Next Article in Journal
Effects of Nitrogen Fertilization on Synthesis of Primary and Secondary Metabolites in Three Varieties of Kacip Fatimah (Labisia Pumila Blume)
Previous Article in Journal
A Comparative Reverse Docking Strategy to Identify Potential Antineoplastic Targets of Tea Functional Components and Binding Mode
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechanisms of Mycotoxin-Induced Neurotoxicity through Oxidative Stress-Associated Pathways

1
Nippon Institute for Biological Science, 9-2221-1, Shin-Machi, Ome, Tokyo 198-0024, Japan
2
Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo, Tokyo 113-8657, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2011, 12(8), 5213-5237; https://doi.org/10.3390/ijms12085213
Submission received: 7 June 2011 / Revised: 21 July 2011 / Accepted: 4 August 2011 / Published: 15 August 2011
(This article belongs to the Section Molecular Toxicology)

Abstract

:
Among many mycotoxins, T-2 toxin, macrocyclic trichothecenes, fumonisin B1 (FB1) and ochratochin A (OTA) are known to have the potential to induce neurotoxicity in rodent models. T-2 toxin induces neuronal cell apoptosis in the fetal and adult brain. Macrocyclic trichothecenes bring about neuronal cell apoptosis and inflammation in the olfactory epithelium and olfactory bulb. FB1 induces neuronal degeneration in the cerebral cortex, concurrent with disruption of de novo ceramide synthesis. OTA causes acute depletion of striatal dopamine and its metabolites, accompanying evidence of neuronal cell apoptosis in the substantia nigra, striatum and hippocampus. This paper reviews the mechanisms of neurotoxicity induced by these mycotoxins especially from the viewpoint of oxidative stress-associated pathways.

1. Introduction

Mycotoxins are fungal metabolites known to be harmful toward human and animal health. To date, disorders caused by mycotoxins have been reported in digestive, urinary, immune and reproduction systems [1], and many in vivo and in vitro studies have been performed in order to clarify the mechanisms of mycotoxin-induced toxicity in these systems. Recently, Surai et al. [2] described that, in many cases, membrane-active properties of various mycotoxins determine their toxicity and incorporation of mycotoxins into membrane structures causes various detrimental changes, resulting in alterations in second messenger systems through damaging membrane receptors. In addition, detrimental effects of mycotoxins on DNA and RNA and protein synthesis together with proapoptotic action further compromise important metabolic pathways and consequently changes in physiological functions including growth, development and reproduction occur. During the last decades, the importance of oxidative stress and lipid peroxidation in all these processes have been pointed out by many researchers [2,3].
Compared with the amount of research on digestive, urinary, immune and reproduction systems, there are few reports of the effects of mycotoxins on neuronal tissues. This paper reviews the mechanisms of neurotoxicity experimentally induced in rats and mice by T-2 toxin, macrocyclic trichothecenes, fumonisin B1 (FB1) and ochratoxin A (OTA) especially from the viewpoint of oxidative stress-associated pathways.

2. T-2 Toxin

T-2 toxin is a cytotoxic secondary fungal metabolite that belongs to thetrichothecene mycotoxin family. They are produced by various species of Fusarium (F. sporotichioides, F. poae, F. equiseti, and F. acuminatum), which can infect corn, wheat, barley and rice crops in the field or during storage [4,5]. T-2 toxin is conjectured to be a major factor in alimentary toxic aleukia in humans [6] and has been implicated in additional mycotoxicoses such as red mold disease in humans and animals [7] and beanhull poisoning in horses [8].
T-2 toxin is a well-known inhibitor of protein synthesis through its high binding affinity to peptidyl transferase which is an integral part of the 60 s ribosomal subunit [911]. Subsequent inhibition of the peptidyl transferase reaction can trigger a ribotoxic stress response that activates c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinases (MAPKs) [11]. Moreover, T-2 toxin interferes with the metabolism of membrane phospholipids and increases liver lipid peroxides [12,13].
Oral, parenteral and cutaneous exposures to T-2 toxin induce lesions in hematopoietic, lymphoid and gastrointestinal tissues and suppress reproductive functions in domestic and laboratory animals [1417]. T-2 toxin can induce apoptosis in many types of cells bearing rapid rates of proliferation [1822]. T-2 toxin also induces apoptosis and fatty change in hepatocytes of mice following the increased expression of both oxidative stress- and apoptosis-related genes (c-fos and c-jun) [23]. Moreover, prenatal exposure of rats to T-2 toxin induces apoptosis in maternal liver, placenta and fetal liver following the increased expression of oxidative stress- and apoptosis-related genes and decreased expression of lipid metabolism- and drug-metabolizing enzyme-related genes in these tissues [24]. Doi et al. [25,26] have reviewed the mode of occurrence and mechanisms of T-2 toxin-induced apoptosis in mice and rats.
To date, the effects of T-2 toxin on the central nervous system (CNS) have received limited attention [27], and therefore, there are only a small number of reports of T-2 toxin-induced neurotoxicity [2830]. Boyd et al. [28] reported that low levels of T-2 toxin were responsible for the changes in the metabolism of brain biogenic monoamines, and Wang et al. [31] showed that ingestion of T-2 toxin leads to changes in amino acid permeability across the blood-brain barrier, which could lead to neurological effects observed in animals exposed to trichothecenes. T-2 toxin may be easily distributed to the fetal brain, and induce fetal death and fetotoxicity mainly in the CNS and skeletal system in addition to maternal toxicity [3235].
Sehata et al. [36] have investigated the mechanisms of apoptosis induction in the fetal brain by oral administration of T-2 toxin (2 mg/kg b.w.) to pregnant rats on day 13 of gestation. In their study, the number of apoptotic neural progenitor cells in the telencephalon increased from 1 h and peaked at 12 h after T-2 toxin treatment. Microarray analysis revealed that the expression of heat shock protein 70 (HSP70), metallothionein (MT)-2 and 1, and heme oxygenase-1 (HO-1) was strongly elevated by T-2 toxin at 12 h, and the expression of the Cu, Zn-superoxide dismutase (Cu, Zn-SOD) gene also increased at 24 h after T-2 toxin treatment. This suggests that oxidative stress might be the main factor behind the T-2 toxin-induced changes in the fetal brain. In addition, the gene expression of liver stearyl-CoA desaturase and farnesyl diphosphate synthase genes which are involved in lipid metabolism was suppressed by T-2 toxin in the fetal brain [36].
T-2 toxin suppresses drug metabolizing enzymes such as glutathione S-transferases (GSTs) [3638]. In addition, a decreased expression in mitochondria-related genes, such as mitochondrial NADH-dehydrogenase and cytochrome oxidase, has been reported in the fetal brain [36], suggesting a dysfunction of the mitochondria. Since mitochondria play an important role in cell survival, these changes in metabolism-related genes may also have a relationship to the induction of apoptosis.
In the study by Sehata et al. [36] on the fetal brain, the expression of MEKK1 gene increased at 12 and 24 h, and the expression of c-jun gene at 24 h after T-2 toxin treatment. These findings suggest that the MAPK-JNK-c-jun pathway might be involved in T-2 toxin-induced apoptosis in the fetal brain. Extracellular signal-related protein kinase (ERK) mediates cell growth and protects cells from apoptosis, whereas stress-activated protein kinase (SAPK)/JNK and p38 MAPK inhibit cell proliferation and may promote apoptosis [39]. Each MAPK is activated by an upstream MAPK kinase, including MEKK1, and JNK activates transcription factors such as c-fos and c-jun. MEKK1 may induce apoptosis by causing a general deregulation of MAPK signaling [40], and JNK and c-jun are important regulators of apoptosis in the nervous system [41].
Differing from the results in the maternal liver, placenta and fetal liver [24], the increase in caspase-2 gene expression with no changes in caspase-9 and Bax-α gene expression was detected in the fetal brain at 24 h after T-2 toxin treatment [36], suggesting an involvement of caspase-2 activation in T-2 toxin-induced apoptosis in the fetal brain. Activation of caspase-2 is induced by reactive oxygen species (ROS), and caspase-2 is said to play a crucial role in the control of apoptosis [4244]. Although it is suggested that the p53-related mitochondrial pathway is involved in the T-2 toxin-induced apoptosis in the maternal and fetal livers [24], apoptosis induction in the fetal brain by T-2 toxin seems to be independent of the p53-related pathway which is the most important pathway in DNA-damaging agent-induced apoptosis of neural progenitor cells in the developing brain [4549].
In microarray analysis on the fetal rat brain from dams exposed to T-2 toxin, the expression of vascular endothelial growth factor (VEGF) gene increased at 12 and 24 h after T-2 toxin treatment [36]. VEGF is expressed in neurons and may play a role in the maintenance of neurons and endothelial cells in the CNS [50]. Therefore, the observed VEGF induction in the fetal brain might indicate a protective reaction to the apoptotic changes in the fetal brain induced by T-2 toxin.
Recently, Chaudhary and Rao [27] evaluated acute toxicity of dermal and subcutaneous exposure of T-2 toxin on brain oxidative stress in adult mice. Mice were exposed to LD50 of T-2 toxin either by the dermal (5.94 mg/kg b.w.) or subcutaneous (1.54 mg/kg b.w.) route and sacrificed at 1, 3 and 7 days post-exposure. They reported that T-2 toxin-treated animals showed a time-dependent increase in ROS generation, glutathione (GSH) depletion, lipid peroxidation and protein carbonyl content in the brain in both routes of exposure. The gene expression profile of antioxidant enzymes showed a significant increase in SOD and catalase via the percutaneous route and glutathione reductase (GR) and glutathione peroxidase (GPx) via the subcutaneous route. This indicates that T-2 toxin induces oxidative damage in adult mouse brain as well as in fetal rat brain. Lipid peroxidation may bring about protein damage and inactivation of membrane-bound enzyme either through direct attack by free radicals or through chemical modification by its end products [51]. Indeed, as mentioned above, protein carbonylation, a sign of oxidative damage, significantly increased in the mouse brain after exposure to T-2 toxin.
Chaudhary and Roa [27] also investigated the role of nuclear factor erythroid 2-related factor (Nrf2) and its downstream targets of phase II antioxidant/detoxifying enzymes in the mouse brain exposed to T-2 toxin. Upon activation, Nrf2 binds to antioxidant responsive element sites in the promoter regions of many detoxification and antioxidant genes, leading to coordinate regulation of downstream targets that boost the cellular detoxification process and antioxidant potential [52,53]. In the study of Chaudhary and Roa [27], however, Nrf2 and its downstream target genes were down-regulated, and the involvement of Nrf2 in augmenting oxidative potential is not significant, although there is oxidative stress.

