Next Article in Journal
A Thermostable Dissolving Microneedle Vaccine with Recombinant Protein of Botulinum Neurotoxin Serotype A
Previous Article in Journal
Comparative Analysis of Aristolochic Acids in Aristolochia Medicinal Herbs and Evaluation of Their Toxicities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Hypoxia and Cellular Senescence, Emerging Toxic Mechanisms of Mycotoxins and Toxins: A New Understanding of the Negative Immune Regulations

1
College of Life Science, Yangtze University, Jingzhou 434025, China
2
Department of Chemistry, Faculty of Science, University of Hradec Králové, 50003 Hradec Králové, Czech Republic
3
College of Physical Education and Health, Chongqing College of International Business and Economics, Chongqing 401520, China
4
School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, China
5
National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan 430070, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Toxins 2022, 14(12), 880; https://doi.org/10.3390/toxins14120880
Submission received: 7 December 2022 / Accepted: 13 December 2022 / Published: 16 December 2022
(This article belongs to the Section Mycotoxins)
Mycotoxin contamination is an important issue that has plagued the world. Mycotoxins not only contaminate food and feed but, more importantly, can further cause poisoning in humans and animals through food and feed. Moreover, children seem to be more sensitive to mycotoxins [1]. The risk of exposure to the trichothecene mycotoxin deoxynivalenol (DON) in adolescents is higher than in adults [2]. Immunotoxicity is a specific toxicity of concern from mycotoxins. Mycotoxins inhibit the proliferation of leukocytes and suppress the immune system [3]. Trichothecene mycotoxins have the ability to induce apoptosis in macrophages [4]. Other toxins, such as aflatoxin M1 and ochratoxin A (OTA), induce a competitive endogenous RNA regulatory network of intestinal immunosuppression [5]. Moreover, some mycotoxins, including zearalenone and DON, aggravate disease progression by inhibiting the inflammatory response following infection of foodborne bacteria (for example, L. monocytogenes) [6] and viruses [7]. On the other hand, the global outbreak of animal diseases is closely related to mycotoxin contamination in the environment, as these toxins reduce the immunity of animals. Moreover, mycotoxin contamination in feed is an important risk factor for animals’ susceptibility to pathogens [8,9].
Currently, studies have focused on the potential mechanisms of mycotoxin immunotoxicity and found that oxidative stress, apoptosis, and autophagy are important toxic effects and mechanisms. Researchers have tried to reveal the upstream regulatory mechanisms from signaling pathways such as hemopoietic cell kinase (HCK) and RNA-activated protein kinase R (PKR) [10,11]. In addition, some crucial regulatory mechanisms have been gradually reported in recent years, such as the TLRs/NF-κB pathway and other crossing pathways, including cyclooxygenase-2 [12], and endoplasmic reticulum stress-activated PERK-eIF2α-ATF4 signaling [13], which play important roles in the immunomodulation of mycotoxins. Moreover, Notch1 signaling is concerned since this pathway is significantly activated after DON exposure [14]. In addition, mycotoxins can inhibit the expression of immune response factors and mediate an intracellular active “immune evasion” process, reducing the body’s cellular immune defense function and altering the cellular immune microenvironment, allowing the toxins to further poison cells [15]. In this regard, the hypoxic microenvironment and hypoxia-inducible factor (HIF) play the unique roles, since HIF-1α inhibits the trichothecene mycotoxin-mediated “immune evasion” process by negatively regulating programmed death-1/programmed death-ligand 1 [16].
