Mold, Mycotoxins and a Dysregulated Immune System: A Combination of Concern?
Abstract
:1. Introduction
2. Mold-Induced Immunological Effects
2.1. Toxic Mold Syndrome
2.2. Mold-Induced Hypersensitivity
2.3. Immune Modulatory Potential of Mycotoxins
3. Mold-Induced Exacerbation of Immune System Disorders
3.1. Mold Exposure and Increased Asthma Severity
3.2. Mycotoxin Exposure and Its Association with Autoimmune Disorders
3.3. Mycotoxin Exposure Is Associated with an Onset of Inflammatory Bowel Disease
3.4. Mycotoxin Exposure and Human Immunodeficiency Virus (HIV) Infection
Mold | Mold Component | Species | Disease/ Model | Measured Surrogate Marker | Outcome | Source |
---|---|---|---|---|---|---|
Increased Asthma severity | ||||||
A. fumigatus | Spores | Human | Asthma | Total serum IgE ↑ Blood eosinophils (cells/µL) ↑ FEV1/FVC (%) ↓ | Asthma severity increased | [50] |
Penicillium spp. | Spores | Human | Asthma | FEV1/FVC (%) ↓ | Asthma severity increased | [50] |
n.a. | GTX Patulin | Mouse | Allergic asthma/ OVA-model | Airway inflammation ↑ OVA-IgE ↑ BAL eosinophils ↑ | Asthma severity increased | [54] |
Involved in autoimmune response | ||||||
Mold water damage | n.a. | Human | n.a. | IgG neuronal antibodies against microtubule-associated protein-2, myelin basic protein, tau, glial fibrillary acidic protein, tubulin, and S-100B | No clincial outcome | [57] |
n.a. | GTX | Mouse | EAE | Neuroinflammation ↑ Demyelination ↑ | Aggravation of autoimmune encephalomyelitis | [58] |
A. fumigatus | A. fumigatus antigens (I and VIII) | Human | RA | IgG ↑ IgA ↑ | Stronger sensitization than control subjects | [62] |
n.a. | OTA DON | Mouse | RA DBA1 model | IgG1 ↑ IG2a ↑ Pro-inflammatory cytokines ↑ | Clinical severity score ↑ | [63] |
Trigger of IBD | ||||||
S. chartarum | Trichothecene group | Human | CU/CSF HLA-DR/DQ susceptible | Mycotoxin test in urine-positive | Pancreatitis improved after withdrawn | [65] |
n.a. | DON | Rat | CU DSS model | Morphological damage in colon ↑ Colonic inflammation ↑ | Exacerbation of onset and symptoms of DSS-induced colitis | [67] |
Worsening of HIV condition | ||||||
Aspergillus spp. | AFB1 | Human | HIV-positive adults | Plasma aflatoxin B1 ↑ HIV-1 RNA ↑ | Higher viral loads in HIV-positive humans with higher AF-ALB | [69,70] |
4. Mode of Action
4.1. Effects of Mold on the Immune System
4.2. Mycotoxins Are Systemically Bioavailable
4.3. Mycotoxins Compromise Barrier Functions
4.4. Mycotoxins Can Influence the Gastrointestinal Microbiota
4.5. Mycotoxins Have the Potential to Induce and Exacerbate Inflammation
4.6. Mycotoxins Interfere with T-Cell Differentiation
4.7. Mycotoxins and Their Potential Exacerbating Effect Regarding HIV Infection and Diseases Progression
5. Conclusions and Future Perspectives
- (1)
- Study objectives should include investigation of the adjuvant activity of mycotoxins of the addressed immune system disorders. For better understanding of the underlying pathological mechanism, future research should investigate the key modes of action including the alteration of barrier functions, exacerbation of inflammation and T-cell differentiation. Moreover, the measurement of relevant disease parameters like the influence on gastrointestinal microbiota in IBD, or axonal demyelination in MS is strongly recommended.
- (2)
- Using appropriate disease models for a valuable translation to human exposure and clinical research is suggested. Therefore, it is necessary to determine state of the art disease models and use disease-specific biomarkers as readout. Real-world human mycotoxin exposure scenarios should be mimicked in model organisms to be able to make estimations about critical threshold concentration for specific vulnerable populations.
- (3)
- Investigations to develop prevention or treatment strategies to face the exacerbation of pre-existing immune system disorders due to mycotoxin contamination should be addressed as well. It should be questioned if the harmful effects are reversible after elimination of the mycotoxin or by interfering with the mycotoxin modes of action. E.g., the development of sensitive and specific detection assays for multiplex mycotoxin detection could be established for comprehensive diagnostics.
- (4)
- Next to mycotoxin exposure, also invasion mechanisms of mold might be of interest depending on the disease model. Under this scope, antifungal treatment options should be investigated as well.
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Robbins, C.A.; Swenson, L.J.; Nealley, M.L.; Kelman, B.J.; Gots, R.E. Health Effects of Mycotoxins in Indoor Air: A Critical Review. Appl. Occup. Environ. Hyg. 2010, 15, 773–784. [Google Scholar] [CrossRef]
- Barlow, G.; Irving, W.L.; Moss, P.J. Infectious diseases. In Kumar and Clark’s Clinical Medicine, 10th ed.; Elsevier: Amsterdam, The Netherlands, 2020. [Google Scholar]
- Benedict, K.; Chiller, T.M.; Mody, R.K. Invasive Fungal Infections Acquired from Contaminated Food or Nutritional Supplements: A Review of the Literature. Foodborne Pathog. Dis. 2016, 13, 343–349. [Google Scholar] [CrossRef]
- Kohler, J.R.; Hube, B.; Puccia, R.; Casadevall, A.; Perfect, J.R. Fungi that Infect Humans. Microbiol. Spectr. 2017, 5. [Google Scholar] [CrossRef] [PubMed]
- Jung, B.; Park, J.; Kim, N.; Li, T.; Kim, S.; Bartley, L.E.; Kim, J.; Kim, I.; Kang, Y.; Yun, K.; et al. Cooperative interactions between seed-borne bacterial and air-borne fungal pathogens on rice. Nat. Commun. 2018, 9, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Venkatesh, N.; Keller, N.P. Mycotoxins in Conversation with Bacteria and Fungi. Front. Microbiol. 2019, 10, 403. [Google Scholar] [CrossRef]
- Health, N.C.f.E. Basic Facts about Mold and Dampness. Available online: https://www.cdc.gov/mold/faqs.htm (accessed on 30 August 2021).
