chapter mycotoxine immunity- blue book 2005

Upload: ceciliapistol

Post on 13-Apr-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    1/46

    P.F. Surai and J.E. Dvorska 93

    93

    Effects of mycotoxins on antioxidantsystems

    A delicate balance between antioxidants andpro-oxidants in the body in general andspecifically in the cell is responsible forregulation of various metabolic pathwaysleading to maintenance of immuno-competence, growth and development andprotection against stress conditions associatedwith commercial poultry production (Surai andDvorska, 2001). This balance can be regulatedby dietary antioxidants, including vitamin E

    (Surai et al., 1999), carotenoids (Surai andSpeake, 1998; Surai et al., 2001) andselenium (Se) (Surai, 2000). On the otherhand, nutritional stress factors have a negativeimpact on this antioxidant/pro-oxidantbalance. In this respect mycotoxins areconsidered to be among the most importantfeed-borne stress factors.

    It is not clear at present whethermycotoxins stimulate lipid peroxidationdirectly by enhancing free radical productionor if the increased tissue susceptibility to lipidperoxidation is a result of a compromised

    antioxidant system. It seems likely that bothprocesses are at work. In most cases lipidperoxidation in tissues caused by mycotoxinswas associated with decreased concentrationsof natural antioxidants. For example, in anexperiment with quail, levels of the primaryliver antioxidants ( tocopherol, tocopherol, carotenoids and ascorbic acid)were significantly decreased as a result of T-

    2 toxin consumption (Dvorska and Surai,2001; Figure 1).

    Similarly, the presence of T-2 toxin in thediet decreased the concentration of tocopherol in the chicken liver (Hoehler andMarquardt, 1996). T-2 toxin consistentlydepressed concentrations of vitamin E inchicken plasma (Coffin and Combs, 1981).Addition of micelle-promoting compounds(taurocholic, monoolein, and oleic acids)

    alleviated depression in plasma vitamin E,indicating interference of T-2 toxin withmicelle formation during vitamin Eabsorption. Similarly, aflatoxin B

    1(AFB

    1) in

    the feed interfered with the accumulation ofcarotenoids in chicken tissues (Schaeffer etal., 1988) inducing pale bird syndrome inbirds. In fact, AFB

    1 caused a significant

    depression of lutein in the toe web, liver,serum and mucosa (Schaeffer et al.,1988a).Pigment restoration was accomplished byfeeding the same diet supplemented withlutein (70 mg/kg). In young chickens AFB

    1

    reduced the lutein content of jejunal mucosaup to 35% while serum lutein was reducedup to 70% (Tyczkowski and Hamilton, 1987),suggesting that AFB

    1 interfered with the

    absorbtion, transport and deposition ofcarotenoids. More precisely, AFB

    1impaired

    lutein absorption in chickens (Tyczkowskiand Hamilton, 1987a). In similar fashion,ochratoxin A (OTA) was shown to affect

    5

    EFFECTS OF MYCOTOXINS ON ANTIOXIDANT STATUSAND IMMUNITY

    1*Peter F. Surai and 2Julia. E. Dvorska1Avian Science Research Centre, Scottish Agricultural College, Auchincruive, Ayr,Scotland, UK and 2Sumy National Agrarian University, Sumy, Ukraine

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    2/46

    94 Effects of mycotoxins on antioxidant status and immunity

    carotenoid assimilation in chickens. Again,depression in uptake of carotenoids byintestinal mucosa and depressed transport inserum were considered to be importantmechanisms of AFB

    1 action on carotenoid

    metabolism (Schaeffer et al.,1988; Huff andHamilton, 1975).

    In general, malabsorption syndrome isconsidered a common result ofmycotoxicoses. For example, aflatoxicosis,ochratoxicosis and T-2 toxicosis wereproduced by feeding diets containing gradedconcentrations of the appropriate toxin tobroiler chickens from hatching until 3 weeksof age (Osborne et al., 1982). In thisexperiment AFB

    1, even at levels lower than

    needed for growth inhibition, produced amalabsorption syndrome characterized bysteatorrhea, hypocarotenoidemia, anddecreased concentrations of bile salts andpancreatic enzymes. T-2 toxin also producedmalabsorption, but at concentrations higherthan required to inhibit growth. Ochra-toxicosis produced mainly hypocarote-noidemia (Osborne et al., 1982). It ispostulated that the decreased level of vitaminA in the quail liver as a result of T-2 toxinconsumption (Dvorska and Surai, 2001) is also

    a reflection of the decreased intestinalabsorption of fat soluble nutrients.