3. Macrocyclic Trichothecenes

The fungus Stachybotrys chartarum, a saprophyte that grows on wet cellulose-containing building materials including wallboard, ceiling tiles and cardboard is often found in low concentrations among the mycoflora identified in water-damaged buildings [5457]. Chronic indoor exposures to S. chartarum and its products or components have been postulated to etiologically contribute to damp building-related illnesses (DBRI) such as debilitating respiratory [58,59] and nonrespiratory symptoms involving immune and neurological impairment [56,6062]. Experimental rodent studies revealed that, while this fungus is not infectious, airway exposure to spores of S. chartarum and its components have the potential to evoke toxicity, inflammation and allergic sensitization in the upper and lower respiratory tracts [56].
Two toxic “chemotypes” of S. chartarum exist. One chemotype elaborates highly toxic macrocyclic trichotecene mycotoxins whereas a second chemotype produces less toxic atranones and simple trichothecenes but no macrocyclic trichothecenes [63]. The former mycotoxins are potent translational inhibitors and stress kinase activators that appear to be a critical underlying cause for a number of adverse effects. Notably, these toxins form covalent protein adducts in vitro and in vivo and, furthermore, cause neurotoxicity and inflammation in the nose and brain of the mouse [64,65]. On the other hand, the latter mycotoxins can induce pulmonary inflammation. Pestka et al. [56] have reviewed the relationship between S. chartarum, trichothecene mycotoxins, and DBRI and proposed new insights into a public health enigma.
Besides inhibiting translation, macrocyclic trichothecenes as well as other trichothecenes can simultaneously activate p38, JNK and ERK and MAPKs in vivo and in vitro [6668] via a process referred to as “ribotoxic stress” [69]. In cell cultures, macrocyclic trichothecenes (Type D trichotecenes) are 10–100 times more potent than Type A (e.g., T-2 toxin) or Type B (e.g., deoxynivalenol) trichothecenes at activating MAPKs, impairing leukocyte proliferation, or inducing apoptosis [11,7075]. The common ability of macrocyclic trichothecenes to cause protein synthesis inhibition via binding to the 18S rRNA of the ribosomal large subunit [76] has been speculated to be a major mechanism underlying induction of cell apoptosis by this group of trichothecenes. Moreover, the potential of macrocyclic trichothecenes to covalently bind to proteins and possibly other macromolecules has major implications relative to their absorption, metabolism, distribution, toxicity, and potential allergenicity [64,65].
Satratoxin G (SG) is one of the most potent macrocyclic trichothecenes produced by S. chartarum [75] and contributes to the above-mentioned DBRI. Roridin A (RA) is a commercially available macrocyclic trichithecene used as a SG surrogate, and roridin L2 (RL2) is a putative biosynthetic precursor of SG. While SG contains an intact macrocyclic ring linking C-4 to C-15, the precursor RL2 contains only an extended carbon chain linked at C-4 [77]. RL2 is said to be nontoxic [78]. Satratoxin H (SH) is another macrocyclic trichothecene mycotoxin derived from the fungus S. chartaum. This mycotoxin is one of the toxic constituents of the toxic mushroom, Podostoma cornu-damae [79].
Murine alveolar type II cells and alveolar macrophages are extremely sensitive to intratracheally instilled S. chartarum spores [80]. The methanol extract of a trichothecene-producing strain of S. chartarum particularly up-regulates DNA damage-responsive and DNA repair genes in the murine alveolar macrophage cell line MH-S early in the treatment, which are suggestive of genotoxic stress [81]. In a follow-up study, extract-induced apoptosis in MH-S cells was observed to precede DNA damage [82]. Moreover, both p38- and p53-mediated signaling events seem to occur in S. chartarum-induced apoptosis of alveolar macrophages.
Macrocyclic tricothecenes also affect the upper respiratory tract (e.g., nasal airway). Using an intranasal instillation model in adult C57BL/6J mice, Islam et al. [83] showed that SG exposure specifically induced apoptosis of the olfactory sensory neurons (OSNs) and subsequent atrophy of the olfactory epithelium (OE). Concurrently, there was bilateral atrophy of the olfactory nerve layer of the olfactory bulbs (OBs) of the brain. In addition, SG induced an acute, neutrophilic rhinitis and encephalitis. Similar findings have also been reported in mice intranasally instilled with RA [84]. In the ethmoid turbinates and OBs in the frontal brain in mice treated with SG, elevated mRNA expression for the proinflammatory cytokines, TNF-α, IL-6 and IL-1, the chemokine macrophage-inflammatory protein-2 (MIP-2), and the proapoptotic genes, Fas, FasL, p75NGFR, p53, Bax, caspase-3 and CAD, was detected at 24 h post instillation (PI). In the same regions of mice treated with RA, up-regulated mRNA expression of Fas, TNF-α, IL-6 and IL-1 and MIP-2 was observed from 6 to 24 h PI, whereas expression of several other proapoptotic genes (p53, Bax, and caspase-activated DNase) was detectable only at 24 h PI.
Following intranasal instillation of mice to SG [83] or RA [84], double-stranded RNA-activated protein kinase (PKR) mRNA concentrations in the nasal turbinates were up-regulated in parallel with OSN apoptosis. PKR, Bax and p53 have been previously reported to mediate apoptosis in murine OSNs [8587]. PKR associates with the ribosome [88] and can selectively shut down translation via phosphorylation of eukaryolic initiation factor 2α (eIF2α) as well as activate NF-κB [89].
Both TNF-α and Fas directly induce apoptosis in OE [90] and in OE organ cultures [91,92], and SG- and RA-induced TNF-α and Fas mRNA expression precedes or is concurrent with OSN apoptosis, induction of caspase-3 mRNA and caspase-3 activation [83,84]. The origins of induced TNF-α are considered to be OSNs and adjacent cells in the OE which would promote autocrine or paracrine responses, respectively. SG and RA also directly induces apoptosis in the OSNs by initiating mitochondrial cell death via an intrinsic pathway involving p53 and Bax. Islam et al. [93] used the PC12 rat pheochromocytoma cell models to elucidate potential mechanisms of SG-induced neuronal cell death. In their experiment, SG-induced apoptosis occurred at 48 h after SG treatment, and the expression of p53, and PKR, Bax and caspase-activated DNase mRNAs was significantly elevated from 6 to 48 h after SG treatment. SG-induced p53 and Bax gene expression is known to drive nuclear translocation of apoptosis-inducing factor (AIF), mitochondrial flavoprotein, in PC-12 cells [93].
In the study by Islam et al. [93], SG-induced apoptosis was not affected by inhibitors of oxidative stress or MAPKs but was suppressed by the PKR inhibitor C16 and by PKR siRNA transfection. PKR inhibition also blocked SG-induced apoptotic gene expression and AIF translocation but not caspase-3 activation. These results indicate that SG-induced apoptosis in PC12 neural cells is mediated by PKR via a caspase-independent pathway possibly involving AIF translocation.
SH is thought to induce caspase-3 activation and apoptosis of PC12 cells through the activation of p38 MAPK and JNK in a GSH-sensitive manner [94]. Moreover, Nusuetrong et al. [79] carried out the study to further elucidate the mechanisms by which SH induces cell death in PC12 cells. They reported that SH causes apoptosis of serum-deprived PC12 cells within 24 h and that SH increases ROS production and lipid peroxidation which are attenuated by incubation of cells with GSH. They suggested that SH-induced increase in apoptosis of serum-deprived PC12 cells, may be partially mediated through the generation of ROS. GSH, the most abundant intracellular thiol, plays an important role in controlling the redox state of cells, and GSH is thought to play a role in apoptotic cell death following its efflux through the GSH-specific membrane channels, carrier-mediated GSH extrusion and oxidative stress [79,95].
The constant activation of inflammatory and apoptotic pathways at low levels of exposure in human neurological system cells may amplify devastation to neurological tissues and lead to neurological system cell damage from indirect events triggered by the presence of SH [96]. This suggests that individuals exposed to SH and microbial organisms resulting in a chronic immune response (inflammation and oxidative stress) could have increased sensitivity to these agents, leading to neural damage, further supporting previous in vivo studies demonstrating CNS tissue damage via inhalation of fungal toxins [97]. The process of inflammation is intended to repair injured tissues; however, this mechanism tends to induce damage to nervous tissues when activated [98,99]. In this context, studies on canines by Caldeón-Garcidueñas et al. [100,101] demonstrated increased iNOS, NF-κB, and TNF-α production among other inflammatory and oxidative stress agents—leading to permanent damage of DNA and CNS tissues due to passage of small particles via olfactory epithelium and lung tissue. The fine particles reach CNS tissue via the olfactory bulb and into brain tissue (frontal cortex and cortical tissues) demonstrating increases in β-amyloid plaques suggestive of pathogenesis similar to Alzheimer’s disease [100,101].

4. Fumonisin B1

Fumonisins belong to the relatively recently discovered group of mycotoxins produced by the fungus Fusarium verticillioides (formerly F. moniliforme), a widespread fungal concomitant of various cereals, predominantly corn [2,102]. In this case, FB1 is the most abundant and toxic; it has been linked to a number of diseases in humans and animals [102,103].
The structures of FB1 and sphingolipids show marked similarities, which may be the reason why FB1 drastically disrupts the normal sphingolipid metabolism [104]. FB1 inhibits ceramide synthase, a key enzyme in de novo sphingolipid biosynthesis and sphingolipid turnover, causing elevated levels of free sphingolipid bases and sphingolipid base metabolites and lowered levels of ceramide [105,106]. FB1-induced inhibition of ceramide synthesis can result in a wide spectrum of changes in lipid metabolism and associated lipid-depending signaling pathways, and it appears to be a major contributor to the carcinogenic and other deleterious effects of FB1 [106].
FB1 is well known to cause equine leukoencephalomalacia (ELEM) [107,108]. This disease is associated primarily with FB1 and is characterized by high mortality [108]. In histopathological examinations, pathognomonic focal necrotic lesions, located primarily in the subcortical white matter are apparent [102]. In addition, the elevation of free sphingoid bases after FB1 treatment has been demonstrated in the brain of ELEM-diagnosed horses [109], suggesting that free sphingoid bases may be important in FB1-related neurotoxicity.
Another emerging neurodevelopmental aspect of FB1 toxicity that implicates the consumption of fumonisins in the etiology of neuronal tubule defects (NTD) in children has recently been suggested [110]. Treatment with FB1 causes NTDs in ex vivo neurulating mouse embryos [111] and this effect is related to the folic acid receptor deficiency as a result of the FB1-dependent lipid rafts depletion [112].
The detrimental effects of FB1 on neuronal tissue have been shown in a number of reports indicating its potential for direct neurotoxicity. For example, FB1 drastically inhibits axonal growth in cultured hippocampal neurons [113], increases levels of sphinganine concentration in the forebrain and brain stem of rats accompanying a concomitant demyelination in the forebrain [114], and disrupts myelination in glial cells but not neurons in aggregating brain cell culture [115] and in developing rats [116]. FB1-dependent changes in neurotransmitter metabolite levels in different brain regions of BALB/c mice [117] and in rat brain [118], and alteration of electrophysiological activity in rat neocortex [199] are also reported.
Osuchowski et al. [120] carried out a study to compare the toxicity of FB1 in mouse brain after an intracerebroventricular (icv) or subcutaneous (sc) infusion with total doses of 0, 10 or 100 μg/kg of FB1. The icv infusion of FB1 led to neuronal degeneration in the cortex, concurrent with disruption of sphingolipid metabolism, i.e., inhibition of de novo ceramide synthesis, stimulation of astrocytes, and activation of proinflammatory cytokine signaling while the sc infusion of FB1 brought about partial inhibition of sphingolipid metabolism in the cortex. From these results and the reports showing that FB1 compromises the endothelial barrier function [121,122], it is suggested that there may be limited blood-brain barrier transfer of FB1 [123] and that FB1 may disrupt central nervous system homeostasis when brain tissue is directly exposed to this mycotoxin.
All cytokines analyzed in the study of Osuchowski et al. [120] are CNS-borne and are expressed on-site by neurons (IL-1β and IL-6), astrocytes (IL-1β, IL-6, TNF-α and interferon-γ; IFN-γ) and microglia (IL-1β, IL-6 and TNF-α) [124]. IL-1β, IL-6 and TNF-α are primarily associated with neuronal injury; thus neuronal damage will be accompanied by their elevated expression. In addition, the increased expression of TNF-α and IL-1β also indirectly enhance neuronal damage via ceramide-mediated signaling, since both cytokines activate brain neutral sphingomyelinase (nSMase), and the role of ceramide-dependent neurodegeneration mediated by nSMase is reported [125,126]. The immunocompetent cells and proinflammatory signaling are first being activated (i.e., astrocytes and probably microglia) and then neurodegeneration follows in mouse brain after FB1 infusion [120].
During the last decades, studies aimed at clarifying the mechanisms of FB1-induced neurotoxicity in cultured cells has been done mainly from the viewpoints of oxidative stress and/or apoptosis. Stockmann-Juvala et al. [127] tried to characterize oxidative stress-related parameters induced by FB1 in three different neural cell lines, human SH-SY5Y neuroblastoma, rat C6 glioblastoma and mouse GT1-7 hypothalamic cells. In their study, FB1 caused a dose-dependent increase of ROS production in C6 and GT1-7 cells but was without an effect in SH-SY5Y cells. Decreased GSH levels, increased malon dialdehyde (MDA)-formation, indicative of lipid peroxidation, and necrotic cell death were observed in all cell lines after incubation with FB1. From these results, they concluded that FB1 induces oxidative stress in human, rat and mouse neural cell cultures. They also suggested that FB1 is cytotoxic to neural cells only at high concentrations in vitro, although systemic toxicity, which may be caused by the inhibition of ceramide synthase, takes place already at very low concentrations of FB1 [104].
Mobio et al. [128130] reported that in rat C6 glioma cells, FB1 inhibits protein synthesis, causes DNA fragmentation and cell death, increases 8-hydroxy-2′-deoxyguanosine (8-OH-dG), and induces lipid peroxidation, and that cytotoxic concentrations of FB1 induce cell cycle arrest in C6 cells (in the G2/M phase after 24 h and in G0/G1 after 48 h incubation with FB1), possibly associated with genotoxic event. On the other hand, Galvano et al. [131,132] reported that FB1 does not increase ROS production or cell death in rat astrocytes although DNA-damage and caspase-3 activation take place. Based on these results, the authors suggested that effects of FB1 are not a result of oxidative injury, but are instead a response that may occur after modulation of protective genes. These theories support the above-mentioned observations in FB1-treated SH-SY5Y cells. In SH-SY5Y cells, lipid peroxidation took place without an increase in ROS production, and was associated with delayed cell death. Moreover, low expression of the anti-apoptotic Bcl-2 protein in GT1-7 and C6 cells can be linked to low basal GSH levels in these cell lines [127], and may increase their susceptibility to radical attack [133135]. SH-SY5Y cells, on the other hand, express higher levels of Bcl-2 [133,136], which may explain why GSH levels in these cells decreased later than in GT1-7 and C6 cells exposed to FB1.
Stockmann-Juvala et al. [137] have also investigated the effects of FB1 on human U-118MG glioblastoma cells. In their study, FB1 increased lipid peroxidation and the production of ROS in U-118 MG cells dose- and time-dependently and these effects were accompanied by decreases in the GSH levels and cell viability. In addition, signs of apoptosis were indicated by increased caspase-3-like protease activity and internucleosomal DNA fragmentation. Based on these results, they concluded that oxidative stress and apoptosis may be involved in the neurotoxicity induced by FB1. There are a number of studies showing FB1-induced apoptosis in different cell types [129131,138140]. Apoptosis is considered to be a common result of oxidative stress caused by ROS production, disturbance of GSH generation and lipid peroxidation [141,142]. In addition, activation of caspase-3 may be one of the events causing an increase in ROS production, and subsequent lipid peroxidation and reduction of intracellular GSH levels.