- Omotayo, O.P.; Omotayo, A.O.; Mwanza, M.; Babalola, O.O. Prevalence of Mycotoxins and Their Consequences on Human Health. Toxicol. Res. 2019, 35, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- D’Mello, J.P. Handbook of Plant and Fungal Toxicants; CRC Press: Boca Raton, FL, USA, 1997. [Google Scholar]
- Ciegler, A.; Bennett, J.W. Mycotoxins and Mycotoxicoses. BioScience 1980, 30, 512–515. [Google Scholar] [CrossRef]
- Gracia-Lor, E.; Zuccato, E.; Hernandez, F.; Castiglioni, S. Wastewater-based epidemiology for tracking human exposure to mycotoxins. J. Hazard. Mater. 2020, 382, 121108. [Google Scholar] [CrossRef]
- Vin, K.; Riviere, G.; Leconte, S.; Cravedi, J.P.; Fremy, J.M.; Oswald, I.P.; Roudot, A.C.; Vasseur, P.; Jean, J.; Hulin, M.; et al. Dietary exposure to mycotoxins in the French infant total diet study. Food Chem. Toxicol. 2020, 140, 111301. [Google Scholar] [CrossRef]
- Arce-Lopez, B.; Lizarraga, E.; Lopez de Mesa, R.; Gonzalez-Penas, E. Assessment of Exposure to Mycotoxins in Spanish Children through the Analysis of Their Levels in Plasma Samples. Toxins 2021, 13, 150. [Google Scholar] [CrossRef]
- Franco, L.T.; Ismail, A.; Amjad, A.; Oliveira, C.A.F.D. Occurrence of toxigenic fungi and mycotoxins in workplaces and human biomonitoring of mycotoxins in exposed workers: A systematic review. Toxin Rev. 2020, 1–16. [Google Scholar] [CrossRef]
- Sadhasivam, S.; Britzi, M.; Zakin, V.; Kostyukovsky, M.; Trostanetsky, A.; Quinn, E.; Sionov, E. Rapid Detection and Identification of Mycotoxigenic Fungi and Mycotoxins in Stored Wheat Grain. Toxins 2017, 9, 302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- WHO. Mycotoxins. Available online: https://www.who.int/news-room/fact-sheets/detail/mycotoxins (accessed on 9 August 2021).
- Lessard, M.; Savard, C.; Deschene, K.; Lauzon, K.; Pinilla, V.A.; Gagnon, C.A.; Lapointe, J.; Guay, F.; Chorfi, Y. Impact of deoxynivalenol (DON) contaminated feed on intestinal integrity and immune response in swine. Food Chem. Toxicol. 2015, 2015, 7–16. [Google Scholar] [CrossRef] [PubMed]
- Gonkowski, S.; Gajecka, M.; Makowska, K. Mycotoxins and the Enteric Nervous System. Toxins 2020, 12, 461. [Google Scholar] [CrossRef]
- Empting, L.D. Neurologic and neuropsychiatric syndrome features of mold and mycotoxin exposure. Toxicol. Ind. Health 2009, 25, 577–581. [Google Scholar] [CrossRef] [PubMed]
- French, P.W.; Ludowyke, R.; Guillemin, G.J. Fungal Neurotoxins and Sporadic Amyotrophic Lateral Sclerosis. Neurotox. Res. 2019, 35, 969–980. [Google Scholar] [CrossRef] [PubMed]
- Sava, V.; Reunova, O.; Velasquez, A.; Sanchez-Ramos, J. Can low level exposure to ochratoxin—A cause parkinsonism? J. Neurol. Sci. 2006, 249, 68–75. [Google Scholar] [CrossRef]
- Chang, C.; Gershwin, M.E. The Myth of Mycotoxins and Mold Injury. Clin. Rev. Allergy Immunol. 2019, 57, 449–455. [Google Scholar] [CrossRef]
- Rudert, A.; Portnoy, J. Mold allergy: Is it real and what do we do about it? Expert Rev. Clin. Immunol. 2017, 13, 823–835. [Google Scholar] [CrossRef]
- Vincent, M.; Percier, P.; De Prins, S.; Huygen, K.; Potemberg, G.; Muraille, E.; Romano, M.; Michel, O.; Denis, O. Investigation of inflammatory and allergic responses to common mold species: Results from in vitro experiments, from a mouse model of asthma, and from a group of asthmatic patients. Indoor Air 2017, 27, 933–945. [Google Scholar] [CrossRef]
- Portnoy, J.M.; Kennedy, K.; Barnes, C.S. Controversies regarding dampness and mold growth in homes. Allergy Asthma Proc. 2007, 28, 257–258. [Google Scholar] [CrossRef]
- Khalili, B.; Bardana, E.J., Jr. Inhalational mold toxicity: Fact or fiction? A clinical review of 50 cases. Ann. Allergy Asthma Immunol. 2005, 95, 239–246. [Google Scholar] [CrossRef]
- Vignal, C.; Djouina, M.; Pichavant, M.; Caboche, S.; Waxin, C.; Beury, D.; Hot, D.; Gower-Rousseau, C.; Body-Malapel, M. Chronic ingestion of deoxynivalenol at human dietary levels impairs intestinal homeostasis and gut microbiota in mice. Arch. Toxicol. 2018, 92, 2327–2338. [Google Scholar] [CrossRef]
- Pisa, D.; Alonso, R.; Jimenez-Jimenez, F.J.; Carrasco, L. Fungal infection in cerebrospinal fluid from some patients with multiple sclerosis. Eur. J. Clin. Microbiol. Infect. Dis. 2013, 32, 795–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mankai, A.; Sakly, W.; Thabet, Y.; Achour, A.; Manoubi, W.; Ghedira, I. Anti-Saccharomyces cerevisiae antibodies in patients with systemic lupus erythematosus. Rheumatol. Int. 2013, 33, 665–669. [Google Scholar] [CrossRef] [PubMed]
- Vaillant, A.A.J.; Vashisht, R.; Zito, P.M. Immediate Hypersensitivity Reactions. Available online: https://www.ncbi.nlm.nih.gov/books/NBK513315/ (accessed on 30 August 2021).