Indeed, hypoxia is involved in the toxic mechanisms of mycotoxins [17,18]. In addition to its occurrence in trichothecenes [19,20], HIF-1α is also involved in the regulation of toxic mechanism of other toxins. OTA can upregulate HIF-1α and then lead to tumorigenesis through altering growth signaling transforming growth factor-β and vascular endothelial growth factor [21]. OTA causes kidney toxicity via the AhR-Smad2/3-HIF-1α pathways [22]. In hypoxic conditions, ROS formation is increased in hypoxic conditions, which contribute to further activation of HIF-1α [23]. As a new toxicity mechanism of mycotoxins, the relationship among hypoxia, oxidative stress, and immunotoxicity needs to be studied. Importantly, recent studies further show that cellular senescence plays a potential function in the immunotoxicity of mycotoxins. Cellular senescence can be triggered by oxidative stress in the DNA damage response [24,25,26]. Mycotoxins induce cellular senescence and cause senescence-associated cell cycle arrest [27,28]. OTA increases the senescence biomarkers expression of p53, γ-H2AX, and the senescence-associated secretory phenotype (SASP) inflammatory factors [29]. Moreover, OTA induces renal cell senescence by modulating the expression of cyclin-dependent kinase 2 [30]. Another mycotoxin, alternariol, induces an arrest in the G2/M transition by increasing the expression of cyclin B and SA-β-gal activity [31,32]. Aflatoxin B1 (AFB1) upregulates CXCL8 expression and causes cell cycle arrest [28]. In addition, AFB1 causes G0G1 cell cycle arrest by downregulating cyclin D1 expression [33]. We believe that other mycotoxins, including trichothecenes, have the capacity of inducing cellular senescence. Actually, in our recent primary experiment, we have observed that trichothecenes, including T-2 toxin, could activate SASP and SA-β-gal and induce RAW264.7 cell senescence, implying that more mycotoxins (including modified ones) have the potential of inducing cellular senescence. Nevertheless, the underlying mechanisms in this context need to be explored. Since cellular senescence and SASP are reported to induce an immunosuppressive environment [34], we suspect that the cell senescence induced by mycotoxins plays a potential role in the immunosuppressive effects (Figure 1). Understanding the function of cellular senescence in immunotoxicity helps to further explore the toxic target of mycotoxins. In addition, whether hypoxia and HIF are involved in this potential mechanism is not yet fully understood, but it should be noted that HIF is a master regulator of the immune evasion process. Other scientific questions, including how mycotoxins induce hypoxia and HIF and what is the molecular mechanism by which cellular senescence regulates immunosuppression, need to be answered in the near future.