5. Ochratoxin A

Ochratoxin A (OTA) is a fungal metabolite produced by Aspergillus ochraceus and Penicillium verrucosum. OTA is found in a variety of plant food products such as cereals. Because of its long half-life, it accumulates in the food chain [143,144] and is frequently detected in human plasma at nanomolar concentrations [145,146]. OTA has been found to be involved in the developments of certain kidney diseases [147] and enzymuria [148] similar to Balkan endemic nephropathy found in humans [147,149]. In addition to nephrotoxicity, the main OTA-induced toxicity, OTA has also been reported to have immunotoxic [150,151], teratogenic [152,153], genotoxic [154] and neurotoxic [155] effects. Although there is still insufficient evidence in humans, there is sufficient evidence in experimental animals for the carcinogenicity of OTA [156].
OTA has complex mechanisms of action that include evocation of oxidative stress, bio-energetic compromise, mitochondrial impairment, inhibition of protein synthesis, production of DNA single-strand breaks and formation of OTA-DNA adducts [155,157160]. The bio-energetic compromise induced by OTA may be responsible for the generation of free radicals and ROS that results in global oxidative damage to DNA and lipids and damage to proteins through generation of oxygen free radicals and nitric oxide [161,162]. OTA renal toxicity and carcinogenicity may be, at least partly, mediated by an Nrf2-dependent signal transduction pathway [53]. Effect of OTA on potential signaling molecules (growth factors, fatty acids, and/or Ca2+) could disrupt PKC-regulated pathways downstream, and down-regulation of genes under transcriptional control of Nrf2 may lead to a reduced oxidative stress response. In addition, OTA-induced down-regulation of genes under hepatocyte nuclear factor 4α(HNF4α)-control may affect key metabolic processes. This could make kidney cells more vulnerable to OTA-induced toxicity leading to tumor development [163]. OTA has also been reported to inhibit succinate-dependent electron transfer in the electron transport chain, but at higher concentrations will also inhibit electron transport at Complex I [164,165], suggesting mitochondrial toxicity.
The developing brain appears to be very susceptible to the deleterious effects of OTA [166169]. OTA has been shown to affect proliferation and migration of neurons [153] and reduce DNA content [170] in the developing rodent brain. OTA is also reported to be neurotoxic to adult male rats [166]. Neurotoxicity is more pronounced in the ventral mesencephalon, hippocampus (HP), and striatum than in the cerebellum (CB) [171]. Recent animal and cellular studies have suggested that OTA may contribute to the development of human and animal systemic problems, including neurodegenerative diseases and brain dysfunction [155].
Sava et al. [155] investigated the time course of acute effects of OTA in the context of DNA damage, DNA repair and global oxidative stress across six brain regions, CB, cortex (CX), HP, midbrain (MB), caudate/putamn (CP) and pons/medulla (PM), in male mice, and they showed that OTA causes acute depletion of striatal dopamine (DA) and its metabolites as well as decreased tyrosine hydroxylase immunoreactivity in the corpus striatum on a background of globally increased oxidative stress evidenced by significant increases in lipid peroxidation and oxidative DNA damage, and transient inhibition of oxidative DNA repair activity (oxyguanosine glyosylase, OGG1) across six brain regions, accompanying evidence for apoptosis in the substantia nigra (SN), striatum and HP or other regions. Unlike the monophasic SOD activation, the oxidative DNA repair response exhibited a biphasic response.
Sava et al. [172] also examined the possibility that OTA can cause parkinsonism in male mice focusing on the effects of chronic low doses of OTA exposure on regional brain oxidative stress and striatal DA metabolism. They reported that continuous administration of low doses of OTA with implanted subcutaneous Alzet minipumps caused a small but significant decrease in striatal DA levels and an up-regulation of anti-oxidant systems and DNA repair. These data suggest the possibility that low dose exposure to OTA will result in an earlier onset of parkinsonism when normal age-dependent decline in striatal DA levels is superimposed on the mycotoxin-induced lesion. In their study, since the CP and MB showed a relatively diminished OGG1 activity and increased oxidative DNA damage, it was postulated that the DA terminals of the striatum would suffer damage. This concept is supported by an earlier report of increased oxidative DNA damage in SN and striatum in post-mortem brain from Parkinson’s disease cases [173] and by the above-mentioned report that acute doses of OTA caused a dose-dependent decrease of striatal DA and a decrease in DA turnover [155]. The regional vulnerability to the toxin was not directly related to the concentration of the toxin in each region. Moreover, as mentioned above, not all regions were equally sensitive to the toxin, even though all brain regions were capable of marked increases in OGG1 activity. Namely, the CP was most sensitive to the toxin while the CB was the least sensitive. In addition, the HP, a primary site of neurodegeneration in Alzheimer’s disease, turns out to exhibit relatively high OTA levels with concurrently pronounced OTA neurotxicity [166]. OTA may also be toxic through other mechanisms. For example, due to its chemical structure, OTA inhibits protein synthesis by competition with phenylalanine in the aminoacylation reaction of phenylalanine-tRNA [174,175] and phenylalanine hydroxylase activity [157], leading to the impairment of the synthesis of DOPA, dopamine and catecholamines or enzymes involved in the metabolism of DNA.
The adult brain retains a reservoir of stem-progenitor cells in the hippocampal “neurogenic zone” capable of proliferative activity throughout life [176,177], and it is known that injury, irradiation, drugs and endogenous factors such as hormones and trophic factors impact neurogenesis [178181]. Therefore, OTA may also impact neurogenesis in adult HP. In addition, subchronic administration of OTA is demonstrated to affect cognitive functions by reducing hippocampal N-methyl-d-aspartate (NMDA) receptor subunits 2A and 2B concentrations in rats [182].
Sava et al. [183] tested neural stem/progenitor cells (NSCs) prepared from HP of adult mouse brain for their vulnerability to OTA in vitro. OTA, added to the cultures in concentrations of 0.01–100 mg/mL, caused a dose- and time-dependent (6–72 h) decrease in viability of both proliferating and differentiating NSC. Along with decreased viability, OTA elicited a pronounced oxidative stress evidenced by a robust increase in total and mitochondrial SOD activity, and OTA also significantly increased OGG1 activity. Proliferating NSC exhibited a greater vulnerability to the toxin than differentiated neurons despite robust DNA repair and antioxidant responses. Such a result is unexpected since DNA repair systems are typically more active and efficient in proliferating cells than in post-mitotic differentiated cells [184,185]. It suggests that OTA’s mechanism of action as an inhibitor of mitochondrial oxidative metabolism may be less critical than its interference with DNA synthesis and mitotic competence. Overall, Sava et al. [183] speculated that OTA exposure may contribute to impaired hippocampal neurogenesis in vivo, resulting in depression and cognitive deficits, conditions reported to be linked to mycotoxin exposure in humans [186188].
Yoon et al. [189] investigated the potential harm caused by environmental exposure to OTA in terms of its effects on neuronal cell viability and proteome profiles using mouse hippocampal HT22 and human neuroblastoma SH-SY5Y cells. Generation of ROS was detected in OTA-treated SH-SY5Y and HT22 cells, however, caspase activation and an increase in p53 phosphorylation were only detected in HT22 cells, even though OTA treatment caused oxidative stress in both two cell lines. The expressions of several proteins (valosin containing protein, propyl 4-hydroxylase, Atp5b protein, nucleophosmin 1, eukaryotic translation elongation factor 1 delta isoform, ornithine aminotransferase, prohibitin, and peroxiredoxin 6), which have been suggested to be implicated in the pathogenesis of neurodegenerative disorders, were up-regulated only in HT22 cells after treatment with OTA, which was interesting because OTA induced the apoptosis of HT22 cells but not of SH-SY5Y cells. Involvement of the mitochondrial dysfunction-related apoptotic process in OTA toxicity in HT22 cells was corroborated by the results showing significant decline in mitochondrial activity in OTA-treated HT22 cells, not in SH-SY5Y cells. Such differences between cell lines might be due in part to the complex natures of protein expression and functional regulation required during the intracellular signaling of apoptosis [190], and the above-mentioned alteration of protein expression profile in HT22 cells after OTA treatment is considered to be related to ROS generation. Because inhibition of expression of propyl 4-hydoxylase is known to attenuate neuronal death associated with oxidative stress [191], ornithine has been shown to be up-regulated during ROS-related apoptotic cell death [192], and valosin containing protein has been proposed to contribute to the conversion of oxidative stress to an endoplasmic reticulum stress response during the pathological processes of a number of neurodegenerative disorders [193]. Contrary to the above-mentioned report by Sava et al. [183], Zhang et al. [194] reported that caspase-9 and caspase-3 were activated in response to OTA treatment and caspase inhibitors were effective in partly counteracting OTA-induced apoptosis-related neurocytotoxicity not only in primary rat cortical neuronal cells but also in human neuroblastoma SHSY5Y cells and that such OTA-induced apoptosis was accompanied by a loss of mitochondria membrane potential. The authors suggest that OTA may contribute to the pathogenesis of neurodegenerative diseases (e.g., Alzheimer’s and Parkinson’s disease) in which apoptotic processes are centrally involved. The reason for the difference in apoptosis-inducing ability of OTA in human neuroblastoma SH-SY5Y cells between the two reports still remains unclear.
Besides the toxic effects of OTA on neuronal cells, Zurich et al. [195] investigated the relationship between OTA toxicity and glial reactivity in serum-free aggregating rat brain cell cultures, and they showed that OTA affects the cytoskeletal integrity of astrocytes as well as the expression of genes pertaining to the brain inflammatory response system, and suggested that a relationship exists between the inflammatory events and the cytoskeletal changes induced by OTA. Furthermore, they also suggested that, by inducing an atypical glial reactivity, OTA may severely affect the neuroprotective capacity of glial cells. Moreover, Hong et al. [196] reported that OTA caused concentration-dependent reductions in neurite outgrowth and cell number, and induced the activation of transcription factors activator protein-1 (AP-1) and NF-κB activation in cultured rat embryonic midbrain cells, and that 15-deoxy-delta 12, 14-prostaglandin J2 (15-deoxy PGJ2), a peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist, blocked OTA-induced neurotoxicity by inhibiting AP and NF-κB activation in cultured rat embryonic midbrain cells.