- Hedayati, M.T.; Pasqualotto, A.C.; Warn, P.A.; Bowyer, P.; Denning, D.W. Aspergillus flavus: Human pathogen, allergen and mycotoxin producer. Microbiology 2007, 153, 1677–1692. [Google Scholar] [CrossRef] [Green Version]
- Bowyer, P.; Fraczek, M.; Denning, D.W. Comparative genomics of fungal allergens and epitopes shows widespread distribution of closely related allergen and epitope orthologues. BMC Genom. 2006, 7, 251. [Google Scholar] [CrossRef] [Green Version]
- Twaroch, T.E.; Curin, M.; Valenta, R.; Swoboda, I. Mold allergens in respiratory allergy: From structure to therapy. Allergy Asthma Immunol. Res. 2015, 7, 205–220. [Google Scholar] [CrossRef] [Green Version]
- Mims, J.W. Asthma: Definitions and pathophysiology. Int. Forum Allergy Rhinol. 2015, 5, S2–S6. [Google Scholar] [CrossRef]
- Dykewicz, M.S.; Rodrigues, J.M.; Slavin, R.G. Allergic fungal rhinosinusitis. J. Allergy Clin. Immunol. 2018, 142, 341–351. [Google Scholar] [CrossRef] [Green Version]
- Moller, J.; Hyldgaard, C.; Kronborg-White, S.B.; Rasmussen, F.; Bendstrup, E. Hypersensitivity pneumonitis among wind musicians—An overlooked disease? Eur. Clin. Respir. J. 2017, 4, 1351268. [Google Scholar] [CrossRef] [Green Version]
- Ohkubo, H.; Okayama, M.; Fukumitsu, K.; Niimi, A. Summer-type hypersensitivity pneumonitis in a patient with rheumatoid arthritis on methotrexate and tacrolimus. Respirol. Case Rep. 2016, 4, e00194. [Google Scholar] [CrossRef] [PubMed]
- Mohr, L.C. Hypersensitivity pneumonitis. Curr. Opin. Pulm. Med. 2004, 10, 401–411. [Google Scholar] [CrossRef]
- Sakamoto, T.; Yamasaki, A.; Funaki, Y.; Harada, T.; Okazaki, R.; Hasegawa, Y.; Sueda, Y.; Fukushima, T.; Morita, M.; Yamamoto, A.; et al. An onion farmer with a case of hypersensitivity pneumonitis caused by Aspergillus niger. Respir. Med. Case Rep. 2018, 23, 60–62. [Google Scholar] [CrossRef] [PubMed]
- Benkerroum, N. Chronic and Acute Toxicities of Aflatoxins: Mechanisms of Action. Int. J. Environ. Res. Public Health 2020, 17, 423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shirani, K.; Zanjani, B.R.; Mahmoudi, M.; Jafarian, A.H.; Hassani, F.V.; Giesy, J.P.; Karimi, G. Immunotoxicity of aflatoxin M1: As a potent suppressor of innate and acquired immune systems in a subacute study. J. Sci. Food Agric. 2018, 98, 5884–5892. [Google Scholar] [CrossRef] [PubMed]
- Reif, D.; Martin, M.; Tan, S.; Houck, K.; Judson, R.S.; Richard, A.M.; Knudsen, T.B.; Dix, D.J.; Kavlock, R.J. Endocrine profiling and prioritization of environmental chemicals using ToxCast data. Environ. Health Perspect. 2010, 118, 1714–1720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marin, D.E.; Pistol, G.C.; Gras, M.A.; Palade, M.L.; Taranu, I. Comparative effect of ochratoxin A on inflammation and oxidative stress parameters in gut and kidney of piglets. Regul. Toxicol. Pharmacol. 2017, 89, 224–231. [Google Scholar] [CrossRef]
- Malir, F.; Ostry, V.; Pfohl-Leszkowicz, A.; Malir, J.; Toman, J. Ochratoxin A: 50 Years of Research. Toxins 2016, 8, 191. [Google Scholar] [CrossRef] [Green Version]
- Wood, L.F.; Wood, M.P.; Fisher, B.S.; Jaspan, H.B.; Sodora, D.L. T Cell Activation in South African HIV-Exposed Infants Correlates with Ochratoxin A Exposure. Front. Immunol. 2017, 8, 1857. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.Y.; DeKruyff, R.H.; Umetsu, D.T. The many paths to asthma: Phenotype shaped by innate and adaptive immunity. Nat. Immunol. 2010, 11, 577–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khurana, S.; Jarjour, N.N. Systematic Approach to Asthma of Varying Severity. Clin. Chest Med. 2019, 40, 59–70. [Google Scholar] [CrossRef]
- Chung, K.F.; Wenzel, S.E.; Brozek, J.L.; Bush, A.; Castro, M.; Sterk, P.J.; Adcock, I.M.; Bateman, E.D.; Bel, E.H.; Bleecker, E.R.; et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur. Respir. J. 2014, 43, 343–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mendell, M.J.; Mirer, A.G.; Cheung, K.; Tong, M.; Douwes, J. Respiratory and allergic health effects of dampness, mold, and dampness-related agents: A review of the epidemiologic evidence. Environ. Health Perspect. 2011, 119, 748–756. [Google Scholar] [CrossRef] [PubMed]
- Vincent, M.; Corazza, F.; Chasseur, C.; Bladt, S.; Romano, M.; Huygen, K.; Denis, O.; Michel, O. Relationship between mold exposure, specific IgE sensitization, and clinical asthma: A case-control study. Ann. Allergy Asthma Immunol. 2018, 121, 333–339. [Google Scholar] [CrossRef]
- Akar-Ghibril, N.; Casale, T.; Custovic, A.; Phipatanakul, W. Allergic Endotypes and Phenotypes of Asthma. J. Allergy Clin. Immunol. Pract. 2020, 8, 429–440. [Google Scholar] [CrossRef]