    The presence of OTA in the dietsignificantly decreased the concentration oftocopherol in the chicken liver (Hoehlerand Marquardt, 1996). Furthermore, aflatoxin-treated barrows had decreased serumtocopherol and retinol concentrationscompared with control and pre-test values,and decreased tocopherol concentration incardiac tissue (Harvey et al., 1994).Aurofusarin decreased vitamin E andcarotenoid concentration in quail egg yolk(Dvorska et al., 2001a), in the liver, yolk sacmembrane and other tissues of newly hatchedquail (Dvorska et al.,2002; Dvorska et al.,2003; Dvorska and Surai, 2004). It isinteresting to note that the carotenoid andvitamin A concentrations in the quail liverwere also decreased when the maternal dietwas supplemented with aurofusarin.

    A pro-oxidant effect of mycotoxins inmany cases could be mediated throughchanges in reduced glutathione (GSH)concentration. For example, Rizzo et al.(1994) demonstrated that T-2 toxin decreasedGSH in rat liver. Treatment of fasted micewith a single dose of T-2 toxin (1.8 or 2.8

    0

    5

    10

    15

    20

    25

    Control T-2 T-2+Zeolite T-2+Mycosorb

    -tocopherol -tocopherol

    carotenoids ascorbate

    g/g

    tissue

    Figure 1. Effect of T-2 toxin with and without supplemental toxin adsorbents on antioxidant concentrations in

    quail liver (adapted from Dvorska and Surai, 2001).

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    3/46

    P.F. Surai and J.E. Dvorska 95

    mg/kg BW) by oral gavage led to a markeddecrease in hepatic GSH (Atroshi et al.,1997).In male broiler chicks, hepatic GSHconcentration decreased after 7 days oftreatment (1.5 mg T-2 toxin/kg BW/day) (Lealet al.,1999). Acute exposure of mice to T-2toxin (4 mg/kg, s.c.) resulted in a progressivedecrease in hepatic GSH, reaching a minimum6-8 hrs after toxin administration (Fricke andJorge, 1991). Intraperitoneal administrationof AFB

    1to rats (2 mg/kg) was also associated

    with decreased GSH in the liver. In contrast,in 3-week-old male chickens daily aflatoxin

    gavage (2 mg/kg BW, in corn oil) for 5 and10 days elevated hepatic GSH; and renal GSHwas elevated after 10 days (Beers et al.,1992a). Similarly, hepatic GSH increased 2and 8 hrs following a single AFB

    1dose and

    continued to increase through five daily dosesof AFB

    1(Beers et al.,1992b).

    There was GSH depletion in cultured rathepatocytes as a result of AFB

    1toxicosis (Liu

    et al.,1999). Similarly, consumption of OTAfor two weeks was associated with a depletionof GSH from the mouse liver (Atroshi et al.,2000). The mycotoxin patulin also decreased

    GSH concentration in rat hepatocytes (Busbeeet al.,1999). Considering that glutathione isresponsible for the maintenance of redoxstatus of the cell (Sies, 1999) and thereforeparticipates in regulation of gene expression(Arrigo, 1999), changes in GSH status couldbe detrimental.

    One of the most important mycotoxinactions is their effect on antioxidant enzymes.Depending on experimental conditions(species, doses, route and duration ofadministration, concentrations of otherantioxidants etc.), antioxidant enzyme

    activities can be increased in response tooxidative stress or decreased by direct orindirect action of mycotoxins. For example,treatment of pig kidney cells with 50 mMfumonisin B

    1 (FB

    1) for 24 hrs significantly

    decreased cellular GSH and increased theactivities of glutathione reductase (Kang andAlexander, 1996). The activities ofglutathione peroxidase (GSH-Px), catalase,

    and Cu/Zn-superoxide dismutase (SOD) werenot changed by this treatment. Oraladministration of T-2 mycotoxin to rats (1.25mg/kg) for 5 days decreased the activity ofliver glutathione-S-transferase (Ahmed andRam, 1986). In contrast, feeding a single doseof T-2 toxin (2 mg/kg BW) to rats increasedactivities of GSH-shuttle enzymes includingGSH-Px, glutathione reductase and glucose-6-phosphate dehydrogenase (Suneja et al.,1989), probably reflecting an adaptiveresponse to oxidative stress. On the otherhand, when male rats were given a diet

    deficient in vitamins C and E and Se and wereadministered orally a single dose ofdeoxynivalenol (DON) or T-2 toxin, there wasa significant decrease in activities of GSH-Px, catalase, SOD and glutathione reductase(Rizzo et al., 1994). Activity of GSH-Px inrat blood was decreased due to consumptionof AFB

    1(Choi et al.,1995). Administration of

    AFB1to rats (2 mg/kg intraperitoneally) caused

    a significant decrease in the activities of SOD,catalase, GSH-Px, glutathione-S-transferaseand glutathione reductase in liver (Rastogi etal., 2001). A significant increase in SOD

    activity occurred in the liver following AFB1exposure of the ducks (Barraud et al.,2001).