6. Conclusions

This paper has reviewed the mechanisms of neurotoxicity induced in rodents and neuronal cell lines by T-2 toxin, macrocyclic trichothecenes, FB1 and OTA.
T-2 toxin, one of the Type A trichothecene mycotoxins, triggers a ribotoxic response through its high binding affinity to peptidyl transferase which is an integral part of the 60 s ribosomal subunit, resulting in activation of JNK/p38 MAPKs. T-2 toxin also interferes with the metabolism of membrane phospholipids and increases liver lipid peroxides. In the fetal brain, oxidative stress is considered to be the main factor behind the T-2 toxin-induced changes, and the MAPK-JNK-c-jun pathway is thought to be involved in T-2 toxin-induced neuronal cell apoptosis. Moreover, activation of caspase-2 is essential to T-2 toxin-induced apoptosis in the fetal brain. T-2 toxin also induces oxidative damage in the adult mouse brain as well as in the fetal rat brain.
Macrocyclic trichothecenes have been postulated to etiologically contribute to DBRI such as debilitating respiratory and nonrespiratory symptoms. The common ability of macrocyclic trichothecenes to cause protein synthesis inhibition via binding to the 18s rRNA of the ribosomal large subunit has been speculated to be a major mechanism underlying induction of cell apoptosis by this group of trichothecenes. In mice, SG or RA exposure specifically induces apoptosis of OSNs and subsequent atrophy of OE. SG or RA also induces apoptosis in, and atrophy of, the olfactory nerve layer of OBs of the brain. Moreover, in the ethmoid turbinates and OBs in the frontal brain in mice treated with SG or RA, elevated mRNA expression for the inflammatory cytokines, chemokine, and proapoptotic genes and increased mRNA concentrations for PKR are detected. In PC12 neural cells, SG-induced apoptosis is mediated by PKR via a caspase-independent pathway possibly involving AIF translocation from mitochondria to the nucleus. On the other hand, SH is thought to induce caspase-3 activation and apoptosis of PC12 cells through the activation of MAPK and JNK in a GSH-sensitive manner.
FB1 is well known to cause ELEM and may be implicated in the etiology of NTD in children. FB1-induced inhibition of ceramide synthesis can result in a wide spectrum of changes in lipid metabolism and associated lipid-dependent pathways. FB1 may disrupt central nervous system homeostasis when brain tissue is directly exposed to this mycotoxin. Namely, the icv infusion of FB1 leads to neuronal degeneration in the cortex, concurrent with disruption of sphingolipid metabolism, i.e., inhibition of de novo ceramide synthesis, stimulation of astrocytes, and activation of proinflammatory cytokine signaling. In in vitro studies, FB1 inhibits protein synthesis, causes DNA fragmentation and cell death, increases 8-OH-dG, and induces lipid peroxidation in rat C6 glioma cells, and oxidative stress and apoptosis may be involved in the neurotoxicity induced in human U-118MG glioblastoma cells by FB1. On the other hand, effects of FB1 are not a result of oxidative injury but are instead a response that may occur after modulation of protective genes in rat astrocyte and SH-SY5Y human neuroblastoma cell cultures.
OTA has complex mechanisms of action that include evocation of oxidative stress, bio-energetic compromise, mitochondrial impairment, inhibition of protein synthesis, production of DNA single-strand breaks and formation of OTA-DNA adducts. OTA causes acute depletion of striatal DA and its metabolites in the corpus striatum on a background of globally increased oxidative stress across six brain regions of rats examined, accompanying evidence for apoptosis in the SN, striatum and HP or other regions. The CP is most sensitive to the toxin while the CB is the least sensitive, and the HP, primary site of neurodegeneration in Alzheimer’s disease, turns out to exhibit relatively high OTA levels with concurrently pronounced OTA neurotoxicity. OTA exposure may contribute to impaired hippocampal neurogenesis in vivo, resulting in depression and cognitive deficits. OTA also induces oxidative stress and apoptosis through caspase activation and increase in p53 phosphorylation in various neural cell cultures. OTA seems to contribute to the pathogenesis of neurodegenerative diseases in humans (e.g., Alzheimer’s and Parkinson’s disease), in which apoptotic processes are essentially involved.

Acknowledgments

The authors would like to thank Pete Aughton, D.A.B.T., ITR Laboratories Canada Inc., for proofreading.