- Fitzpatrick, A.M.; Gaston, B.M.; Erzurum, S.C.; Teague, W.G.; Severe Asthma Research Program; National Institutes of Health. Features of severe asthma in school-age children: Atopy and increased exhaled nitric oxide. J. Allergy Clin. Immunol. 2006, 118, 1218–1225. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Biagini Myers, J.M.; Brandt, E.B.; Ryan, P.H.; Lindsey, M.; Mintz-Cole, R.A.; Reponen, T.; Vesper, S.J.; Forde, F.; Ruff, B.; et al. β-Glucan exacerbates allergic asthma independent of fungal sensitization and promotes steroid-resistant TH2/TH17 responses. J. Allergy Clin. Immunol. 2017, 139, 54–65. [Google Scholar] [CrossRef] [Green Version]
- Schutze, N.; Lehmann, I.; Bonisch, U.; Simon, J.C.; Polte, T. Exposure to mycotoxins increases the allergic immune response in a murine asthma model. Am. J. Respir. Crit. Care Med. 2010, 181, 1188–1199. [Google Scholar] [CrossRef]
- Campbell, A.W.; Thrasher, J.D.; Madison, R.A.; Vojdani, A.; Gray, M.R.; Johnson, A. Neural autoantibodies and neurophysiologic abnormalities in patients exposed to molds in water-damaged buildings. Arch. Environ. Health 2003, 58, 464–474. [Google Scholar] [CrossRef]
- Gray, M.R.; Thrasher, J.D.; Crago, R.; Madison, R.A.; Arnold, L.; Campbell, A.W.; Vojdani, A. Mixed mold mycotoxicosis: Immunological changes in humans following exposure in water-damaged buildings. Arch. Environ. Health 2003, 58, 410–420. [Google Scholar] [CrossRef]
- Abou-Donia, M.B.; Lieberman, A.; Curtis, L. Neural autoantibodies in patients with neurological symptoms and histories of chemical/mold exposures. Toxicol. Ind. Health 2018, 34, 44–53. [Google Scholar] [CrossRef]
- Fraga-Silva, T.F.C.; Mimura, L.A.N.; Leite, L.C.T.; Borim, P.A.; Ishikawa, L.L.W.; Venturini, J.; Arruda, M.S.P.; Sartori, A. Gliotoxin Aggravates Experimental Autoimmune Encephalomyelitis by Triggering Neuroinflammation. Toxins 2019, 11, 443. [Google Scholar] [CrossRef] [Green Version]
- Lieberman, A.; Curtis, L. Mold Exposure and Mitochondrial Antibodies. Altern. Ther. Health Med. 2020, 26, 44–47. [Google Scholar] [PubMed]
- Islam, M.T.; Mishra, S.K.; Tripathi, S.; de Alencar, M.; JMC, E.S.; Rolim, H.M.L.; de Medeiros, M.; Ferreira, P.M.P.; Rouf, R.; Uddin, S.J.; et al. Mycotoxin-assisted mitochondrial dysfunction and cytotoxicity: Unexploited tools against proliferative disorders. IUBMB Life 2018, 70, 1084–1092. [Google Scholar] [CrossRef]
- Olsson, Å.R.; Skogh, T.; Wingren, G. Comorbidity and lifestyle, reproductive factors, and environmental exposures associated with rheumatoid arthritis. Ann. Rheum. Dis. 2001, 60, 934–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schønheyder, H.; Andersen, I.; Andersen, P. Serum antibodies to Aspergillus fumigatus in patients with rheumatic diseases. Sabouraudia 1983, 21, 149–157. [Google Scholar] [CrossRef]
- Jahreis, S.; Kuhn, S.; Madaj, A.M.; Bauer, M.; Polte, T. Mold metabolites drive rheumatoid arthritis in mice via promotion of IFN-gamma- and IL-17-producing T cells. Food Chem. Toxicol. 2017, 109, 405–413. [Google Scholar] [CrossRef] [PubMed]
- Stange, E.F.; Schroeder, B.O. Microbiota and mucosal defense in IBD: An update. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 963–976. [Google Scholar] [CrossRef] [Green Version]
- Gunn, S.R.; Gunn, G.G.; Mueller, F.W. Reversal of Refractory Ulcerative Colitis and Severe Chronic Fatigue Syndrome Symptoms Arising from Immune Disturbance in an HLA-DR/DQ Genetically Susceptible Individual with Multiple Biotoxin Exposures. Am. J. Case Rep. 2016, 17, 320–325. [Google Scholar] [CrossRef] [Green Version]
- Brewer, J.H.; Thrasher, J.D.; Hooper, D. Chronic illness associated with mold and mycotoxins: Is naso-sinus fungal biofilm the culprit? Toxins 2013, 6, 66–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Payros, D.; Menard, S.; Laffitte, J.; Neves, M.; Tremblay-Franco, M.; Luo, S.; Fouche, E.; Snini, S.P.; Theodorou, V.; Pinton, P.; et al. The food contaminant, deoxynivalenol, modulates the Thelper/Treg balance and increases inflammatory bowel diseases. Arch. Toxicol. 2020, 94, 3173–3184. [Google Scholar] [CrossRef] [PubMed]
- Fan, W.; Lv, Y.; Ren, S.; Shao, M.; Shen, T.; Huang, K.; Zhou, J.; Yan, L.; Song, S. Zearalenone (ZEA)-induced intestinal inflammation is mediated by the NLRP3 inflammasome. Chemosphere 2018, 190, 272–279. [Google Scholar] [CrossRef]
- Jolly, P.E.; Shuaib, F.M.; Jiang, Y.; Preko, P.; Baidoo, J.; Stiles, J.K.; Wang, J.S.; Phillips, T.D.; Williams, J.H. Association of high viral load and abnormal liver function with high aflatoxin B1-albumin adduct levels in HIV-positive Ghanaians: Preliminary observations. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess. 2011, 28, 1224–1234. [Google Scholar] [CrossRef] [PubMed]