Therefore, under these circumstances, we are pleased to have this opportunity to compile a Special Issue entitled “Hypoxia, Cellular Senescence, and Immunosuppression: Emerging Toxic Mechanisms of Mycotoxins and Toxins” for Toxins (https://www.mdpi.com/journal/toxins/special_issues/31W44JMZ11, accessed on 16 December 2022). The aim of this Special Issue is to assemble reviews and original research articles on mycotoxins and toxins, including their new toxic mechanisms and signaling and especially on hypoxia, cell senescence, and immunosuppressive effects. We welcome researchers to submit their relevant research articles and excellent work on mycotoxins and toxins to this Special Issue with the aim of sharing the latest ideas and progresses in this field with the readers of Toxins.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Alvito, P.; Pereira-da-Silva, L. Mycotoxin Exposure during the First 1000 Days of Life and Its Impact on Children’s Health: A Clinical Overview. Toxins 2022, 14, 189. [Google Scholar] [CrossRef] [PubMed]
  2. Deng, Y.; You, L.; Nepovimova, E.; Wang, X.; Musilek, K.; Wu, Q.; Wu, W.; Kuca, K. Biomarkers of deoxynivalenol (DON) and its modified form DON-3-glucoside (DON-3G) in humans. Trends Food Sci. Technol. 2021, 110, 551–558. [Google Scholar] [CrossRef]
  3. Kraft, S.; Buchenauer, L.; Polte, T. Mold, Mycotoxins and a Dysregulated Immune System: A Combination of Concern? Int. J. Mol. Sci. 2021, 22, 2269. [Google Scholar] [CrossRef]
  4. Liu, N.; Yang, Y.; Chen, J.; Jia, H.; Zhang, Y.; Jiang, D.; Wu, G.; Wu, Z. 3-Acetyldeoxynivalenol induces lysosomal membrane permeabilization-mediated apoptosis and inhibits autophagic flux in macrophages. Environ. Pollut. 2020, 265, 114697. [Google Scholar] [CrossRef] [PubMed]
  5. Gao, Y.N.; Wang, Z.W.; Yang, X.; Wang, J.Q.; Zheng, N. Aflatoxin M1 and ochratoxin A induce a competitive endogenous RNA regulatory network of intestinal immunosuppression by whole-transcriptome analysis. Sci. Total Environ. 2022, 854, 158777. [Google Scholar] [CrossRef]
  6. Cai, G.; Zhong, F.; Cao, Q.; Bai, Y.; Zou, H.; Gu, J.; Yuan, Y.; Zhu, G.; Liu, Z.; Bian, J. ZEA and DON inhibited inflammation after L. monocytogenes infection and induced ribosomal hyperfunction. Ecotoxicol. Environ. Saf. 2022, 236, 113470. [Google Scholar] [CrossRef]
  7. Liu, D.; Wang, Q.; He, W.; Ge, L.; Huang, K. Deoxynivalenol aggravates the immunosuppression in piglets and PAMs under the condition of PEDV infection through inhibiting TLR4/NLRP3 signaling pathway. Ecotoxicol. Environ. Saf. 2022, 231, 113209. [Google Scholar] [CrossRef]
  8. Xu, X.; Nicholson, P.; Ritieni, A. Effects of fungal interactions among Fusarium head blight pathogens on disease development and mycotoxin accumulation. Int. J. Food Microbiol. 2007, 119, 67–71. [Google Scholar] [CrossRef]
  9. Antonissen, G.; Martel, A.; Pasmans, F.; Ducatelle, R.; Verbrugghe, E.; Vandenbroucke, V.; Li, S.; Haesebrouck, F.; Van Immerseel, F.; Croubels, S. The impact of Fusarium mycotoxins on human and animal host susceptibility to infectious diseases. Toxins 2014, 6, 430–452. [Google Scholar] [CrossRef] [Green Version]
  10. Zhou, H.R.; Jia, Q.; Pestka, J.J. Ribotoxic stress response to the trichothecene deoxynivalenol in the macrophage involves the SRC family kinase Hck. Toxicol. Sci. 2005, 85, 916–926. [Google Scholar] [CrossRef]
  11. Zhou, H.R.; Lau, A.S.; Pestka, J.J. Role of double-stranded RNA-activated protein kinase R (PKR) in deoxynivalenol-induced ribotoxic stress response. Toxicol. Sci. 2003, 74, 335–344. [Google Scholar] [CrossRef] [Green Version]
  12. Sun, Y.; Huang, K.; Long, M.; Yang, S.; Zhang, Y. An update on immunotoxicity and mechanisms of action of six environmental mycotoxins. Food Chem. Toxicol. 2022, 163, 112895. [Google Scholar] [CrossRef]