References

  1. Haschek, WM; Voss, KA; Beasley, VR. Selected Mycotoxins Affecting Animal and Human Health. In Handbook of Toxicologic Pathology; Haschek, WM, Rousseaux, CG, Walling, MA, Eds.; Academic Press: San Diego, CA, USA, 2002; pp. 645–699. [Google Scholar]
  2. Surai, PF; Mezes, M; Melnichuk, SD; Fotina, TI. Mycotoxins and animal health: From oxidative stress to gene expression. Krmiva 2008, 50, 35–43. [Google Scholar]
  3. Chandra, J; Samali, A; Orrenius, S. Triggering and modulation of apoptosis by oxidative stress. Free Radic. Biol. Med 2000, 29, 323–333. [Google Scholar]
  4. Desjardins, AE; Hohn, TM; McComic, SP. Trichothecene biosynthesis in Fusarium species: chemistry, genetics, and significance. Microbiol. Mol. Biol. Rev 1993, 57, 595–604. [Google Scholar]
  5. Nelson, PE; Dignani, MC; Anaissie, EJ. Taxonomy, biology, and clinical aspects of Fusarium species. Clin Microbiol Rev 1994, 7, 479–504. [Google Scholar]
  6. Joffe, AZ. Foodborne Diseases: Alimentary Toxic Aleukia. In Handbook of Foodborne Diseases of Biological Origin; Rochcigle, M, Ed.; CRC Press: Boca Raton, FL, USA, 1983; pp. 353–495. [Google Scholar]
  7. Saito, M; Ohtsubo, K. Trichothecene Toxins of Fusarium Species. In Mycotoxins; Purchase, IFH, Ed.; Elsevier Scientific Publication: New York, NY, USA, 1977; pp. 264–280. [Google Scholar]
  8. Ueno, Y; Ishii, K; Saki, K; Kanadera, K; Tsunoda, S; Tanoka, H; Enomoto, M. Toxicological approaches to the metabolites of fusaria. IV. Microbial survey on “bean-hulls poisoning of horses” with the isolation of toxic trichothecenes, neosonaniol and T-2 toxin of Fusarium solani M-1-1. Jpn J Exp Med 1972, 42, 187–203. [Google Scholar]
  9. Bennet, JW; Klich, M. Mycotoxins. Clin. Microbiol. Rev 2003, 16, 497–516. [Google Scholar]
  10. Eriksen, GS; Petterson, H. Toxicological evaluation of trichothecenes in animal feed. Anim. Feed Sci. Technol 2004, 114, 205–239. [Google Scholar]
  11. Shifrin, VI; Anderson, P. Trichothecene mycotoxins trigger a ribotoxic stress response that activates c-jun N-terminal kinase and p38 mitogen-activated protein kinase and induces apoptosis. J Biol Chem 1999, 274, 13985–13992. [Google Scholar]
  12. Chang, IM; Mar, WC. Effect of T-2 toxin on lipid peroxidation in rats: Elevation of conjugated diene formation. Toxicol. Lett 1988, 40, 275–280. [Google Scholar]
  13. Eriksen, GS; Petterson, H; Lund, H. Comparative cytotoxicity of deoxynivalenol, nivalenol, triacetylated derivatives and de-epoxy metabolites. Food Chem. Toxicol 2004, 42, 619–624. [Google Scholar]
  14. IARC. Toxins derived from Fusarium sporotrichoides: T-2 toxin. In IARC Monographson the Evaluation of Carcinogenic Risks to Humans; IARC: Lyon, France, 1993; pp. 467–488. [Google Scholar]
  15. Sharma, RP. Immunotoxicity of mycotoxins. J. Dairy Sci 1993, 76, 892–897. [Google Scholar]
  16. Stanford, GK; Hood, RD; Haynes, AW. Effects of prenatal administration of T-2 toxin to mice. Res Commu Chem Pathol Pharmacol 1975, 10, 743–746. [Google Scholar]
  17. Williams, PP. Effects of T-2 mycotoxin on gastrointestinal tissues: A review of in vivo and in vitro models. Arch Environ Contam Toxicol 1989, 18, 374–387. [Google Scholar]
  18. Shinozuka, J; Li, G; Kiatipattanasakul, W; Uetsuka, K; Nakayama, H; Doi, K. T-2 toxin-induced apoptosis in lymphoid organs of mice. Exp. Toxicol. Pathol 1997, 49, 387–392. [Google Scholar]
  19. Li, G; Shinozuka, J; Uetsuka, K; Nakayama, H; Doi, K. T-2 toxin-induced apoptosis in Peyer’s patches of mice. J. Toxicol. Pathol 1997, 10, 59–61. [Google Scholar]
  20. Shinozuka, J; Suzuki, M; Noguchi, N; Sugimoto, T; Uetsuka, K; Nakayama, H; Doi, K. T-2 toxin-induced apoptosis in hematopoietic tissues of mice. Toxicol. Pathol 1998, 26, 674–681. [Google Scholar]
  21. Li, G; Shinozuka, J; Uetsuka, K; Nakayama, H; Doi, K. T-2 toxin-induced apoptosis in intestinal crypt epithelial cells of mice. Exp. Toxicol. Pathol 1997, 49, 447–450. [Google Scholar]
  22. Albarenque, SM; Shinozuka, J; Iwamoto, S; Nakayama, H; Doi, K. T-2 toxin-induced acute skin lesions in Wistar-derived hypotrichotic WBN/ILA-Ht rats. Histol. Histopathol 1999, 14, 337–342. [Google Scholar]
  23. Shinozuka, J; Miwa, S; Fujimura, H; Toriumi, W; Doi, K. Hepatotoxicity of T-2 Toxin, Trichothecene Mycotoxin. In New Strategies for Mycotoxin Research in Asia (Proceedings of ISMYCO Bangkok ‘06); Kumagai, S, Ed.; Japanese Association of Mycotoxicology: Tokyo, Japan, 2007; pp. 62–66. [Google Scholar]
  24. Sehata, S; Kiyosawa, N; Atsumi, F; Ito, K; Yamoto, T; Teranishi, M; Uetsuka, K; Nakayama, H; Doi, K. Microarray analysis of T-2 toxin-induced liver, placenta and fetal liver lesions in pregnant rats. Exp. Toxicol. Pathol 2005, 57, 15–28. [Google Scholar]
  25. Doi, K; Shinozuka, J; Sehata, S. T-2 toxin and apoptosis. J. Toxicol. Pathol 2006, 19, 15–27. [Google Scholar]
  26. Doi, K; Ishigami, N; Sehata, S. T-2 toxin-induced toxicity in pregnant mice and rats. Int. J. Mol. Sci 2008, 9, 2146–2158. [Google Scholar]
  27. Chaudhary, M; Rao, PV. Brain oxidative stress after dermal and subcutaneous exposure of T-2 toxin in mice. Food Chem. Toxicol 2010, 48, 3436–3442. [Google Scholar]
  28. Boyd, KE; Fitzpatrick, DW; Wilson, JR; Wilson, LM. Effect of T-2 toxin on brain biogenic monoamines in rats and chickens. Can. J. Vet. Res 1988, 52, 181–185. [Google Scholar]
  29. Martin, LJ; Morse, JD; Anthony, A. Quantitative cytophotometric analysis of brain neuronal RNA and protein changes in acute T-2 mycotoxin poisoned rats. Toxicon 1986, 24, 933–941. [Google Scholar]
  30. Wang, J; Fitzpatrick, DW; Wilson, JR. Effects of the trichothecene mycotoxin T-2 toxin on neurotransmitters and metabolites in discrete areas of the rat brain. Food Chem. Toxicol 1998, 36, 947–953. [Google Scholar]
  31. Wang, J; Fitzpatrick, DW; Wilson, JR. Effect of T-2 toxin on blood-brain barrier permeability monoamine oxidase activity and protein synthesis in rats. Food Chem. Toxicol 1998, 36, 955–961. [Google Scholar]
  32. Ishigami, N; Shinozuka, J; Katayama, K; Uetsuka, K; Nakayama, H; Doi, K. Apoptosis in the developing mouse embryos from T-2 toxin-inoculated dams. Histol. Histopathol 1999, 14, 729–733. [Google Scholar]
  33. Ishigami, N; Shinozuka, J; Katayama, K; Uetsuka, K; Nakayama, H; Doi, K. Apoptosis in mouse fetuses from dams exposed to T-2 toxin at different days of gestation. Exp. Toxicol. Pathol 2001, 52, 493–501. [Google Scholar]
  34. Rousseaux, CG; Schiefer, HB. Maternal toxicity, embryolethality and abnormal fetal development in CD-1 mice following one oral dose of T-2 toxin. J. Appl. Toxicol 1987, 7, 281–288. [Google Scholar]
  35. Stanford, GK; Hood, RD; Hayes, AW. Effect of prenatal administration of T-2 toxin to mice. Res. Commun. Chem. Path. Pharmacol 1975, 10, 743–746. [Google Scholar]
  36. Sehata, S; Kiyosawa, N; Makino, T; Atsumi, F; Ito, K; Yamoto, T; Teranishi, M; Baba, Y; Uetauka, K; Nakayama, H; Doi, K. Morphological and microarray analysis of T-2 toxin-induced rat fetal brain lesion. Food Chem. Toxicol 2004, 42, 1727–1736. [Google Scholar]
  37. Galtier, P; Paulin, F; Eeckhoutte, C; Larrieu, G. Comparative effects of T-2 toxin and diacetoxyscirpenol on drug metabolizing enzymes in rat tissues. Food Chem. Toxicol 1989, 27, 215–220. [Google Scholar]
  38. Guerre, P; Eeckhoutte, C; Burgat, V; Galtier, P. The effects of T-2 toxin exposure on liver drug metabolizing enzymes in rabbit. Food Add. Contam 2000, 17, 1019–1026. [Google Scholar]
  39. Jarpe, MB; Widmann, C; Knall, C; Schlesinger, TK; Gibson, S; Yujiri, T; Fanger, GR; Gelfand, EW; Johnson, GL. Anti-apoptotic versus pro-apoptotic signal transduction: checkpoints and stop signs along the roard to death. Oncogene 1998, 17, 1475–1582. [Google Scholar]
  40. Bold, S; Weidle, UH; Kolch, W. The kinase domain of MEKK1 induces apoptosis by dysregulation of MAP kinase pathways. Exp. Cell Res 2003, 283, 80–90. [Google Scholar]
  41. Ham, J; Eilers, A; Whitfield, J; Neame, SJ; Shah, B. c-JUN and the transcriptional control of neuronal apoptosis. Biochem. Pharmacol 2000, 60, 1015–1021. [Google Scholar]
  42. Annunziato, L; Amoroso, S; Pannaccione, A; Cataldi, M; Pignataro, G; D’Alessio, S; Sirabella, R; Second, A; Sibaud, L; DiRenzo, GF. Apoptosis induced in neuronal cells by oxidative stress: role played by caspases and intracellular calcium ions. Toxicol. Lett 2003, 139, 125–133. [Google Scholar]
  43. Troy, CM; Shelanski, ML. Caspase-2 redux. Cell Death Differ 2003, 10, 101–107. [Google Scholar]
  44. Huang, P; Akagawa, K; Yokoyama, Y; Nohara, K; Kano, K; Morimoto, K. T-2 toxin initially activates caspase-2 and induces apoptosis in U937 cells. Toxicol. Lett 2007, 170, 1–10. [Google Scholar]
  45. Kanemitsu, H; Yamauchi, H; Komatsu, M; Yamamoto, S; Okazaki, S; Uchida, K; Nakayama, H. 6-Mercaptopurine (6-MP) induces cell cycle arrest and apoptosis of neural progenitor cells in the developing rat brain. Neurotox. Teratol 2009, 31, 104–109. [Google Scholar]
  46. Katayama, K; Ueno, M; Yamauchi, H; Nakayama, H; Doi, K. Microarray analysis of genes in fetal central nervous system after ethylnitrosourea administration. Birth Defects Res. Part B 2005, 74, 255–260. [Google Scholar]
  47. Nam, C; Yamauchi, H; Nakayama, H; Doi, K. Etoposide induces apoptosis and cell cycle arrest of neuroepithelial cells in a p53-related manner. Neurotox. Teratol 2009, 28, 664–672. [Google Scholar]
  48. Ueno, M; Katayama, K; Yamauchi, H; Nakayama, H; Doi, K. Cell cycle and cell death regulation of neural progenitor cells in the 5-azacytidine (5AzC)-treated developing fetal brain. Exp Neurol 2006, 198, 154–166. [Google Scholar]
  49. Woo, GH; Bak, EJ; Nakayama, H; Doi, K. Molecular mechanisms of hydroxyurea (HU)-induced apoptosis in the mouse fetal brain. Neurotox. Teratol 2006, 28, 125–134. [Google Scholar]
  50. Ogunshola, OO; Antic, A; Donoghue, MJ; Fan, S-Y; Kim, H; Stewart, WB; Madri, JA; Ment, LR. Paracrine and autocrine function of neuronal vascular endothelial growth factor (VEGF) in the central nervous system. J. Biol. Chem 2002, 277, 11410–11415. [Google Scholar]
  51. Halliwell, B; Gutteridge, JMC. Free Radicals in Biology and Medicine, 3rd ed; Oxford University Press: New York, NY, USA, 1999. [Google Scholar]
  52. Lee, J-M; Jiang, L; Johnson, DA; Stein, TD; Kraft, AD; Calkins, MJ; Jakel, RJ; Jofnson, JA. Nrf2, a multiorgan protector? FASEB J 2005, 19, 1061–1066. [Google Scholar]
  53. Boesch-Saadatmandi, C; Wagner, AE; Graeser, AC; Hundhausen, C; Wollram, S; Rimbach, G. Ochratoxin A impairs Nrf2-dependent gene expression in porcine kidney tubulus cells. J. Anim. Phys. Anim. Nutr 2009, 93, 547–555. [Google Scholar]
  54. Boutin-Forzano, S; Charpin-Kadouch, C; Chabbi, S; Bennedjai, N; Dumon, H; Charpin, D. Wall relative humidity: A simple and reliable index for predicting Stachybotrys chartarum infestation in dwellings. Indoor Air 2004, 14, 196–199. [Google Scholar]
  55. Tsumori, T; Reijula, K; Johnsson, T; Hemminki, K; Hintikka, EL; Lindroos, O; Kalso, S; Koukila-Kahkola, P; Mussalo-Rauhamaa, H; Haahtela, T. Mycotoxins in crude building materials from water-damaged buildings. Appl. Environ. Microbiol 2000, 66, 1899–1904. [Google Scholar]
  56. Pestka, JJ; Yike, I; Dearborn, DG; Ward, MDW; Harkema, JR. Stachybotrys chartarum, trichothecene mycotoxins, and damp building-related illness: New insights into a public health enigma. Toxicol. Sci 2008, 104, 4–26. [Google Scholar]
  57. Shelton, BG; Kirkland, KH; Flanders, WD; Morris, GK. Profiles of airborne fungi in buildings and outdoor environments in the United States. Appl. Environ. Microbiol 2002, 68, 1743–1753. [Google Scholar]
  58. Hodgson, MJ; Morey, P; Leung, WY; Morrow, L; Miller, D; Jarvis, BB; Robbins, H; Halsey, JF; Storey, E. Building-associated pulmonary disease from exposure to Stachybotrys chartarum and Aspergillus versicolor. J. Occup. Environ. Med 1998, 40, 241–249. [Google Scholar]
  59. Johanning, E; Biagini, R; Hull, D; Morey, P; Jarvis, B; Landsbergis, P. Health and immunology study following exposure to toxigenic fungi (Stachybotrys chartarum) in a water-damaged office environment. Int. Arch. Occup. Environ. Health 1996, 68, 207–218. [Google Scholar]
  60. Gordon, WA; Cantor, JB; Johanning, E; Charatz, HJ; Ashman, TA; Breeze, JL; Haddad, L; Abramowitz, S. Cognitive impairment associated with toxigenic fungal exposure: A replication and extension of previous findings. Appl. Neuropsychol 2004, 11, 65–74. [Google Scholar]
  61. Hossain, MA; Ahmed, MS; Ghannoum, MA. Attributes of Stachybotrys chartarum and its association with human disease. J. Allergy Clin. Immunol 2004, 113, 200–208. [Google Scholar]
  62. Kirburn, KH. Role of molds and myxotoxins in being sick in buildings: Neurobehavioral and pulmonary impairment. Adv. Appl. Microbiol 2004, 55, 339–359. [Google Scholar]
  63. Andersen, B; Nielsen, KF; Jarvis, BB. Characterization of Stachybotrys from water-damaged buildings based on morphology, growth, and metabolite production. Mycologia 2002, 94, 392–403. [Google Scholar]
  64. Gregory, L; Pestka, JJ; Dearborn, DG; Rand, TG. Localization of satratoxin-G in Stachybotrys chartarum spores and spore-impacted mouse lung using immunocytochemistry. Toxicol. Pathol 2004, 32, 26–34. [Google Scholar]
  65. Yike, I; Distler, AM; Ziady, AG; Dearborn, DG. Mycotoxin adducts on human serum albumin: Biomerkers of exposure to Stachybotrys chartarum. Environ. Health Perspect 2006, 114, 1221–1226. [Google Scholar]
  66. Chung, YJ; Zhou, HR; Pestka, JJ. Transcriptional and posttranscriptional roles for p38 mitogen-activated protein kinase in upregulation of TNF-α expression by deoxynivalenol (vomitoxin). Toxicol. Appl. Pharmacol 2003, 193, 188–201. [Google Scholar]
  67. Moon, Y; Pestka, JJ. Deoxynivalenol-induced mitogen-activated protein kinase phosphorylation and IL-6 expression in mice suppressed by fish oil. J. Nutr. Biochem 2003, 14, 717–726. [Google Scholar]
  68. Zhou, HR; Lau, AS; Pestka, JJ. Role of double-stranded RNA-activated protein kinase R (PKR) in deoxynivalenol-induced ribotoxic stress response. Toxicol. Sci 2003, 74, 335–344. [Google Scholar]
  69. Iordanov, MS; Pribnow, D; Magun, JL; Dinh, TH; Pearson, JA; Chen, SL; Magun, BE. Ribotoxic stress response: Activation of the stress-activated protein kinase JNK1 by inhibitors of the peptidyl transferase reaction and by sequence-specific RNA damage to the alphasarcin/ricin loop in the 28S rRNA. Mol. Cell Biol 1997, 17, 3373–3381. [Google Scholar]
  70. Chung, YJ; Jarvis, B; Pestka, JJ. Modulation of lipopolysaccharide-induced proinflammatory cytokine production by satratoxins and other macrocyclic trichothecenes in the murine macrophage. J. Toxicol. Environ. Health A 2003, 66, 379–391. [Google Scholar]
  71. Chung, YJ; Yang, GH; Islam, Z; Pestka, JJ. Up-regulation of macrophage inflammatory protein-2 and complement 3A receptor by the trichothecenes deoxynivalenol and satratoxin G. Toxicology 2003, 186, 51–65. [Google Scholar]
  72. Hughes, BJ; Hsieh, GC; Jarvis, BB; Sharma, RP. Effects of macrocyclic trichothecene mycotoxins on the murine immune system. Arh. Environ. Contam. Toxicol 1989, 18, 388–395. [Google Scholar]
  73. Hughes, BJ; Jarvis, BB; Sharma, RP. Effects of macrocyclictrichothecene congeners on the viability and mitogenesis of mirine splenic lymphocytes. Toxicol Lett 1990, 50, 57–67. [Google Scholar]
  74. Pestka, JJ; Forsell, JH. Inhibition of human lymphocyte transformation by the macrocyclic trichothecene roridin A and verrucarin A. Toxicol Lett 1988, 41, 215–222. [Google Scholar]
  75. Yang, G-H; Jarvis, BB; Chung, Y-J; Pestka, JJ. Apoptosis induction by the satratoxins and other trichothecene mycotoxins: relationship to ERK, p38 MAPK, and SAPK/JNK activation. Toxicol. Appl. Pharmacol 2000, 164, 149–160. [Google Scholar]
  76. Cundliffe, E; Davies, JE. Inhibition of initiation, elongation, and termination of eukaryotic protein synthesis by trichothecene fungal toxins. Antimicrob. Agents Chemother 1977, 11, 491–499. [Google Scholar]
  77. Nielsen, KF; Huttunen, K; Hyvarinen, A; Andersen, B; Jarvis, BB; Hirvonen, MR. Metabolite profiles of Stachybotrys isolates from water-damaged buildings and their induction of inflammatory mediators and cytotoxicity in macrophages. Mycopathologia 2002, 154, 201–205. [Google Scholar]
  78. Islam, Z; Shinozuka, J; Harkema, JR; Pestka, JJ. Purification and comparative neurotoxicity of the trichothecenes satratoxin G and roridin L2 from Stachybotrys chartarum. J. Toxicol. Environ. Health A 2009, 72, 1242–1251. [Google Scholar]
  79. Nusuetrong, P; Pengsuparp, T; Meksuriyen, D; Tanitsu, M; Kikuchi, H; Muzugaki, M; Shimazu, K; Oshima, Y; Nakahata, N; Yoshida, M. Satratoxin H generates reactive oxygen species and lipid peroxides in PC12 cell. Biol. Pharm. Bull 2008, 31, 1115–1120. [Google Scholar]
  80. Rand, TG; Mahoney, M; White, K; Oulton, M. Microanatomical changes in alveolar type II cells in juvenile mice intratracheally exposed to Stachybotrys chartarum spores and toxin. Toxicol. Sci 2002, 65, 239–245. [Google Scholar]
  81. Wang, H; Yadav, JS. Global gene expression changes underlying Stachybotrys chartarum toxin-induced apoptosis in murine alveolar macrophages: Evidence of multiple signal transduction pathways. Apoptosis 2007, 12, 535–548. [Google Scholar]
  82. Wang, H; Yadav, JS. DNA damage, redox changes, and associated stress-inducible signaling events underlying the apoptosis and cytotocity in murine alveolar macrophage cell line MH-S by methanol-extracted Stachybotrys chartarum toxins. Toxicol. Appl. Pharmacol 2006, 214, 297–308. [Google Scholar]
  83. Islam, Z; Harkema, JR; Pestka, JJ. Satratoxin G from the black mold Stachybotrys chartarum evokes olfactory sensory neuron loss and inflammation in the murine nose and brain. Environ. Health Perspect 2006, 114, 1099–1107. [Google Scholar]
  84. Islam, Z; Amuzie, CJ; Harkema, JR; Pestka, JJ. Neurotoxicity and inflammation in the nasal airways of mice exposed to the macrocyclic trichothecene mycotoxin roridin A: Kinetic and potentiation by bacterial lipipolysaccharide coexposure. Oxford J. Life Sci. Med. Toxicol. Sci 2007, 98, 526–541. [Google Scholar]
  85. Chang, RC; Suen, KC; Ma, CH; Elyaman, W; Ng, HK; Hugon, J. Involvement of double-stranded RNA-dependent protein kinase and phosphorylation of eukaryotic initiation factor-2alpha in neuronal degeneration. J. Neurochem 2002, 83, 1215–1225. [Google Scholar]
  86. Ge, Y; Tsukatani, T; Nishimura, T; Furukawa, M; Miwa, T. Cell death of olfactory receptor neurons in a rat with nasosinusitis infected artificially with Staphylococcus. Chem. Senses 2002, 27, 521–527. [Google Scholar]
  87. Huang, CC; Chen, K; Huang, TY. Immunohistochemical studies of sensory neurons in rat olfactory epithelium. Eur. Arch. Otorhinolaryngol 1995, 252, 86–91. [Google Scholar]
  88. Wu, S; Kumar, KU; Kaufmam, RJ. Identification and requirement of three ribosome binding domains in dsRNA-dependent protein kinase (PKR). Biochemistry 1998, 37, 13816–13826. [Google Scholar]
  89. Garcia, MA; Meurs, EF; Esteban, M. The dsRNA protein kinase PKR: Virus and cell control. Biochemie 2007, 89, 799–811. [Google Scholar]
  90. Cowan, CM; Roskams, AJ. Apoptosis in the mature and developing olfactory neuroepithelium. Microsc. Res. Technol 2002, 58, 204–215. [Google Scholar]
  91. Farbman, AI; Buchholz, JA; Suzuki, Y; Coines, A; Speert, D. A molecular basis of cell death in olfactory epithelium. J. Comp. Neurol 1999, 414, 306–314. [Google Scholar]
  92. Suzuki, Y; Farbman, AI. Tumor necrosis factor-alpha-induced apoptosis in olfactory epithelium in vitro: Possible roles of caspase 1 (ICE), caspase-2 (ICH-1), and caspase-3 (CPP32). Exp. Neurol 2000, 165, 35–45. [Google Scholar]
  93. Islam, Z; Hegg, CC; Bae, HY; Pestka, JJ. Satratoxin G-induced apoptosis in PC-12 neuronal cells is mediated by PKR and caspase independent. Toxicol. Sci 2008, 105, 142–152. [Google Scholar]
  94. Nusuetrong, P; Yoshida, M; Tanitsu, MA; Kikuchi, H; Mizugaki, M; Shimazu, K; Pengsuparp, T; Meksuriyen, D; Oshima, Y; Nakahata, N. Involvementof reactive oxygen species and stress activated MAPKs in satoratoxin H-induced apoptosis. Eur. J. Pharmacol 2005, 507, 239–246. [Google Scholar]
  95. Chandra, J; Samali, A; Orrenius, S. Triggering and modulation of apoptosis by oxidative stress. Free Radic. Biol. Med 2000, 29, 323–333. [Google Scholar]
  96. Karunasena, E; Larrañaga, MD; Simoni, JS; Douglas, DR; Straus, DC. Building-associated neurological damage modeled in human cells: A mechanism of neurotoxic effects by exposure to mycotoxins in the indoor environment. Mycopathologia 2010, 170, 377–390. [Google Scholar]
  97. Thrasher, JD; Crawley, S. The biocontaminants and complexity of damp indoor spaces; more than what meets the eyes. Toxicol. Ind. Health 2009, 25, 583–615. [Google Scholar]
  98. Campbell, IL. Neuropathogenic acions of cytokines assessed in transgenic mice. Int. J. Dev. Neurosci 1995, 13, 275–284. [Google Scholar]
  99. Peters, A; Vweronesi, B; Calderon-Garciduenas, J; Gehr, P; Chen, LC; Greiser, M; Reed, W; Rothen-Rutishauser, B; Schurch, S; Schulz, H. Translocation and potential neurological effects of fine and ultrafine particles a critical update. Part. Fibre Toxicol 2006, 3, 1–13. [Google Scholar]
  100. Calderón-Garcidueñas, L; Azzarelli, B; Acuna, H; Garcia, R; Gambling, TM; Osnaya, N; Monroy, S; Tizapantzi, MDR; Carson, JL; Villarreal-Calderon, A; et al. Air pollution and brain damage. Toxicol. Pathol 2002, 30, 373–389. [Google Scholar]
  101. Calderón-Garcidueñas, L; Maronpot, RR; Torres-Jardon, R; Henríquez-Roldán, C; Schoonhoven, R; Acuña-Ayala, H; Villarreal-Carderón, A; Nakamura, J; Fernando, R; Reed, W; et al. DNA damage in nasal and brain tissues of canines exposed to air pollutants is associated with evidence of chronic brain inflammation and neurodegeneration. Toxico. Pathol 2003, 31, 524–538. [Google Scholar]