- Jolly, P.E.; Inusah, S.; Lu, B.; Ellis, W.O.; Nyarko, A.; Phillips, T.D.; Williams, J.H. Association between high aflatoxin B1 levels and high viral load in HIV-positive people. World Mycotoxin. J. 2013, 6, 255–261. [Google Scholar] [CrossRef] [PubMed]
- Correale, J.; Gaitan, M.I.; Ysrraelit, M.C.; Fiol, M.P. Progressive multiple sclerosis: From pathogenic mechanisms to treatment. Brain 2017, 140, 527–546. [Google Scholar] [CrossRef] [Green Version]
- Pospíšil, A.F.M. Role of ergot alkaloids in the immune system. In Ergot the Genus Claviceps, 1st ed.; CRC Press: Boca Raton, FL, USA, 1999; p. 18. [Google Scholar]
- Jolly, P.E. Aflatoxin: Does it contribute to an increase in HIV viral load? Future Microbiol. 2014, 9, 121–124. [Google Scholar] [CrossRef]
- Hellings, P.W.; Steelant, B. Epithelial barriers in allergy and asthma. J. Allergy Clin. Immunol. 2020, 145, 1499–1509. [Google Scholar] [CrossRef]
- Gao, Y.; Meng, L.; Liu, H.; Wang, J.; Zheng, N. The Compromised Intestinal Barrier Induced by Mycotoxins. Toxins 2020, 12, 619. [Google Scholar] [CrossRef]
- Brown, R.; Priest, E.; Naglik, J.R.; Richardson, J.P. Fungal Toxins and Host Immune Responses. Front. Microbiol. 2021, 12, 643639. [Google Scholar] [CrossRef]
- Mintz-Cole, R.A.; Brandt, E.B.; Bass, S.A.; Gibson, A.M.; Reponen, T.; Khurana Hershey, G.K. Surface availability of beta-glucans is critical determinant of host immune response to Cladosporium cladosporioides. J. Allergy Clin. Immunol. 2013, 132, 159–169. [Google Scholar] [CrossRef]
- Leitão, E.A.; Bittencourt, V.C.B.; Haido, R.M.T.; Valente, A.P.; Peter-Katalinic, J.; Letzel, M.; de Souza, L.M.; Barreto-Bergter, E. β-Galactofuranose-containing O-linked oligosaccharides present in the cell wall peptidogalactomannan of Aspergillus fumigatus contain immunodominant epitopes. Glycobiology 2003, 13, 681–692. [Google Scholar] [CrossRef]
- Horner, W.E.; Helbling, A.; Salvaggio, J.E.; Lehrer, S.B. Fungal allergens. Clin. Microbiol. Rev. 1995, 8, 161–179. [Google Scholar] [CrossRef]
- Vijay, H.M.; Kurup, V.P. Fungal allergens. Clin. Allergy Immunol. 2008, 21, 141–160. [Google Scholar]
- Rivera, A. Protective immune responses to fungal infections. Parasite Immunol. 2014, 36, 453–462. [Google Scholar] [CrossRef]
- Rivera, A.; Hohl, T.M.; Collins, N.; Leiner, I.; Gallegos, A.; Saijo, S.; Coward, J.W.; Iwakura, Y.; Pamer, E.G. Dectin-1 diversifies Aspergillus fumigatus–specific T cell responses by inhibiting T helper type 1 CD4 T cell differentiation. J. Exp. Med. 2011, 208, 369–381. [Google Scholar] [CrossRef]
- Romani, L. Immunity to fungal infections. Nat. Rev. Immunol. 2011, 11, 275–288. [Google Scholar] [CrossRef]
- Tang, J.; Lin, G.; Langdon, W.Y.; Tao, L.; Zhang, J. Regulation of C-Type Lectin Receptor-Mediated Antifungal Immunity. Front. Immunol. 2018, 9, 123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Drummond, R.A.; Saijo, S.; Iwakura, Y.; Brown, G.D. The role of Syk/CARD9 coupled C-type lectins in antifungal immunity. Eur. J. Immunol. 2011, 41, 276–281. [Google Scholar] [CrossRef] [PubMed]
- Werner, J.L.; Metz, A.E.; Horn, D.; Schoeb, T.R.; Hewitt, M.M.; Schwiebert, L.M.; Faro-Trindade, I.; Brown, G.D.; Steele, C. Requisite Role for the Dectin-1 β-Glucan Receptor in Pulmonary Defense against Aspergillus fumigatus. J. Immunol. 2009, 182, 4938–4946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Creasia, D.A.; Thurman, J.D.; Wannemacher, R.W., Jr.; Bunner, D.L. Acute inhalation toxicity of T-2 mycotoxin in the rat and guinea pig. Fundam. Appl. Toxicol. 1990, 14, 54–59. [Google Scholar] [CrossRef]
- Capasso, L.; Longhin, E.; Caloni, F.; Camatini, M.; Gualtieri, M. Synergistic inflammatory effect of PM10 with mycotoxin deoxynivalenol on human lung epithelial cells. Toxicon 2015, 104, 65–72. [Google Scholar] [CrossRef] [PubMed]
- Amuzie, C.J.; Harkema, J.R.; Pestka, J.J. Tissue distribution and proinflammatory cytokine induction by the trichothecene deoxynivalenol in the mouse: Comparison of nasal vs. oral exposure. Toxicology 2008, 248, 39–44. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Shen, H.; Yi, L.; Shao, P.; Soulika, A.M.; Meng, X.; Xing, L.; Yan, X.; Zhang, X. Oral administration of aflatoxin G1 induces chronic alveolar inflammation associated with lung tumorigenesis. Toxicol. Lett. 2015, 232, 547–556. [Google Scholar] [CrossRef]
- Guindon-Kezis, K.A.; Mulder, J.E.; Massey, T.E. In vivo treatment with aflatoxin B1 increases DNA oxidation, base excision repair activity and 8-oxoguanine DNA glycosylase 1 levels in mouse lung. Toxicology 2014, 321, 21–26. [Google Scholar] [CrossRef]
- Bahrami, R.; Shahbazi, Y.; Nikousefat, Z. Aflatoxin M1 in milk and traditional dairy products from west part of Iran: Occurrence and seasonal variation with an emphasis on risk assessment of human exposure. Food Control. 2016, 62, 250–256. [Google Scholar] [CrossRef]
- Romero, A.; Ares, I.; Ramos, E.; Castellano, V.; Martínez, M.; Martínez-Larrañaga, M.R.; Anadón, A.; Martínez, M.A. Mycotoxins modify the barrier function of Caco-2 cells through differential gene expression of specific claudin isoforms: Protective effect of illite mineral clay. Toxicology 2016, 353–354, 21–33. [Google Scholar] [CrossRef]
- Jia, X.; Chen, F.; Pan, W.; Yu, R.; Tian, S.; Han, G.; Fang, H.; Wang, S.; Zhao, J.; Li, X.; et al. Gliotoxin promotes Aspergillus fumigatus internalization into type II human pneumocyte A549 cells by inducing host phospholipase D activation. Microbes Infect. 2014, 16, 491–501. [Google Scholar] [CrossRef]
- Zhang, C.; Chen, F.; Liu, X.; Han, X.; Hu, Y.; Su, X.; Chen, Y.; Sun, Y.; Han, L. Gliotoxin Induces Cofilin Phosphorylation to Promote Actin Cytoskeleton Dynamics and Internalization of Aspergillus fumigatus Into Type II Human Pneumocyte Cells. Front. Microbiol. 2019, 10, 1345. [Google Scholar] [CrossRef]
- Lee, R.J.; Workman, A.D.; Carey, R.M.; Chen, B.; Rosen, P.L.; Doghramji, L.; Adappa, N.D.; Palmer, J.N.; Kennedy, D.W.; Cohen, N.A. Fungal Aflatoxins Reduce Respiratory Mucosal Ciliary Function. Sci. Rep. 2016, 6, 33221. [Google Scholar] [CrossRef]
- Cui, J.; Wang, J.; Huang, S.; Jiang, X.; Li, Y.; Wu, W.; Zhang, X. Sterigmatocystin induced apoptosis in human pulmonary cells in vitro. Exp. Toxicol. Pathol. 2017, 69, 695–699. [Google Scholar] [CrossRef]
- Patel, R.; Hossain, M.A.; German, N.; Al-Ahmad, A.J. Gliotoxin penetrates and impairs the integrity of the human blood-brain barrier in vitro. Mycotoxin. Res. 2018, 34, 257–268. [Google Scholar] [CrossRef]
- Gon, Y.; Hashimoto, S. Role of airway epithelial barrier dysfunction in pathogenesis of asthma. Allergol. Int. 2018, 67, 12–17. [Google Scholar] [CrossRef]
- Parikh, K.; Antanaviciute, A.; Fawkner-Corbett, D.; Jagielowicz, M.; Aulicino, A.; Lagerholm, C.; Davis, S.; Kinchen, J.; Chen, H.H.; Alham, N.K.; et al. Colonic epithelial cell diversity in health and inflammatory bowel disease. Nature 2019, 567, 49–55. [Google Scholar] [CrossRef]
- Fukui, H. Increased Intestinal Permeability and Decreased Barrier Function: Does It Really Influence the Risk of Inflammation? Inflamm. Intest. Dis. 2016, 1, 135–145. [Google Scholar] [CrossRef] [PubMed]
- Shi, N.; Li, N.; Duan, X.; Niu, H. Interaction between the gut microbiome and mucosal immune system. Mil. Med. Res. 2017, 4, 14. [Google Scholar] [CrossRef]
- De Luca, F.; Shoenfeld, Y. The microbiome in autoimmune diseases. Clin. Exp. Immunol. 2019, 195, 74–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guerre, P. Mycotoxin and Gut Microbiota Interactions. Toxins 2020, 12, 769. [Google Scholar] [CrossRef]
- Zhang, W.; Zhang, S.; Wang, J.; Shan, A.; Xu, L. Changes in intestinal barrier functions and gut microbiota in rats exposed to zearalenone. Ecotoxicol. Environ. Saf. 2020, 204, 111072. [Google Scholar] [CrossRef] [PubMed]
- Payros, D.; Dobrindt, U.; Martin, P.; Secher, T.; Bracarense, A.P.; Boury, M.; Laffitte, J.; Pinton, P.; Oswald, E.; Oswald, I.P. The Food Contaminant Deoxynivalenol Exacerbates the Genotoxicity of Gut Microbiota. mBio 2017, 8, e00007-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Danicke, S.; Winkler, J.; Meyer, U.; Frahm, J.; Kersten, S. Haematological, clinical-chemical and immunological consequences of feeding Fusarium toxin contaminated diets to early lactating dairy cows. Mycotoxin. Res. 2017, 33, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theofani, E.; Semitekolou, M.; Morianos, I.; Samitas, K.; Xanthou, G. Targeting NLRP3 Inflammasome Activation in Severe Asthma. J. Clin. Med. 2019, 8, 1615. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhen, Y.; Zhang, H. NLRP3 Inflammasome and Inflammatory Bowel Disease. Front. Immunol. 2019, 10, 276. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.Y.; Zhan, D.L.; Chen, Y.Y.; Wang, W.H.; He, C.Y.; Lin, Y.; Lin, Y.C.; Lin, Z.N. Aflatoxin B1 enhances pyroptosis of hepatocytes and activation of Kupffer cells to promote liver inflammatory injury via dephosphorylation of cyclooxygenase-2: An in vitro, ex vivo and in vivo study. Arch. Toxicol. 2019, 93, 3305–3320. [Google Scholar] [CrossRef] [PubMed]
- Kankkunen, P.; Valimaki, E.; Rintahaka, J.; Palomaki, J.; Nyman, T.; Alenius, H.; Wolff, H.; Matikainen, S. Trichothecene mycotoxins activate NLRP3 inflammasome through a P2X7 receptor and Src tyrosine kinase dependent pathway. Hum. Immunol. 2014, 75, 134–140. [Google Scholar] [CrossRef]