  13. Liu, X.; Wang, Z.; Wang, X.; Yan, X.; He, Q.; Liu, S.; Ye, M.; Li, X.; Yuan, Z.; Wu, J.; et al. Involvement of endoplasmic reticulum stress-activated PERK-eIF2α-ATF4 signaling pathway in T-2 toxin-induced apoptosis of porcine renal epithelial cells. Toxicol. Appl. Pharmacol. 2021, 432, 115753. [Google Scholar] [CrossRef]
  14. Xiao, Y.; Wang, J.; Wang, J.; Wang, H.; Wu, S.; Bao, W. Analysis of the roles of the Notch1 signalling pathway in modulating deoxynivalenol cytotoxicity. Ecotoxicol. Environ. Saf. 2022, 246, 114183. [Google Scholar] [CrossRef] [PubMed]
  15. Wu, Q.; Wu, W.; Franca, T.C.C.; Jacevic, V.; Wang, X.; Kuca, K. Immune Evasion, a Potential Mechanism of Trichothecenes: New Insights into Negative Immune Regulations. Int. J. Mol. Sci. 2018, 19, 3307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. You, L.; Wang, X.; Wu, W.; Nepovimova, E.; Wu, Q.; Kuca, K. HIF-1α inhibits T-2 toxin-mediated "immune evasion" process by negatively regulating PD-1/PD-L1. Toxicology 2022, 480, 153324. [Google Scholar] [CrossRef]
  17. You, L.; Zhao, Y.; Kuca, K.; Wang, X.; Oleksak, P.; Chrienova, Z.; Nepovimova, E.; Jaćević, V.; Wu, Q.; Wu, W. Hypoxia, oxidative stress, and immune evasion: A trinity of the trichothecenes T-2 toxin and deoxynivalenol (DON). Arch. Toxicol. 2021, 95, 1899–1915. [Google Scholar] [CrossRef] [PubMed]
  18. Wu, Q.; Wu, W.; Kuca, K. From hypoxia and hypoxia-inducible factors (HIF) to oxidative stress: A new understanding of the toxic mechanism of mycotoxins. Food Chem. Toxicol. 2020, 135, 110968. [Google Scholar] [CrossRef] [PubMed]
  19. Habrowska-Górczyńska, D.E.; Kowalska, K.; Urbanek, K.A.; Domińska, K.; Sakowicz, A.; Piastowska-Ciesielska, A.W. Deoxynivalenol modulates the viability, ROS production and apoptosis in prostate cancer cells. Toxins 2019, 11, 265. [Google Scholar] [CrossRef] [Green Version]
  20. Tian, J.; Yan, J.; Wang, W.; Zhong, N.; Tian, L.; Sun, J.; Min, Z.; Ma, J.; Lu, S. T-2 toxin enhances catabolic activity of hypertrophic chondrocytes through ROS-NF-κB-HIF-2α pathway. Toxicol. In Vitro 2012, 26, 1106–1113. [Google Scholar] [CrossRef]
  21. Raghubeer, S.; Nagiah, S.; Chuturgoon, A. Ochratoxin A upregulates biomarkers associated with hypoxia and transformation in human kidney cells. Toxicol. In Vitro 2019, 57, 211–216. [Google Scholar] [CrossRef]
  22. Pyo, M.C.; Choi, I.G.; Lee, K.W. Transcriptome analysis reveals the AhR, Smad2/3, and HIF-1α pathways as the mechanism of ochratoxin A toxicity in kidney cells. Toxins 2021, 13, 190. [Google Scholar] [CrossRef]
  23. Chen, R.; Lai, U.H.; Zhu, L.; Singh, A.; Ahmed, M.; Forsyth, N.R. Reactive oxygen species formation in the brain at different oxygen levels: The role of hypoxia inducible factors. Front. Cell Dev. Biol. 2018, 6, 132. [Google Scholar] [CrossRef] [Green Version]
  24. Cohn, R.L.; Gasek, N.S.; Kuchel, G.A.; Xu, M. The heterogeneity of cellular senescence: Insights at the single-cell level. Trends Cell Biol. 2022. [Google Scholar] [CrossRef]
  25. Calcinotto, A.; Kohli, J.; Zagato, E.; Pellegrini, L.; Demaria, M.; Alimonti, A. Cellular senescence: Aging, cancer, and injury. Physiol. Rev. 2019, 99, 1047–1078. [Google Scholar] [CrossRef]