  102. Dutton, MF. Fumonisins, mycotoxins of increasing importance: their nature and their effects. Pharmacol. Ther 1996, 70, 137–161. [Google Scholar]
  103. Howard, PC; Eppley, RM; Stack, ME; Warbritton, A; Voss, KA; Lorentzen, RJ; Kovach, RM; Bucci, TJ. Fumonisin B1 carcinogenicity in a 2-year feeding study using F344 rats and B6C3 F1 mice. Environ. Health Perspect 2001, 109, 277–282. [Google Scholar]
  104. Wang, E; Norred, WP; Bacon, CW; Riley, RT; Merrill, AH, Jr. Inhibition of sphingolipid biosynthesis by fumonisins. J. Biol. Chem 1991, 22, 14486–14490. [Google Scholar]
  105. Merrill, AH, Jr; Sullards, MC; Wang, E; Voss, KA; Riley, RT. Sphingolipid metabolism: Role in signal transduction and disruption by fumonisins. Environ. Health Perspect 2001, 109, 283–289. [Google Scholar]
  106. Riley, RT; Enongene, E; Voss, KA; Norred, WP; Meredith, FI; Sharma, RP; Spitsbergen, J; Williams, DE; Carlson, DB; Merrill, AH, Jr. Sphingolipid perturbations as mechanisms for fumonisin carcinogenesis. Environ. Health Perspect 2001, 109, 301–308. [Google Scholar]
  107. Ross, PF; Rice, LG; Reagor, JC; Osweiler, GD; Wilson, TM; Nelson, HA; Owens, DL; Plattner, RD; Harlin, KA; Richard, JL; et al. Fumonisin B1 concentrations in feeds from 45 confirmed equine leukoencephalomalacia cases. J. Vet. Diagn. Invest 1991, 3, 238–241. [Google Scholar]
  108. Wilson, TM; Ross, PF; Rice, LG; Osweiler, GD; Nelson, HA; Owen, DL; Plattner, RD; Reggiardo, C; Noon, TH; Pickrell, JW. Fumonisin B1 levels associated with an epizootics of equine leukoencephalomalacia. J Vet Diagn Invest 1990, 2, 213–216. [Google Scholar]
  109. Goel, S; Schumacher, J; Lenz, SD; Kemppanien, BW. Effects of fusarium moniliforme isolates on tissue and serum sphingolipid concentrations in horses. Vet. Hum. Toxicol 1996, 38, 265–270. [Google Scholar]
  110. Marasas, WF; Riley, RT; Hendricks, KA; Stevens, VL; Sadler, TW; Gelineau-van Wanes, J. Fumonisins disrupt sphingolipid metabolism, folate transport, and neural tube development in embryo culture and in vivo: A potential risk factor for human neural tube defects among populations consuming fumonisin-contaminated maize. J. Nutr 2004, 134, 711–716. [Google Scholar]
  111. Sadler, TW; Merrill, AH; Stevens, VL; Sullards, MC; Wang, E; Wang, P. Prevention of fumonisin B1-induced neural tube defects by folic acid. Teratology 2002, 66, 169–176. [Google Scholar]
  112. Stevens, VL; Tang, J. Fumonisin B1-induced sphingolipid depletion inhibits vitamin uptake via the glycosylphosphatidylinositol-anchored folate receptor. J. Biol. Chem 1997, 272, 18020–18025. [Google Scholar]
  113. Harel, R; Futerman, AH. Inhibition of sphingolipid synthesis affects axonal outgrowth in cultured hippocampal neurons. J. Biol. Chem 1993, 268, 14476–14481. [Google Scholar]
  114. Kwon, OS; Slikker, W, Jr; Davies, DL. Biochemical and morphological effects of fumonisin B1 on primary cultures of rat cerebrum. Neurotoxicol. Teratol 2000, 22, 565–572. [Google Scholar]
  115. Monnet-Tschudi, F; Zurich, MG; Sorg, O; Matthieu, JM; Honegger, P; Schilter, B. The naturally occurring food mycotoxin fumonisin B1 impairs myelin formation in aggregating brain cell culture. Neurotoxicology 1999, 20, 41–48. [Google Scholar]
  116. Kwon, OS; Schmued, LC; Slikker, W, Jr. Fumonisin B1 in developing rats alter brain sphinganine levels and myelination. Neurotoxicolog 1997, 18, 571–580. [Google Scholar]
  117. Tsunoda, M; Dugyala, RR; Sharma, RP. Fumonisin B1-induced increases in neurotransmitter metabolite levels in different brain regions of BALB/c mice. Comp. Biochem. Physiol. C. Pharmacol. Toxicol. Endocrnol 1998, 120, 457–465. [Google Scholar]
  118. Porter, JK; Voss, KA; Chamberlain, WJ; Bacon, CW; Norred, WP. Neurotransmitters in rats fed fumonisin B1. Proc. Soc. Exp. Biol. Med 1993, 202, 360–364. [Google Scholar]
  119. Banczerowski-Pelyhe, I; Vilagi, I; Detri, L; Doczi, J; Kovacs, F; Kukorelli, T. In vivo and in vitro electrophysiological monitoring of rat neocortical activity after dietary fumonisin exposure. Mycopathologia 2002, 153, 149–156. [Google Scholar]
  120. Osuchowski, MF; Edwards, GL; Sharma, RP. Fumonisin B1-induced neurodegeneration in mice after intracerebroventricular infusion is concurrent with disruption of sphingolipid metabolism and activation of proinflammatory signaling. Neurotoxicology 2005, 26, 211–221. [Google Scholar]
  121. Bouhet, S; Hourcade, E; Loiseau, N; Fikry, A; Martinez, S; Roselli, M; Galtier, P; Mengheri, E; Oswald, IP. The mycotoxin fumonisin B1 alters the proliferation and the barrier function of porcine intestinal epithelial cells. Toxicol. Sci 2004, 77, 165–171. [Google Scholar]
  122. Ramasamy, S; Wang, E; Hennig, B; Merrill, AH, Jr. Fumonisin B1 alters sphingolipid metabolism and disrupt the barrier function of endothelial cells in culture. Toxicol. Appl. Pharmacol 1995, 133, 343–348. [Google Scholar]
  123. Osuchowski, MF; He, Q; Sharma, RP. Fumoniin B1 toxicity in the brain during coexisting lipopolysaccharide-related endotoxemia in BALB/c mice. Toxicol. Sci 2003, 72, 252–253. [Google Scholar]
  124. Szelenyi, J. Cytokines and the central nervous system. Brain Res. Bull 2001, 54, 329–338. [Google Scholar]
  125. Buccoliero, R; Futerman, AH. The roles of ceramide and complex sphingolipids in neuronal cell function. Pharmacol. Res 2003, 47, 409–419. [Google Scholar]
  126. Pettus, BJ; Chalfant, CE; Hannun, YA. Ceramide in apoptosis: An overview and current perspectives. Biochem. Biophys. Acta 2002, 1585, 114–125. [Google Scholar]
  127. Stockmann-Juvalla, H; Mikkola, J; Naarala, J; Loikkanen, J; Elovaara, E; Savolainen, K. Oxidative stress induced by fumonisin B1 in continuous human and rodent neural cell cultures. Free Radic. Res 2004, 38, 933–942. [Google Scholar]
  128. Mobio, TA; Anane, R; Baudrimont, I; Carratū, MR; Shier, TW; Dano, SD; Ueno, Y; Creppy, EE. Epigenetic properties of fumonisin B1: cell cycle arrest and DNA base modification in C6 glioma cells. Toxicol. Appl. Pharmacol 2000, 164, 91–96. [Google Scholar]
  129. Mobio, TA; Baudrimont, I; Sanni, A; Shier, TW; Saboureau, D; Dano, SD; Ueno, Y; Steyn, PS; Creppy, EE. Prevention by vitamin E of DNA fragmentation and apoptosis induced by fumonisin B1 in C6 glioma cells. Arch. Toxicol 2000, 74, 112–119. [Google Scholar]
  130. Mobio, TA; Tavan, E; Baudrimont, I; Anane, R; Carratū, MR; Sanni, A; Gbeassor, MF; Shier, TW; Narbonne, J-F; Creppy, EE. Comparative study of the toxic effects of fumonisin B1 in rat C6 glioma cells and p53-null mouse embryo fibroblasts. Toxicology 2003, 183, 65–75. [Google Scholar]
  131. Galvano, F; Campisi, A; Russo, A; Galvano, G; Palumbo, M; Renis, M; Barcellona, ML; Perez-Polo, JR; Vanella, A. DNA damage in astrocytes exposed to fumonisin B1. Neurochem. Res 2002, 27, 345–351. [Google Scholar]
  132. Galvano, F; Russo, A; Cardile, V; Galvano, G; Vanella, A; Renis, M. DNA damage in human fibroblasts exposed to fumonisin B1. Food Chem. Toxicol 2002, 40, 25–31. [Google Scholar]
  133. Ellerby, LM; Ellerby, HM; Park, SM; Holleran, AL; Murphy, AN; Fiskum, G; Kane, DJ; Testa, MP; Kayalar, C; Bredesen, DE. Shift of cellular oxidation-reduction potential in neural cells expressing Bcl-2. J Neurochem 1996, 67, 1259–1267. [Google Scholar]
  134. Kane, DJ; Sarafian, TA; Anton, R; Hahn, H; Butler, GE; Selverstone, VJ; Ord, T; Bredesen, DE. Bcl-2 inhibition of neural death: decreased generation of reactive oxygen species. Science 1993, 262, 1274–1277. [Google Scholar]
  135. Tjalkens, RB; Ewing, MM; Philbert, MA. Differential cellular regulation of the mitochondrial permeability transition in an in vitro model of 1,3-dinitrobenzene-induced encephalopathy. Brain Res 2000, 874, 165–177. [Google Scholar]
  136. Reed, JC; Meister, L; Tanaka, S; Cuddy, M; Yum, S; Geyer, C; Pleasure, D. Differential expression of bcl-2 protooncogene in neuroblastoma and other human tumor cell lines of neuronal origin. Cancer Res 1991, 51, 6529–6538. [Google Scholar]
  137. Stockmann-Juvala, H; Mikkola, J; Naarala, J; Loikkanen, J; Elovaara, E; Savolainen, K. Fuminisin B1-induced toxicity and oxidative damage in U-118MG glioblastoma cells. Toxicology 2004, 202, 173–183. [Google Scholar]
  138. Schmelz, EM; Dombrink-Kurzman, MA; Roberts, PC; Kozutsumi, Y; Kawasaki, T; Merrill, AH, Jr. Induction of apoptosis by fumonisin B1 in HT29 cells is mediated by the accumulation of endogenous free sphingoid bases. Toxicol Appl Pharmacol 1998, 148, 252–260. [Google Scholar]
  139. Tolleson, WH; Dooley, KL; Sheldon, WG; Thurman, JD; Bucci, TJ; Howard, PC. The Mycotoxin Fumonisin Induces Apoptosis in Cultured Human Cells and in Livers and Kidneys of Rats. In Advances in Experimental and Medical Biology Fumonisins in Food; Jackson, LS, DeVries, JW, Bullerman, LB, Eds.; Plenum Press: New York, NY, USA, 1996; pp. 237–250. [Google Scholar]
  140. Tolleson, WH; Melchior, WB; Morris, SM; McGarrity, LJ; Domon, OE; Muskhelishvili, L; James, SJ; Howard, PC. Apoptotic and anti-proliferaive effects of fuminisin B1 in human keratinocytes, fibroblasts, esophageal epithelial cells and hepatoma cells. Carcinogenesis 1996, 17, 239–249. [Google Scholar]
  141. Higuchi, Y. Chromosomal DNA fragmentation in apoptosis and necrosis induced by oxidative stress. Biochem. Pharmacol 2003, 66, 1527–1535. [Google Scholar]
  142. Slater, AFG; Nobel, CSI; van den Dobbelsteen, DJ; Orrenius, S. Signaling mechanisms and oxidative stress in apoptosis. Toxicol Lett 1995, 82/83, 149–153. [Google Scholar]
  143. Galtier, P. Pharmacokinetics of ochratoxin A in animals. IARC Sci Publ 1991, 187–200. [Google Scholar]
  144. Pfohl-Leszkowicz, A; Manderville, RA. Ochratoxin A: An overview on toxicity and carcinogenicity in animals and humans. Mol. Nutr. Food Res 2007, 51, 61–99. [Google Scholar]
  145. Garies, M; Wolff, J. Relevance of mycotoxin contaminated feed for farm animals and carryover of mycotoxins to food of animal origin. Mycoses 2000, 43, 79–83. [Google Scholar]
  146. Mally, A; Hard, GC; Dekant, W. Ochratoxin A as a potential etiologic factor in endemic nephropathy: lesions from toxicity studies in rats. Food Chem. Toxicol 2007, 45, 2254–2260. [Google Scholar]
  147. Krogh, P. Role of ochratoxin in disease causation. Food Chem. Toxicol 1992, 30, 213–224. [Google Scholar]
  148. Kane, A; Creppy, EE; Roschenthaler, R; Dirheimer, G. Changes in urinary and renal tubular enzymes caused by subchronic administration of ochratoxin A in rats. Toxicology 1986, 42, 233–243. [Google Scholar]
  149. Petkova-Bocharova, T; Chernozemsky, IN; Castegnaro, M. Ochratoxin A in human blood in relation to Balkan endemic nephropathy and urinary system tumors in Bulgaria. Food Addit. Contam 1988, 5, 299–301. [Google Scholar]
  150. Lea, T; Steinen, K; Stormer, FC. Mechanism of ochratoxin A-induced immunosuppression. Mycopathologia 1989, 107, 153–159. [Google Scholar]
  151. Stromer, FC; Lea, T. Effects of ochratoxin A upon early and late events in human T-cell proliferation. Toxicology 1995, 95, 45–50. [Google Scholar]
  152. Arora, RG; Frolen, H; Fellner-Feldegg, H. Inhibition of ochratoxin A teratogenesis by zearalenone and diethylstilbesterol. Food Chem. Toxicol 1983, 21, 779–783. [Google Scholar]
  153. Fukui, Y; Hayasaka, S; Itoh, M; Takeuchi, Y. Development of neurons and synapses in ochratoxin A-induced microcephalic mice: a quantitative assessment of somatosensory cortex. Neurotoxicol. Teratol 1992, 14, 191–196. [Google Scholar]
  154. Pfohl-Leszkowicz, A; Chakor, K; Creppy, EE; Dirheimer, G. DNA adduct formation in mice treated with ochratoxin A. IARC Sci Publ 1991, 245–253. [Google Scholar]
  155. Sava, V; Reunova, O; Velasquez, A; Harbison, R; Sanchez-Ramos, J. Acute neurotoxic effects of the fungal netabolite ochratoxin-A. Neurotoxicology 2006, 27, 82–92. [Google Scholar]
  156. Kuiper-Goodman, T; Hilts, C; Billiard, SM; Kiparissis, Y; Richard, ID; Hayward, S. Health risk assessment of ochratoxin A for all age-sex strata in a market economy. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess 2010, 27, 212–240. [Google Scholar]
  157. Creppy, EE; Chakor, K; Fisher, MJ; Dirheimer, G. The mycotoxin ochratoxin A is a substrate for phenylalanine hydroxylase in isolated rat hepatocytes and in vivo. Arch. Toxicol 1990, 64, 279–284. [Google Scholar]
  158. Creppy, EE; Kane, D; Dirheimer, G; Lafarge-Frayssinet, C; Mousset, S; Frayssinet, C. Genotoxicity of ochratoxin A in mice: DNA single-strand break evaluation in spleen, liver and kidney. Toxicol. Lett 1985, 28, 29–35. [Google Scholar]
  159. Dirheimer, G; Creppy, EE. Mechanism of action of ochratoxin A. IARC Sci Publ 1991, 171–186. [Google Scholar]
  160. Gautier, JC; Holzhaeuser, D; Markovic, J; Gremaud, E; Schilter, B; Turesky, RJ. Oxidative damage and stress response from ochratoxin exposure in rats. Free Radic. Biol. Med 2001, 30, 1089–1098. [Google Scholar]
  161. Bryan, NS; Rassaf, T; Maloney, RE; Rodriguez, CM; Saijo, F; Rodriguez, JR; Feelisch, M. Cellular targets and mechanisms of nitros(yl)ation: An insight into their nature and kinetics in vivo. Proc. Natl. Acad. Sci. USA 2004, 101, 4308–4313. [Google Scholar]
  162. Thomas, JA; Mallis, RJ. Aging and oxidation of reactive protein sulfhydryls. Exp. Gerontol 2001, 36, 1519–1526. [Google Scholar]
  163. Marin-Kuan, M; Nestler, S; Verguet, C; Bezençon, C; Piguet, D; Mansourian, R; Holzwarth, J; Grigorov, M; Delatour, T; Mantel, P; et al. A toxicogenomics approach to identify new plausible epigenetic mechanisms of ochratoxin A carcinogenicity in rat. Toxicol. Sci 2006, 89, 120–134. [Google Scholar]
  164. Aleo, MD; Wyatt, RD; Schnellmann, RG. Mitochondrial dysfunction is an early event in ochratoxin A but not oosporein toxicity to rat renal proximal tubules. Toxicol. Appl. Pharmacol 1991, 107, 73–80. [Google Scholar]
  165. Wei, YH; Lu, CY; Lin, TN; Wei, RD. Effect of ochratoxin A on rat liver mitochondrial respiration and oxidative phosphorylation. Toxicology 1985, 36, 119–130. [Google Scholar]
  166. Belmadani, A; Tramu, G; Betbeder, AM; Creppy, EE. Subchronic effects of ochratoxin A on young adult rat brain and partial prevention by aspartate, a sweetener. Hum. Exp. Toxicol 1998, 17, 380–386. [Google Scholar]
  167. Hayes, AW; Cain, JA; Moore, BG. Effects of aflatoxin B1, ochratoxin A and rubratoxin B on infant rats. Food Cosmet. Toxicol 1977, 15, 23–27. [Google Scholar]
  168. Hayes, AW; Hood, RD; Lee, HL. Teratogenic effects of ochratoxin A in mice. Teratology 1974, 9, 93–97. [Google Scholar]
  169. Wangikar, PB; Dwivedi, P; Sharma, AK; Sinha, N. Effect in rats of simultaneous prenatal exposure to ochratoxin A and aflatoxin B(1). II. Histopathological features of teratological anomalies induced in fetuses. Birth Defects Res. B 2004, 71, 352–358. [Google Scholar]
  170. Tamura, M; Hirata, Y; Matsutani, T. Neurochemical effects of prenatal treatment with ochtatoxin A on fetal and adult mouse brain. Neurochem. Sci 1988, 13, 1139–1147. [Google Scholar]
  171. Belmadani, A; Tramu, G; Betbeder, AM; Steyn, PS; Creppy, EE. Regional selectivity to ochratoxin A, distribution and cytotoxicity in rat brain. Arch. Toxicol 1998, 72, 656–662. [Google Scholar]
  172. 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]
  173. Sanchez-Ramos, J; Overvik, E; Ames, BN. A marker of oxyradical-mediated DNA damage (oxo8dG) is increased in nigro-striatum of Parkinson’s disease brain. Neurodegeneration (incorporated into Exp. Neurol.) 1994, 3, 197–204. [Google Scholar]
  174. Bunge, I; Dirheimer, G; Roschenthaler, R. In vivo and in vitro inhibition of protein synthesis in Bacillus stearothermophilus by ochratoxin A. Biochem Biophys Res Commun 1978, 83, 398–405. [Google Scholar]
  175. Creppy, EE; Kern, D; Steyn, PS; Vleggaar, R; Roschenthaler, R; Dirheimer, G. Comparative study of the effect of ochratoxin a analogues on yeast aminoacyl-tRNA synthetases and on the growth and protein synthesis of hepatoma cells. Toxicol. Lett 1983, 19, 217–224. [Google Scholar]
  176. Palmer, TD; Takahashi, J; Gage, FH. The adult rat hippocampus contains primordial neural stem cells. Mol Cell Neurosci 1997, 8, 389–404. [Google Scholar]
  177. Song, HJ; Stevens, CF; Gage, FH. Neuronal stem cells from adult hippocampus develop essential properties of functional CNS neurons. Nat. Neurosci 2002, 5, 438–445. [Google Scholar]
  178. Chen, H; Tung, YC; Li, B; Iqbal, K; Grundke-Iqbal, I. Trophic factors counteract elevated FGF-2-induced inhibition of adult neurogenesis. Neurobiol. Aging 2006, 28, 1148–1162. [Google Scholar]
  179. Kawai, T; Takagi, N; Mochizuki, N; Besshoh, S; Sakanishi, K; Nakahara, M; Takeo, S. Inhibitor of vascular endothelial growth factor receptor tyrosine kinase attenuates cellular proliferation and differentiation to mature neurons in the hippocampal dentate gyrus after transient forebrain ischemia in the adult rats. Neuroscience 2006, 141, 1209–1216. [Google Scholar]
  180. Lagace, DC; Yee, JK; Bolanos, CA; Eisch, AJ. Juvenile administration of methylphenidate attenuates adult hippocampal neurogenesis. Biol. Psychiatry 2006, 60, 1121–1130. [Google Scholar]
  181. Rossi, C; Angelucci, A; Costantin, L; Braschi, C; Mazzantini, M; Babbini, F; Fabbri, ME; Tessarollo, L; Maffei, L; Berardi, N; Caleo, M. Brain-derived neurotrophic factor (BDNF) is required for the enhancement of hippocampal neurogenesis following environmental enrichment. Eur J Neurosci 2006, 24, 1850–1856. [Google Scholar]
  182. Delibas, N; Altuntas, I; Yonden, Z; Ozcelik, N. Ochratoxin A reduces NMDA receptor subunits 2A and 2B concentrations in rat hippocampus: partial protective effect of melatonin. Hum. Exp. Toxicol 2003, 22, 335–339. [Google Scholar]
  183. Sava, V; Velasquez, A; Song, S; Sanchez-Ramos, J. Adult hippocampal neural stem/progenitor cells in vitro are vulnerable to the mycotoxin ochratoxin A. Toxicol. Sci 2007, 98, 187–197. [Google Scholar]
  184. Alexander, P. The role of DNA lesions in processes leading to aging in mice. Sym. Soc. Exp. Biol 1967, 21, 29–50. [Google Scholar]
  185. Korr, H; Schultz, B. Unscheduled DNA synthesis in various types of cells of the mouse brain in vivo. Exp. Brain Res 1989, 74, 573–578. [Google Scholar]
  186. Crago, BR; Gray, MR; Nelson, LA; Davis, M; Arnold, L; Thrasher, JD. Psychological, neuropsychological, and electrocortical effects of mixed mold exposure. Arch. Environ. Health 2003, 58, 452–563. [Google Scholar]
  187. Gordon, WA; Cantor, JB. The diagnosis of cognitive impairment associated with exposure to mold. Adv. Appl. Microbiol 2004, 55, 361–374. [Google Scholar]
  188. Rea, WJ; Didriksen, N; Simon, TR; Pan, Y; Fenyves, EJ; Griffiths, B. Effects of toxic exposure to molds and mycotoxins in building-related illnesses. Arch. Environ. Health 2003, 58, 399–405. [Google Scholar]
  189. Yoon, S; Cong, W-T; Bang, Y; Lee, SN; Yoon, CS; Kwack, SJ; Kang, TS; Lee, KY; Choi, J-K; Choi, HJ. Proteome response to ochratoxin A-induced apoptotic cell death in mouse hippocampal HT22 cells. Neurotoxicology 2009, 30, 666–676. [Google Scholar]
  190. Sato, A; Miyazaki, E; Satake, A; Hiramoto, A; Hiraoka, O; Miyake, T; Kim, HS; Wataya, Y. Proteome and transcriptome analysis of cell death induced by 5-fluoro-2′-deoxyuridine. Nucleic Acids Symp. Ser (Oxf) 2007, 51, 433–434. [Google Scholar]
  191. Siddiq, A; Ayoub, IA; Chavez, JC; Aminova, L; Shah, S; LaManna, JC; Patton, SM; Connor, JR; Cherny, RA; Volitakis, I; et al. Hypoxia-indicible factor prolyl 4-hydroxylase inhibition. A target for neurprotection in the central nervous system. J. Biol. Chem 2005, 280, 41732–41743. [Google Scholar]
  192. Lei, T; He, Q; Cai, Z; Zhou, Y; Wang, Y; Si, L; Cai, Z; Chiu, JF. Proteomic analysis of chromium cytotoxicity in cultured rat lung epithelial cells. Proteomics 2008, 8, 2420–2429. [Google Scholar]
  193. Noguchi, M; Takata, T; Kimura, Y; Manno, A; Murakami, K; Koike, M; Ohizumi, H; Hori, S; Kakizuka, A. ATPase activity of p97/valosin-containing protein is regulated by oxidative modification of the evolutionally conserved cysteine 522 residue in Waker A motif. J. Biol. Chem 2005, 280, 41332–41341. [Google Scholar]
  194. Zhang, X; Boesch-Saadatmandi, C; Lou, Y; Wolffram, S; Huebbe, P; Rimbach, G. Ochratoxin A induces apoptosis in neuronal cells. Genes Nutr 2009, 4, 41–48. [Google Scholar]
  195. Zurich, MG; Lengacher, S; Braissant, O; Monnet-Tschudi, F; Pellerin, L; Honegger, P. Unusual astrocyte reactivity caused by the food mycotoxin ochratoxin A in aggregating rat brain cell cultures. Neuroscience 2005, 134, 771–782. [Google Scholar]
  196. Hong, JT; Lee, MK; Park, KS; Jung, KM; Lee, RD; Jung, HK; Park, KL; Yang, KJ; Chung, YS. Inhibitory effect of peroxisome proliferator-activated receptor gamma agonist on ochratoxin A-induced cytotoxicity and activation of transcription factors in cultured rat embryonic midbrain cells. J. Toxicol. Environ Health A 2002, 65, 407–418. [Google Scholar]

Share and Cite

MDPI and ACS Style

Doi, K.; Uetsuka, K. Mechanisms of Mycotoxin-Induced Neurotoxicity through Oxidative Stress-Associated Pathways. Int. J. Mol. Sci. 2011, 12, 5213-5237. https://doi.org/10.3390/ijms12085213

AMA Style

Doi K, Uetsuka K. Mechanisms of Mycotoxin-Induced Neurotoxicity through Oxidative Stress-Associated Pathways. International Journal of Molecular Sciences. 2011; 12(8):5213-5237. https://doi.org/10.3390/ijms12085213

Chicago/Turabian Style

Doi, Kunio, and Koji Uetsuka. 2011. "Mechanisms of Mycotoxin-Induced Neurotoxicity through Oxidative Stress-Associated Pathways" International Journal of Molecular Sciences 12, no. 8: 5213-5237. https://doi.org/10.3390/ijms12085213

Article Metrics

Back to TopTop