- Chu, Q.; Wang, S.; Jiang, L.; Jiao, Y.; Sun, X.; Li, J.; Yang, L.; Hou, Y.; Wang, N.; Yao, X.; et al. Patulin induces pyroptosis through the autophagic-inflammasomal pathway in liver. Food Chem. Toxicol. 2021, 147, 111867. [Google Scholar] [CrossRef] [PubMed]
- Chavan, S.S.; Pavlov, V.A.; Tracey, K.J. Mechanisms and Therapeutic Relevance of Neuro-immune Communication. Immunity 2017, 46, 927–942. [Google Scholar] [CrossRef] [Green Version]
- Moulton, C.D.; Pavlidis, P.; Norton, C.; Norton, S.; Pariante, C.; Hayee, B.; Powell, N. Depressive symptoms in inflammatory bowel disease: An extraintestinal manifestation of inflammation? Clin. Exp. Immunol. 2019, 197, 308–318. [Google Scholar] [CrossRef] [Green Version]
- Passaro, A.P.; Lebos, A.L.; Yao, Y.; Stice, S.L. Immune Response in Neurological Pathology: Emerging Role of Central and Peripheral Immune Crosstalk. Front. Immunol. 2021, 12, 676621. [Google Scholar] [CrossRef]
- Becher, B.; Spath, S.; Goverman, J. Cytokine networks in neuroinflammation. Nat. Rev. Immunol. 2017, 17, 49–59. [Google Scholar] [CrossRef]
- Guillemin, G.J.; Cullen, K.M.; Lim, C.K.; Smythe, G.A.; Garner, B.; Kapoor, V.; Takikawa, O.; Brew, B.J. Characterization of the kynurenine pathway in human neurons. J. Neurosci. 2007, 27, 12884–12892. [Google Scholar] [CrossRef] [Green Version]
- O’Farrell, K.; Harkin, A. Stress-related regulation of the kynurenine pathway: Relevance to neuropsychiatric and degenerative disorders. Neuropharmacology 2017, 112, 307–323. [Google Scholar] [CrossRef] [Green Version]
- Abautret-Daly, Á.; Dempsey, E.; Riestra, S.; de Francisco-García, R.; Parra-Blanco, A.; Rodrigo, L.; Medina, C.; Connor, T.J.; Harkin, A. Association between psychological measures with inflammatory anddisease-related markers of inflammatory bowel disease. Int. J. Psychiatry Clin. Pract. 2017, 21, 221–230. [Google Scholar] [CrossRef]
- Valkanova, V.; Ebmeier, K.P.; Allan, C.L. CRP, IL-6 and depression: A systematic review and meta-analysis of longitudinal studies. J. Affect. Disord. 2013, 150, 736–744. [Google Scholar] [CrossRef]
- Benito-León, J.; Laurence, M. The Role of Fungi in the Etiology of Multiple Sclerosis. Front. Neurol. 2017, 8, 535. [Google Scholar] [CrossRef] [PubMed]
- Hachim, M.Y.; Elemam, N.M.; Maghazachi, A.A. The Beneficial and Debilitating Effects of Environmental and Microbial Toxins, Drugs, Organic Solvents and Heavy Metals on the Onset and Progression of Multiple Sclerosis. Toxins 2019, 11, 147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Purzycki, C.B.; Shain, D.H. Fungal toxins and multiple sclerosis: A compelling connection. Brain Res. Bull. 2010, 82, 4–6. [Google Scholar] [CrossRef] [PubMed]
- Guo, S.; Al-Sadi, R.; Said, H.M.; Ma, T.Y. Lipopolysaccharide causes an increase in intestinal tight junction permeability in vitro and in vivo by inducing enterocyte membrane expression and localization of TLR-4 and CD14. Am. J. Pathol. 2013, 182, 375–387. [Google Scholar] [CrossRef] [Green Version]
- Leonard, B.; Maes, M. Mechanistic explanations how cell-mediated immune activation, inflammation and oxidative and nitrosative stress pathways and their sequels and concomitants play a role in the pathophysiology of unipolar depression. Neurosci. Biobehav. Rev. 2012, 36, 764–785. [Google Scholar] [CrossRef] [PubMed]
- Steinberg, B.E.; Silverman, H.A.; Robbiati, S.; Gunasekaran, M.K.; Tsaava, T.; Battinelli, E.; Stiegler, A.; Bouton, C.E.; Chavan, S.S.; Tracey, K.J.; et al. Cytokine-specific neurograms in the sensory vagus nerve. Bioelectron. Med. 2016, 3, 7–17. [Google Scholar] [CrossRef]
- Suzuki, K. Chronic Inflammation as an Immunological Abnormality and Effectiveness of Exercise. Biomolecules 2019, 9, 223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pinkerton, J.W.; Kim, R.Y.; Robertson, A.A.B.; Hirota, J.A.; Wood, L.G.; Knight, D.A.; Cooper, M.A.; O’Neill, L.A.J.; Horvat, J.C.; Hansbro, P.M. Inflammasomes in the lung. Mol. Immunol. 2017, 86, 44–55. [Google Scholar] [CrossRef]
- Friedrich, M.; Pohin, M.; Powrie, F. Cytokine Networks in the Pathophysiology of Inflammatory Bowel Disease. Immunity 2019, 50, 992–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wichmann, G.; Herbarth, O.; Lehmann, I. The mycotoxins citrinin, gliotoxin, and patulin affect interferon-gamma rather than interleukin-4 production in human blood cells. Environ. Toxicol. 2002, 17, 211–218. [Google Scholar] [CrossRef] [PubMed]
- Luft, P.; Oostingh, G.J.; Gruijthuijsen, Y.; Horejs-Hoeck, J.; Lehmann, I.; Duschl, A. Patulin influences the expression of Th1/Th2 cytokines by activated peripheral blood mononuclear cells and T cells through depletion of intracellular glutathione. Environ. Toxicol. 2008, 23, 84–95. [Google Scholar] [CrossRef] [PubMed]
- Reider, N.; Reider, D.; Ebner, S.; Holzmann, S.; Herold, M.; Fritsch, P.; Romani, N. Dendritic cells contribute to the development of atopy by an insufficiency in IL-12 production. J. Allergy Clin. Immunol. 2002, 109, 89–95. [Google Scholar] [CrossRef]
- Chen, X.Q.; Yang, J.; Hu, S.P.; Nie, H.X.; Mao, G.Y.; Chen, H.B. Increased expression of CD86 and reduced production of IL-12 and IL-10 by monocyte-derived dendritic cells from allergic asthmatics and their effects on Th1- and Th2-type cytokine balance. Respiration 2006, 73, 34–40. [Google Scholar] [CrossRef]
- Aghaei, H.; Farhadi, E.; Akhtari, M.; Shahba, S.; Mostafaei, S.; Jamshidi, A.; Poursani, S.; Mahmoudi, M.; Nicknam, M.H. Copy number variation of IL17RA gene and its association with the ankylosing spondylitis risk in Iranian patients: A case-control study. BMC Med. Genet. 2020, 21, 147. [Google Scholar] [CrossRef]
- Shahba, S.; Jafari Shakib, R.; Jamshidi, A.; Vojdanian, M.; Akhtari, M.; Aslani, S.; Poursani, S.; Nikokar, I.; Mahmoudi, M. Association study of copy number variation in BMP8A gene with the risk of ankylosing spondylitis in Iranian population. J. Cell Biochem. 2019, 120, 8359–8365. [Google Scholar] [CrossRef]
- Su, J.; Liu, D.; Wang, Q.; Lin, J.; Song, S.; Huang, K. Long-Time Instead of Short-Time Exposure in Vitro and Administration in Vivo of Ochratoxin A Is Consistent in Immunosuppression. J. Agric. Food Chem. 2019, 67, 7485–7495. [Google Scholar] [CrossRef]
- Wong, J.; Magun, B.E.; Wood, L.J. Lung inflammation caused by inhaled toxicants: A review. Int. J. Chron. Obstruct. Pulmon. Dis. 2016, 11, 1391–1401. [Google Scholar] [CrossRef] [Green Version]
- Rocha, O.; Ansari, K.; Doohan, F.M. Effects of trichothecene mycotoxins on eukaryotic cells: A review. Food Addit. Contam. 2005, 22, 369–378. [Google Scholar] [CrossRef] [PubMed]
- Königs, M.; Lenczyk, M.; Schwerdt, G.; Holzinger, H.; Gekle, M.; Humpf, H.U. Cytotoxicity, metabolism and cellular uptake of the mycotoxin deoxynivalenol in human proximal tubule cells and lung fibroblasts in primary culture. Toxicology 2007, 240, 48–59. [Google Scholar] [CrossRef]
- Islam, Z.; Gray, J.S.; Pestka, J.J. p38 Mitogen-activated protein kinase mediates IL-8 induction by the ribotoxin deoxynivalenol in human monocytes. Toxicol. Appl. Pharmacol. 2006, 213, 235–244. [Google Scholar] [CrossRef]
- Ciacci-Zanella, J.R.; Jones, C. Fumonisin B1, a mycotoxin contaminant of cereal grains, and inducer of apoptosis via the tumour necrosis factor pathway and caspase activation. Food Chem. Toxicol. 1999, 37, 703–712. [Google Scholar] [CrossRef]
- Berek, L.; Mesterhazy, A.; Teren, J.; Molnar, J. Effects of mycotoxins on human immune functions in vitro. Toxicol. In Vitro 2001, 15, 25–30. [Google Scholar] [CrossRef]
- Jiang, Y.; Jolly, P.E.; Preko, P.; Wang, J.S.; Ellis, W.O.; Phillips, T.D.; Williams, J.H. Aflatoxin-related immune dysfunction in health and in human immunodeficiency virus disease. Clin. Dev. Immunol. 2008, 2008, 790309. [Google Scholar] [CrossRef]
- Ramyaa, P.; Krishnaswamy, R.; Padma, V.V. Quercetin modulates OTA-induced oxidative stress and redox signalling in HepG2 cells—Up regulation of Nrf2 expression and down regulation of NF-kappaB and COX-2. Biochim. Biophys. Acta 2014, 1840, 681–692. [Google Scholar] [CrossRef]
- Gan, F.; Zhang, Z.; Hu, Z.; Hesketh, J.; Xue, H.; Chen, X.; Hao, S.; Huang, Y.; Cole Ezea, P.; Parveen, F.; et al. Ochratoxin A promotes porcine circovirus type 2 replication in vitro and in vivo. Free Radic. Biol. Med. 2015, 80, 33–47. [Google Scholar] [CrossRef]
- Ivanov, A.V.; Valuev-Elliston, V.T.; Ivanova, O.N.; Kochetkov, S.N.; Starodubova, E.S.; Bartosch, B.; Isaguliants, M.G. Oxidative Stress during HIV Infection: Mechanisms and Consequences. Oxid. Med. Cell Longev. 2016, 2016, 8910396. [Google Scholar] [CrossRef] [Green Version]
- Hunt, P.W. HIV and inflammation: Mechanisms and consequences. Curr. HIV/AIDS Rep. 2012, 9, 139–147. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kraft, S.; Buchenauer, L.; Polte, T. Mold, Mycotoxins and a Dysregulated Immune System: A Combination of Concern? Int. J. Mol. Sci. 2021, 22, 12269. https://doi.org/10.3390/ijms222212269
Kraft S, Buchenauer L, Polte T. Mold, Mycotoxins and a Dysregulated Immune System: A Combination of Concern? International Journal of Molecular Sciences. 2021; 22(22):12269. https://doi.org/10.3390/ijms222212269
Chicago/Turabian StyleKraft, Stephanie, Lisa Buchenauer, and Tobias Polte. 2021. "Mold, Mycotoxins and a Dysregulated Immune System: A Combination of Concern?" International Journal of Molecular Sciences 22, no. 22: 12269. https://doi.org/10.3390/ijms222212269