  26. Gorgoulis, V.; Adams, P.D.; Alimonti, A.; Bennett, D.C.; Bischof, O.; Bishop, C.; Campisi, J.; Collado, M.; Evangelou, K.; Ferbeyre, G.; et al. Cellular senescence: Defining a path forward. Cell 2019, 179, 813–827. [Google Scholar] [CrossRef]
  27. Huang, C.W.; Liao, W.R.; How, C.M.; Yen, P.L.; Wei, C.C. Chronic exposure of zearalenone inhibits antioxidant defense and results in aging-related defects associated with DAF-16/FOXO in Caenorhabditis elegans. Environ. Pollut. 2021, 285, 117233. [Google Scholar] [CrossRef]
  28. Chao, H.; Ma, H.; Sun, J.; Yuan, S.; Dong, P.; Zhao, A.; Li, L.; Shen, W.; Zhang, X. Whole-transcriptome analysis of non-coding RNA alteration in porcine alveolar macrophage exposed to aflatoxin B1. Toxins 2022, 14, 373. [Google Scholar] [CrossRef]
  29. Yang, X.; Liu, S.; Huang, C.; Wang, H.; Luo, Y.; Xu, W.; Huang, K. Ochratoxin A induced premature senescence in human renal proximal tubular cells. Toxicology 2017, 382, 75–83. [Google Scholar] [CrossRef]
  30. Dubourg, V.; Nolze, A.; Kopf, M.; Gekle, M.; Schwerdt, G. Weighted correlation network analysis reveals CDK2 as a regulator of a ubiquitous environmental toxin-induced cell-cycle arrest. Cells 2020, 9, 143. [Google Scholar] [CrossRef]
  31. Solhaug, A.; Holme, J.A.; Haglund, K.; Dendele, B.; Sergent, O.; Pestka, J.; Lagadic-Gossmann, D.; Eriksen, G.S. Alternariol induces abnormal nuclear morphology and cell cycle arrest in murine RAW 264.7 macrophages. Toxicol. Lett. 2013, 219, 8–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Solhaug, A.; Eriksen, G.S.; Holme, J.A. Mechanisms of action and toxicity of the mycotoxin alternariol: A review. Basic Clin. Pharmacol. Toxicol. 2016, 119, 533–539. [Google Scholar] [CrossRef] [PubMed]
  33. Li, H.; Guan, K.; Zuo, Z.; Wang, F.; Peng, X.; Fang, J.; Cui, H.; Zhou, Y.; Ouyang, P.; Su, G.; et al. Effects of aflatoxin B(1) on the cell cycle distribution of splenocytes in chickens. J. Toxicol. Pathol. 2019, 32, 27–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Toso, A.; Revandkar, A.; Di Mitri, D.; Guccini, I.; Proietti, M.; Sarti, M.; Pinton, S.; Zhang, J.; Kalathur, M.; Civenni, G.; et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 2014, 9, 75–89. [Google Scholar] [CrossRef]
Figure 1. Mycotoxins, hypoxia, cellular senescence, and immunosuppression.
Figure 1. Mycotoxins, hypoxia, cellular senescence, and immunosuppression.
Toxins 14 00880 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wu, Q.; You, L.; Wu, W.; Wang, X. Hypoxia and Cellular Senescence, Emerging Toxic Mechanisms of Mycotoxins and Toxins: A New Understanding of the Negative Immune Regulations. Toxins 2022, 14, 880. https://doi.org/10.3390/toxins14120880

AMA Style

Wu Q, You L, Wu W, Wang X. Hypoxia and Cellular Senescence, Emerging Toxic Mechanisms of Mycotoxins and Toxins: A New Understanding of the Negative Immune Regulations. Toxins. 2022; 14(12):880. https://doi.org/10.3390/toxins14120880

Chicago/Turabian Style

Wu, Qinghua, Li You, Wenda Wu, and Xu Wang. 2022. "Hypoxia and Cellular Senescence, Emerging Toxic Mechanisms of Mycotoxins and Toxins: A New Understanding of the Negative Immune Regulations" Toxins 14, no. 12: 880. https://doi.org/10.3390/toxins14120880

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop