; 10 μM) for 12 h, and then treated with either hydrogen peroxide (0-2000 μM) or mercury (0-100 μM) for another 24 h. Viability was measured via MTT assay after 24 h of treatment.
Collapse
|
22
|
Magaye RR, Savira F, Hua Y, Kelly DJ, Reid C, Flynn B, Liew D, Wang BH. The role of dihydrosphingolipids in disease. Cell Mol Life Sci 2019; 76:1107-1134. [PMID: 30523364 PMCID: PMC11105797 DOI: 10.1007/s00018-018-2984-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/06/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022]
Abstract
Dihydrosphingolipids refer to sphingolipids early in the biosynthetic pathway that do not contain a C4-trans-double bond in the sphingoid backbone: 3-ketosphinganine (3-ketoSph), dihydrosphingosine (dhSph), dihydrosphingosine-1-phosphate (dhS1P) and dihydroceramide (dhCer). Recent advances in research related to sphingolipid biochemistry have shed light on the importance of sphingolipids in terms of cellular signalling in health and disease. However, dihydrosphingolipids have received less attention and research is lacking especially in terms of their molecular mechanisms of action. This is despite studies implicating them in the pathophysiology of disease, for example dhCer in predicting type 2 diabetes in obese individuals, dhS1P in cardiovascular diseases and dhSph in hepato-renal toxicity. This review gives a comprehensive summary of research in the last 10-15 years on the dihydrosphingolipids, 3-ketoSph, dhSph, dhS1P and dhCer, and their relevant roles in different diseases. It also highlights gaps in research that could be of future interest.
Collapse
Affiliation(s)
- Ruth R Magaye
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Feby Savira
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Yue Hua
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Darren J Kelly
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| | - Christopher Reid
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Bernard Flynn
- Australian Translational Medicinal Chemistry Facility, Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Danny Liew
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Bing H Wang
- Monash Centre of Cardiovascular Research and Education in Therapeutics, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia.
| |
Collapse
|
23
|
Chen C, Riley RT, Wu F. Dietary Fumonisin and Growth Impairment in Children and Animals: A Review. Compr Rev Food Sci Food Saf 2018; 17:1448-1464. [DOI: 10.1111/1541-4337.12392] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/24/2018] [Accepted: 08/01/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Chen Chen
- Dept. of Food Science and Human Nutrition; Michigan State Univ.; East Lansing MI 48824 U.S.A
- Inst. of Quality Standards and Testing Technology for Agro-Products; Chinese Academy of Agricultural Sciences; Beijing 100081 China
| | - Ronald T. Riley
- Dept. of Environmental Health Science; Univ. of Georgia; Athens GA 30602 U.S.A
| | - Felicia Wu
- Dept. of Food Science and Human Nutrition; Michigan State Univ.; East Lansing MI 48824 U.S.A
| |
Collapse
|
24
|
Fu P, Ebenezer DL, Ha AW, Suryadevara V, Harijith A, Natarajan V. Nuclear lipid mediators: Role of nuclear sphingolipids and sphingosine-1-phosphate signaling in epigenetic regulation of inflammation and gene expression. J Cell Biochem 2018; 119:6337-6353. [PMID: 29377310 DOI: 10.1002/jcb.26707] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/23/2018] [Indexed: 12/18/2022]
Abstract
Phospholipids, sphingolipids, and cholesterol are integral components of eukaryotic cell organelles, including the nucleus. Recent evidence shows characteristic features of nuclear lipid composition and signaling, which are distinct from that of the cytoplasm and plasma membrane. While the nuclear phosphoinositol lipid signaling in cell cycle regulation and differentiation has been well described, there is a paucity on the role of nuclear sphingolipids and sphingolipid signaling in different physiological and pathophysiological human conditions. In this prospective, we describe the role of sphingolipids and specifically focus on the sphingoid bases, such as sphingosine, ceramide, and sphingosine-1-phosphate (S1P) generation and catabolism in nuclear signaling and function. Particularly, S1P generated in the nucleus by phosphorylation of SPHK2 modulates HDAC activity either by direct binding or through activation of nuclear reactive oxygen species and regulates cell cycle and pro-inflammatory gene expression. Potential implication of association of SPHK2 with the co-repressor complexes and generation of S1P in the nucleus on chromatin remodeling under normal and pathological conditions is discussed. A better understanding of sphingolipid signaling in the nucleus will facilitate the design and development of new and novel therapeutic approaches to modulate expression of pro-inflammatory and cell cycle dependent genes in human pathologies such as cancer, bacterial lung infection, neurodegeneration, and cystic fibrosis.
Collapse
Affiliation(s)
- Panfeng Fu
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - David L Ebenezer
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois
| | - Alison W Ha
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois
| | | | - Anantha Harijith
- Department of Pediatrics, University of Illinois, Chicago, Illinois
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois.,Department of Medicine, University of Illinois, Chicago, Illinois
| |
Collapse
|
25
|
Knutsen HK, Barregård L, Bignami M, Brüschweiler B, Ceccatelli S, Cottrill B, Dinovi M, Edler L, Grasl-Kraupp B, Hogstrand C, Hoogenboom LR, Nebbia CS, Petersen A, Rose M, Roudot AC, Schwerdtle T, Vleminckx C, Vollmer G, Wallace H, Dall'Asta C, Gutleb AC, Humpf HU, Galli C, Metzler M, Oswald IP, Parent-Massin D, Binaglia M, Steinkellner H, Alexander J. Appropriateness to set a group health-based guidance value for fumonisins and their modified forms. EFSA J 2018; 16:e05172. [PMID: 32625807 PMCID: PMC7009576 DOI: 10.2903/j.efsa.2018.5172] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The EFSA Panel on Contaminants in the Food Chain (CONTAM) established a tolerable daily intake (TDI) for fumonisin B1 (FB 1) of 1.0 μg/kg body weight (bw) per day based on increased incidence of megalocytic hepatocytes found in a chronic study with mice. The CONTAM Panel considered the limited data available on toxicity and mode of action and structural similarities of FB 2-6 and found it appropriate to include FB 2, FB 3 and FB 4 in a group TDI with FB 1. Modified forms of FBs are phase I and phase II metabolites formed in fungi, infested plants or farm animals. Modified forms also arise from food or feed processing, and include covalent adducts with matrix constituents. Non-covalently bound forms are not considered as modified forms. Modified forms of FBs identified are hydrolysed FB 1-4 (HFB 1-4), partially hydrolysed FB 1-2 (pHFB 1-2), N-(carboxymethyl)-FB 1-3 (NCM-FB 1-3), N-(1-deoxy-d-fructos-1-yl)-FB 1 (NDF-FB 1), O-fatty acyl FB 1, N-fatty acyl FB 1 and N-palmitoyl-HFB 1. HFB 1, pHFB 1, NCM-FB 1 and NDF-FB 1 show a similar toxicological profile but are less potent than FB 1. Although in vitro data shows that N-fatty acyl FBs are more toxic in vitro than FB 1, no in vivo data were available for N-fatty acyl FBs and O-fatty acyl FBs. The CONTAM Panel concluded that it was not appropriate to include modified FBs in the group TDI for FB 1-4. The uncertainty associated with the present assessment is high, but could be reduced provided more data are made available on occurrence, toxicokinetics and toxicity of FB 2-6 and modified forms of FB 1-4.
Collapse
|
26
|
Stereoselective preparation of quaternary 2-vinyl sphingosines and ceramides and their effect on basal sphingolipid metabolism. Chem Phys Lipids 2017; 205:34-41. [PMID: 28445710 DOI: 10.1016/j.chemphyslip.2017.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/16/2017] [Accepted: 04/20/2017] [Indexed: 11/20/2022]
Abstract
The dicyclohexylborane-mediated addition of allene 1 to (E)-2-tridecenal affords a quaternary protected 2-amino-2-vinyl-1,3-diol in good yield as a single diastereomer. This compound is readily transformed into the four stereoisomers of the quaternary (E)-2-vinyl analogs of sphingosine. The metabolic fate and the effect of these compounds on the basal sphingolipid metabolism in human A549 lung adenocarcinoma cells has been studied, together with the ceramide analog of the most relevant vinylsphingosine derivative.
Collapse
|
27
|
Qian G, Tang L, Lin S, Xue KS, Mitchell NJ, Su J, Gelderblom WC, Riley RT, Phillips TD, Wang JS. Sequential dietary exposure to aflatoxin B1 and fumonisin B1 in F344 rats increases liver preneoplastic changes indicative of a synergistic interaction. Food Chem Toxicol 2016; 95:188-95. [PMID: 27430420 DOI: 10.1016/j.fct.2016.07.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 10/21/2022]
Abstract
Dietary co-exposure to aflatoxin B1 (AFB1) and fumonisin B1 (FB1) and their interaction on hepatocellular carcinogenesis is of particular concern in toxicology and public health. In this study we evaluated the liver preneoplastic effects of single and sequential dietary exposure to AFB1 and FB1 in the F344 rat carcinogenesis model. Serum biochemical alterations, liver histopathological changes, and the formation of liver glutathione S transferase positive (GST-P+) foci were the major outcome parameters examined. Compared to the AFB1-only treatment, the FB1-only treatment induced less dysplasia, and more apoptosis and mitoses. Sequential AFB1 and FB1 treatment lead to increased numbers of dysplasia, apoptosis and foci of altered hepatocytes, as compared to either mycotoxin treatment alone. More importantly, sequential exposure to AFB1 and FB1 synergistically increased the numbers of liver GTP-P+ foci by approximately 7.3-and 12.9-fold and increased the mean sizes of GST-P+ foci by 6- and 7.5-fold, respectively, as compared to AFB1- or FB1-only treatment groups. In addition, liver ALT and AST levels were significantly increased after sequential treatment as compared to single treatment groups. The results demonstrate the interactive effect of dietary AFB1 and FB1 in inducing liver GST-P+ foci formation and provide information to model future intervention studies.
Collapse
Affiliation(s)
- Guoqing Qian
- Department of Environmental Health Science, University of Georgia, Athens, GA, USA
| | - Lili Tang
- Department of Environmental Health Science, University of Georgia, Athens, GA, USA
| | - Shuhan Lin
- Department of Environmental Health Science, University of Georgia, Athens, GA, USA
| | - Kathy S Xue
- Department of Environmental Health Science, University of Georgia, Athens, GA, USA
| | - Nicole J Mitchell
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Jianjia Su
- Department of Environmental Health Science, University of Georgia, Athens, GA, USA
| | - Wentzel C Gelderblom
- Institute of Biomedical and Microbial Biotechnology, Cape Peninsula University of Technology, PO Box 1906, Bellville, 7535, South Africa
| | - Ronald T Riley
- Department of Environmental Health Science, University of Georgia, Athens, GA, USA; USDA-ARS, Toxicology and Mycotoxin Research Unit, R.B. Russell Research Center, National Poultry Disease Research Center, Athens, GA, USA
| | - Timothy D Phillips
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Jia-Sheng Wang
- Department of Environmental Health Science, University of Georgia, Athens, GA, USA.
| |
Collapse
|