    When considering detrimental effects ofmycotoxins on antioxidant systems it isnecessary to be aware that combinations ofseveral mycotoxins can be more toxic thanindividual mycotoxins, as was shown for acombination of T-2 toxin and OTA (Kubenaet al.,1989).

    One of the most important targets formycotoxins is embryonic development. Sincechicken embryo tissues contain high levelsof PUFA, they are vulnerable to peroxidation

    and oxidative stress caused by mycotoxinscould be lethal. As mentioned previously,aurofusarin increased late mortality of quailembryos (Dvorska et al.,2001a). Furthermore,contamination of the diet with T-2 toxinmarkedly decreased egg production andimpaired hatchability (Tobias et al.,1992).Confirmation of a possible association of thiseffect with oxidative stress came from data

    05-Surai.p65 19/12/2004, 23:2495

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    4/46

    96 Effects of mycotoxins on antioxidant status and immunity

    indicating that increased dietary vitamin Eduring the first week of the experimentsignificantly decreased the number of infertileeggs and significantly improved the hatchingpercentage (Tobias et al.,1992).

    Increased lipid peroxidation as a

    consequence of mycotoxicoses

    As illustrated in Table 1, OTA has astimulating effect on lipid peroxidation. Inmost of cases, accumulation of thiobarbituricacid reactive substances (TBARS) was usedas a measurement of lipid peroxidation.Ethane exhalation, EPR-registered freeradicals, hydroxyl radical formation, single-

    strand cleavage DNA, DNA adduct formationas well as LDH release were also used toconfirm pro-oxidant properties of OTA.Various in vitro and in vivo systems werealso used including liver microsomes,phospholipid vesicles, primary cell cultures,whole organ and whole body measurements.

    T-2 toxin was also shown to have pro-oxidant properties (Table 2). Those propertieswere confirmed with rat, mouse and quailliver tissue and yeast. TBARS accumulationwas the method used in most of the studies,however, conjugate diene formation and DNA

    fragmentation were also demonstrated. Effectof AFB

    1 on lipid peroxidation has been

    studied in rat liver and kidney as well as incultured hepatocytes and in anin vitromodel

    Table 1. Stimulation of lipid peroxidation by ochratoxin A.

    Mycotoxin Tissue Lipid peroxidation Protective effect Referencesmeasurement of antioxidants

    OTA in vitro Rat liver TBARS - Rahimtula et al.,1988,microsomes Gautier et al.,2001; Khan et

    al., 1989

    OTA in feed Rats Ethane exhalation - Rahmitula et al.,1988OTA in feed Rats MDA in serum, - Meki and Hussein, 2001liver, kidney

    OTA in vitro Phospholipid TBARS , oxygen - Omar et al., 1990vesicles uptake

    OTA and its Bacillus brevis Free radical Vitamin E Hoehler et al.,1996analogs bacteria generation (EPR)

    OTA in feed Chicken liver TBARS - Hoehler and Marquardt,1996; Hoehler et al.,1997

    OTA in vitro Vero cells in TBARS SOD, catalase Baudrimont et al.,1997a;culture aspartame 1997b

    OTA in diet Rat liver TBARS - Hoehler et al.,1997OTA in vitro A model oxid- OH, single- - Gillman et al.,1999

    ation system strand cleavageDNA

    OTA in vitro Astrocytes and TBARS, LDH - Belmadani et al.,1999neurons release

    OTA in feed Mouse and rat DNA adduct Retinol, ascorbic Grosse et al.,1997kidney formation acid, vitamin E

    OTA in feed Rats DNA adduct Aspartame Creppy et al.,1998formation

    OTA in vitro Rat kidney TBARS - Gautier et al.,2001microsomes

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    5/46

    P.F. Surai and J.E. Dvorska 97

    system (Table 3). Similar to the examplesabove, TBARS accumulation was substantiallyincreased as well as conjugate dieneproduction. At the same time GSHconcentration and activities of antioxidantenzymes significantly declined as a result ofAFB

    1action.

    Fumonisin B1 also stimulated lipid

    peroxidation in rat liver, rat liver nucleifraction, primary rat hepatocytes, Vero cellsin culture and phosphatidyl choline (PC)bilayers. In those systems TBARSaccumulation and DNA strand breaks wereincreased (Table 4). DON increased TBARSformation in rat and mouse liver anddecreased GSH in rat brain and spleen. Thereare also data indicating pro-oxidant propertiesof zearalenone (Karagezyan et al., 1995;Ghedira-Chekir et al., 1999) and citrinin(Ribeiro et al.,1997). Aurofusarin has beenshown to decrease antioxidant defences andstimulate lipid peroxidation in quail egg andtissues of newly hatched quail (Dvorska etal.,2002; 2003; Dvorska and Surai, 2004).

    It is clear from these data that mycotoxinsstrongly promote lipid peroxidation in variousin vitro and in vivosystems. This effect wasobvious no matter which measurement was

    used to assess the process of lipidperoxidation.

    Mycotoxins and apoptosis

    The maintenance of tissue homeostasisinvolves the removal of superfluous anddamaged cells. This process is often referredto as programmed cell deathor apoptosis,since it is thought that cells activate anintrinsic death program contributing to theirown demise (Sastre et al., 1996). Severalprocesses, such as initiation of death signalsat the plasma membrane, expression of pro-apoptotic oncoproteins, activation of deathproteases, endonucleases etc., ultimatelycoalesce to a common irreversible execution

    phase leading to cell demise. A balancebetween cell death and cell survival factorsplays a major role in the decision process asto whether a cell should live or die (Ray etal.,2000).

    Apoptosis is distinguishable from necrosis.When cell death is induced by osmotic,physical or chemical damage, early disruptionof external and internal membranes takesplace with subsequent liberation of denatured

    Table 2. Stimulation of lipid peroxidation by T-2 toxin.Mycotoxin Tissue Lipid peroxidation Protective effect References

    measurement of antioxidants

    T-2 in feed Rat liver TBARS Se, ascorbic Tsuchida et al.,1984;acid, vitamin E Suneja et al.,1989;

    Schuster et al.,1987; Rizzoet al.,1994

    T-2 in feed Rat liver nuclei TBARS - Ahmed and Ram, 1986T-2 in feed Mouse liver TBARS - Karppanen et al.,1989T-2 in feed Mouse MDA in liver after Vila et al.,2002

    24-48 hrs aftersupplementation

    T-2 in feed Mouse liver DNA CoQ10

    and Atroshi et al.,1997

    fragmentation vitamin ET-2 in feed Quail liver TBARS - Dvorska and Surai, 2001T-2 in feed Rat tissues Conjugate diene - Chang and Mar, 1988T-2 in feed Chicken Liver MDA Lycopene Leal et al.,1999T-2 in feed Chicken, duck, Liver MDA Mezes et al.,1999

    goose

    05-Surai.p65 19/12/2004, 23:2497

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    6/46

    98 Effects of mycotoxins on antioxidant status and immunity

    proteins into the cellular space and inductionof an inflammatory response in the vicinityof the dying cell (Sastre et al., 1996). Incontrast, apoptosis is characterised by cellshrinkage, nuclear pyknosis, chromatincondensation, DNA cleavage into fragmentsof regular sizes and activation of proteasescalled caspases (Dare et al.,2001).

    Reactive oxygen species (ROS) arethought to play a major role in apoptosis(Hockenbery et al.,1993; Ratan et al.,1994),being involved in both initiation andexecution of apoptosis (Herdener et al.,2000). GSH depletion increases thepercentage of apoptotic cells in a givenpopulation; and increased GSH concentrationis shown to decrease the percentage ofapoptosis in fibroblasts (Sastre et al.,1996).In fact, GSH depletion sensitises cells forintracellular induction of apoptosis (Zuckerand Bauer, 1997). Therefore, a decrease inGSH, or an increase in GSSG or perhaps a

    change in the ratio of the two constitutes atrigger for apoptosis (Beaver and Waring,1995). For example ROS from mitochondriacan cause apoptosis after GSH depletion(Zucker et al.,1997). Therefore, apoptosis isinduced by oxidative damage either directlyfrom oxygen free radicals or hydrogenperoxide or from their generation in cells byinjurious agents. For example, hydrogenperoxide is considered a common mediatorfor the apoptosis induced by variousanticancer drugs (Simizu et al.,1998). In linewith those findings there are data showingprotective effects of catalase and SOD fromdifferent inducers of apoptosis (Sandstrom andButtke, 1993; Herdener et al.,2000). Indeed,intracellular induction of apoptosis dependson ROS production and can be efficientlyblocked by antioxidants (Langer et al.,1996;Schaefer et al.,1995).

    As previously mentioned, in many casesmycotoxins decreased cellular GSH, which

    Table 3. Stimulation of lipid peroxidation by aflatoxin.Mycotoxin Tissue Lipid peroxidation Protective effect References

    measurement of antioxidants

    In feed Rat liver TBARS , Se , vitamin E Shen et al.,1994conjugated dienes

    In feed Rat liver MDA , NO melatonin - El-Gibaly et al.,2003loaded chitosan

    In feed Rat liver MDA , NO melatonin Meki et al.,2001In feed Rat testis MDA Vitamin E Verma and Nair, 2001In vitro Cultured rat TBARS , LDH SOD, catalase Shen et al.,1995

    hepatocytes release

    In feed Rat liver TBARS Semecarpus Premalatha et al.,anacardiumnut extract 1997

    In vitro Cultured TBARS ROS Silvia Liu et al.,1999primary rat formation miltorrhizahepatocytes extract

    Intraperitoneally Rat liver TBARS Vitamin E, ternatin Souza et al.,1999In feed Rat liver GSH-Px Se, vitamin E Choi et al.,1995Intraperitoneally Rat liver and TBARS Picroliv Rastogi et al.,2001

    kidney

    Intraperitoneally Rat liver GSH, SOD, - Rastogi et al.,2001bCatalase, GSH-Px

    In vitro Primary rat TBARS , GSH , - Yang et al.,2000hepatocytes ROS generation

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    7/46

    P.F. Surai and J.E. Dvorska 99

    can trigger apoptosis. In general, T-2 toxinis the most potent apoptotic agent among

    mycotoxins. However, there are also reportsindicating apoptosis caused by FB

    1, OTA and

    AFB1. For example, based on the DNA

    fragmentation profile in gel electrophoresisand the morphological changes seen inelectron microscopy, the induction ofapoptotic nuclear changes by variousmycotoxins was investigated in HL-60human promyelotic leukemia cells (Ueno et

    al.,1995). The results showed that T-2 toxin,nivalenol (NIV), DON, OTA, citrinin, AFB

    1

    and some other mycotoxins induced DNAfragmentation. Morphological evidence ofapoptosis was related to the toxicity of themycotoxins and more toxic NIV and DONresulted in more late stage apoptotic eventsthan FB

    1. The results suggested that DNA

    damage and apoptosis rather than plasmamembrane damage and necrosis may beresponsible for the observed cytotoxicity.

    Table 4. Stimulation of lipid peroxidation by fumonisin, deoxynivalenol, aurofusarin and citrinin.

    Mycotoxin Tissue Lipid peroxidation Protective effect Referencesmeasurement of antioxidants

    Fumonisin Rat liver TBARS , DNA Catalase, Sahu et al.,1998nuclei strand breaks mannitol

    Fumonisin B1

    Primary rat TBARS Vitamin E Abel and Gelderblom, 1998hepatocytes

    Fumonisin B1

    Rat liver TBARS - Abel and Gelderblom,1998; Lemmer et al.,1999

    Fumonisin B1

    Phosphati- Rate of - Yin et al.,1998dyl choline peroxidation ,bilayers free radical

    Formation ,Acceleration ofchain formation

    Fumonisin B1

    Vero cells TBARS - Abado-Becognee et al.,1998

    Fumonisin B1

    Glioma cells, TBARS , DNA - Mobio et al.,2003Mouse fragmentationfibroblasts

    Fumonisin B1

    Macrophage MDA - Ferrante et al.,2002cell line

    DON in feed Rat liver TBARS Se, ascorbic Rizzo et al.,1994acid, vitamin E

    DON + Mouse liver TBARS - Karppanen et al.,1989 3-AcDON

    in feedAurofusarin Quail egg yolk TBARS - Dvorska et al.,2001b, in feed Dvorska, 2001

    Aurofusarin Liver of newly TBARS - Dvorska et al.,2002 in feed hatched quail Dvorska and Surai, 2004

    Zearalenone Rat liver TBARS - Karagezyan et al.,1995injected IV

    Zearalenone Vero cells TBARS Vitamin E Ghedira-Chekir et al.,1999 in vitro

    Citrinin in feed Rat liver TBARS - Ribeiro et al.,1997

    05-Surai.p65 19/12/2004, 23:2499

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    8/46

    100 Effects of mycotoxins on antioxidant status and immunity

    Fumonisin B1, T-2 toxin, fusarenon-X andDON could induce apoptosis of liver cells,kidney cells, gastrointestinal epithelial cellsand immunological cells as well as severalcell lines. The possible mechanisms ofapoptosis induction are not well understood.It seems likely that sphingol, p21 gene,protein kinase and intracellular Ca2+ levelmight be involved in the cellular apoptosisinduction by Fusariummycotoxins (Wang andZhang, 2000).

    T-2 TOXIN

    The rank order of the potency of trichothecenemycotoxins to induce internucleosomal DNAfragmentation was found to be T-2, satratoxinG, roridin A >> diacetoxyscirpenol >baccharin B-5 >> nivalenol, deoxynivalenol,3-acetyldeoxynivalenol, fusarenon-X, baccharinB-4 vehicle control (Nagase et al.,2001). Theauthors showed that T-2-induced apoptosisinvolved activation of caspase-3 throughcytosolic accumulation of cytochrome c alongwith caspase-9 activation. Both T-2 and HT-2

    induced apoptosis after 24 hrs in HL-60 cells,with T-2 being somewhat more potent thanHT-2 and the apoptotic process was almostcompletely blocked in the presence of acaspase inhibitor (Holme et al., 2003).Development of apoptosis and changes inlymphocyte subsets were examined inthymus, mesenteric lymph nodes and Peyerspatches of mice up to 24 hrs after oralinoculation with T-2 toxin (10 mg/kg). Thedegree of lymphocyte apoptosis wasprominent in the thymus, moderate in thePeyers patches, and somewhat mild in the

    mesenteric lymph nodes (Nagata et al.,2001). Cytocentrifugation and lightmicroscopy of leukocyte-enriched cellsamples from the pronephros (the primaryhematopoietic compartment in the fish)demonstrated T-2-related increases inapoptotic bodies and the apoptotic probesconfirmed apoptosis in fish (Gogal et al.,2000).

    Female mice were treated orally with T-2toxin (10 mg/kg BW) and thymus and spleenwere examined to detect apoptotic changes(Shinozuka et al., 1997). The number ofapoptotic lymphocytes in the thymusincreased dramatically from 9 to 24 hrs aftertreatment and began to increase in the spleenat 12 hrs after treatment indicating thatthymus was more sensitive to apoptosis thanspleen. In another experiment, mice weregiven 5 mg/kg BW of T-2 toxin and werekilled 12 hrs later (Ihara et al.,1997). Massivecellular destruction was observed in the

    thymus and spleen and electron microscopyrevealed the presence of apoptotic bodies.Similarly, in the liver of mice given 2.5 mg/kg T-2 toxin and killed 2 hrs later, theinduction of apoptotic cellular lesions wasobserved. Electron microscopic characteristicsof damaged lymphocytes were shrinkage ofthe cell body, nuclear chromatin condensationand fragmentation (Li et al.,1997). Of ninetrichothecene mycotoxins tested, T-2 toxinwas the most potent agent to induce apoptosisin the thymus in young female mice (Islamet al.,1998; 1998a). It is interesting that both

    the acetyl group at the C-4 position and theisovaleryl or 3-hydroxyisovaleryl group atthe C-8 position of the T-2 toxin moleculeare important for inducing apoptotic changesin the thymus (Islam et al.,1998). T-2 toxinwas shown to induce apoptosis in thehematopoietic (bone marrow) tissues offemale mice (Shinozuka et al., 1998). Itshould be noted that the gastrointestinal tractis also sensitive to trichothecene-inducedapoptosis, since such changes were observedin the gastric mucosa, gastric glandularepithelium and intestinal crypt cell

    epithelium (for review see Bondy and Pestka,2000). These changes could impair nutrientassimilation as well as decrease defencesagainst enteric pathogens and endotoxin.

    To examine the effect of T-2 toxin on thedeveloping embryo, pregnant mice weredosed orally with T-2 toxin (3 mg/kg BW) at11 days of gestation (Ishigami et al.,1999).Ultrastructural changes characteristic of

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    9/46

    P.F. Surai and J.E. Dvorska 101

    apoptosis were observed. For example, in somelayers of the central nervous system moderatepyknosis or karyorrhexis was seen. Using DNAfragmentation and fluorescence microscopyassays Yang et al. (2000a) demonstrated thatrank order of trichothecene-mediated apoptosisis similar to their cytotoxicity. Therefore,cytotoxicity of trichothecene mycotoxins ismediated through an apoptotic process. Inparticular, the embryotoxic effect of T-2 toxinon the mouse fetus is mediated throughapoptosis. T-2 toxin (2 mg/kg BW) was givenorally to pregnant mice at various gestational

    days and the fetuses were examined 24 hrs later.T-2 toxin-induced apoptosis was detected. SinceT-2 toxin readily crosses the placenta, T-2 toxincould directly cause fetal apoptosis (Ishigamiet al.,2001).

    It was shown that in human promyeloticcell line HL-60, T-2 toxin induced apoptosisat 10 ng/ml levels within 2-6 hrs (Ueno etal.,1995). Apoptosis in HL-60 cells inducedby T-2 toxin was dose dependent when cellswere treated with concentrations of 5-100 ng/ml for more than 2 hrs (Yoshino et al.,1996).The authors suggested that the Ca2+ signal

    triggered by T-2 toxin is transduced by theactivation of endonuclease and protease, andultimately evokes apoptosis.

    Molecular mechanisms of T-2 stimulationof apoptosis are not clear at present.However, recently it has been shown thattrichothecene mycotoxins trigger a ribotoxicstress response that activates the stress-activated kinases c-Jun N-terminal kinase and/or p38 mitogen-activated kinase and inducesapoptosis (Shifrin and Anderson, 1999). Suchactivation may signal cell survival or induceapoptosis in various cell types depending on

    the conditions, e.g. length of signal.However, the pathway leading from theactivation of these kinases to the activationof caspases and apoptosis has not beenelucidated and needs further research.Albarenque et al. (2001) suggested that theelevation of TNF-mRNA expression mayplay an important role in T-2 toxin-inducedepidermal cell apoptosis. The induction of c-

    fos and perhaps of c-jun mRNAs may beassociated with T-2 toxin-induced epidermalcell apoptosis (Albarenque et al.,2001).

    FUMONISIN B1

    The effects of FB1on apoptosis in various in

    vitro and in vivo studies have receivedsubstantial attention over the last few years.For example, FB

    1-treated keratinocytes

    developed morphological features consistentwith apoptosis. In particular, they released

    nucleosomal DNA fragments into themedium 2-3 days after exposure to 0.1 mMFB

    1and showed increased DNA strand breaks

    (Tolleson et al., 1996). FB1 induced a

    significant and dose-related increase ofmodifications of DNA bases (8-OH-dG) andDNA fragmentation in both C6 glioma andmouse embryonic fibroblasts cells (Mobio etal.,2003). In the same study, apoptotic C6glioma cells were also observed after FB

    1

    incubation. Recently genes that inhibit FB1-

    induced apoptosis in African green monkeykidney fibronlasts (CV-1 cells) and two mouse

    embryo fibroblasts have been identified (Joneset al., 2001). Primary mouse embryofibroblasts underwent apoptosis following FB

    1

    treatment (Ciacci-Zanella et al.,1999). Theauthors also demonstrated that the tumournecrosis factor (TNF) pathway and caspasesplay an important role in FB

    1-induced

    apoptosis. Effects of exposure of humanfibroblasts to FB

    1were investigated. After 72

    hrs of treatment, FB1 (50 and 100 mM)

    induced DNA damage, an enhancement ofcaspase-3-activity and cleavage of poly(ADP-ribose)polymerase compared to controls

    (Galvano et al.,2002).The cytotoxicity and genotoxicity of FB

    1

    in rabbit kidney RK13 cells were examined.Exposure to FB

    1caused a significant increase

    in micronucleus frequency in a concentration-and in a time-dependent manner and anincreased number of the cells were dying bythe process of apoptosis (Rumora et al.,2002).The author suggested that nanomolar

    05-Surai.p65 19/12/2004, 23:24101

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    10/46

    102 Effects of mycotoxins on antioxidant status and immunity

    concentrations of FB1 induced apoptosis,which subsequently may proceed tosecondary necrosis. Treatment of monkeykidney cells (CV-1 cells) with FB

    1led to cell

    cycle arrest and apoptosis (Zhang et al.,2001). It was suggested that the ability ofFB

    1 to alter gene expression and signal

    transduction pathways may be necessary forits carcinogenic and toxic effects.

    In general, FB1 induces apoptosis of

    hepatocytes and of proximal tubule epithelialcells. More advanced lesions in both organswere characterised by simultaneous cell loss

    (apoptosis and necrosis) and proliferation(Voss et al., 2001). A dose-dependentincrease in TNF-induced apoptosis wasobserved in porcine renal epithelial cellspretreated with FB

    1. In particular, cells treated

    with FB1 showed increased DNA

    fragmentation and terminal uridine nucleotideend labeling in response to TNF-treatment(Johnson et al.,2003). In the same study, FB

    1

    also increased DNA synthesis and resultedin cell cycle arrest in the G2/M phase of thecell cycle.

    FB1toxicity caused induction of cytokine

    networks in liver with involvement of theTNF- signaling pathway (Bhandari andSharma, 2002). The authors suggested thatincreased expression of caspase 8 involvedin the TNF- signaling pathway maycontribute to the apoptosis, whereas IL-1Rainduction could contribute to the proliferatingeffects observed in FB

    1 toxicity. Similarly,

    FB1 caused an activation of the cytokine

    network in liver, particularly the TNF-signaling pathway, suggesting its involvementin hepatotoxic mechanisms (Bhandari et al.,2002). Induction of apoptosis was suggested

    to be a consequence of ceramide synthaseinhibition and disruption of sphingolipidmetabolism by FB

    1(Dragan et al.,2001). It

    seems likely that the elevated endogenoussphinganine acts as a contributing factor tothe fumonisin-induced cell death (Yu et al.,2001). Moreover recent results of Mobio etal . (2000) showed that cytotoxicconcentrations of FB

    1 induce cellular cycle

    arrest in phase G(2)/M in rat C6 glioma cells.Furthermore, under experimental conditions(9 or 18 mM FB

    1in the medium) there was

    an induced DNA fragmentation and ladderingand many apoptotic bodies in glioma cells.

    As mentioned above, FB1 caused

    morphological changes (i.e., cell shrinkage,membrane blebbing) and time-dependentincreases in DNA fragmentation characteristicfor apoptosis. For example, Kim et al. (2001)demonstrated that FB

    1kills LLC-PK(1) kidney

    cells by inducing apoptosis. This includedinitial disruption of sphingolipid metabolism

    and accumulation of sphinganine (or ametabolite), which, in turn, inducedexpression of calmodulin. FB

    1, but not AFB

    1,

    induced the apoptosis of swine alveolarmacrophages (AM) with evidence of DNAladdering and nuclear fragmentation.However, both FB

    1 and AFB

    1 exposure

    induced the expression of apoptosis-relatedheat shock protein 72 in AM (Liu et al.,2002).

    FB1has been shown to cause apoptosis in

    a variety of cell types and tissues, but theapoptotic potential of other fumonisins and

    fumonisin metabolites could substantiallyvary. For example, Seefelder et al. (2003)exposed human proximal tubule-derivedcells (IHKE cells) to FB

    1, FB

    2, FB

    3, hydrolyzed

    FB1 and N-palmitoyl-hydrolyzed FB

    1 and

    investigated caspase-3 activation, chromatincondensation and DNA fragmentation. Theresults demonstrated that all compounds ledto increased sphinganine levels in IHKE cells,but only FB

    1was able to induce apoptosis. In

    the short-term studies, increases in tissueconcentration of the ceramide synthase substratesphinganine and the sphinganine:sphingosine

    ratio were correlated with apoptosis (Voss etal.,2002).

    Although it has been demonstrated thatFB

    1induces apoptosis in many cell lines, the

    precise mechanism of apoptosis is not fullyunderstood. For example, FB

    1-induced

    apoptosis involves the activation of caspase3, which is associated with the repression ofprotein kinase C and possibly its downstream

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    11/46

    P.F. Surai and J.E. Dvorska 103

    effectors, NF-B and TNF- (Gopee et al.,2003).After intravenous injection of male rats

    with FB1(1.25 mg/kg), hepatic and renal GSH

    concentrations were depressed andproliferation and apoptosis were observed inthe outer medulla of the kidney cell (Lim etal.,1996). When male mice were injectedwith FB

    1subcutaneously at doses of 0, 0.25,

    0.75, 2.25 and 6.75 mg/kg BW daily for 5days, a dose-dependent increase in apoptosisin liver and kidney was observed (Tsunodaet al., 1998). Similar observations were

    described by Sharma et al. (1997). Male micewere injected subcutaneously with vehicleor 2.25 mg/kg/day of FB

    1 for 5 days and

    sampled 1 day after the last treatment. FB1

    increased apoptotic cells in liver (Bhandariet al., 2002). Similarly, histopathologyrevealed the occurrence of apoptosis in theliver of rats exposed to FB

    1 (Pozzi et al.,

    2001).Multiple daily doses of FB

    1after surgery

    elevated the number of apoptotic hepatocytesin rats (Li et al.,2000). Including FB

    1in the

    diets of rats increased hepatocellular and renal

    tubule epithelial cell apoptosis (Howard etal., 2001). It is interesting that female ratsdemonstrated more sensitivity than male ratsin the induction of hepatocellular apoptosisand mitosis. Mild toxic effects, includingapoptosis, proliferation of bile duct epithelialcells, and early signs of fibrosis were noticedin the liver of rats fed FB

    1(25 mg/FB

    1/kg diet;

    Gelderblom et al.,2001). Six hours afteradministration of FB

    1, marked morphologic

    changes of rat hepatocytes included theappearance of small vacuoles along the marginof the cell membrane. Electron microscopic

    analysis revealed margination of nuclearchromatin and swollen mitochondria withamorphous matrical deposit (Moon et al.,2000). The authors suggested that FB

    1-induced

    alteration of hepatocyte membrane lipidcomposition is an early key event in the FB

    1

    apoptotic effect. Therefore, the induction oftubular epithelial cell apoptosis was theprimary response of the kidneys to dietary

    FB1. Apoptosis was present at all doses (99-484 ppm for 28 days) in the kidney of malerats and occurred in females at doses of 153-484 ppm FB

    1(Tolleson et al.,1996). Wistar

    rats were fed diets containing 0 (control) or100 ppm FB

    1 for 12 weeks. Necrosis and

    apoptosis of tubular epithelial cells in thekidney were observed and increased mitoticfigures and lymphocytic infiltrate in the smallintestine were found (Theumer et al.,2002).Increased hepatocellular apoptosis was alsodetected in the mice consuming dietscontaining 72 and 143 mmol/kg FM

    1(Howard

    et al.,2002).FB

    1 can induce renal injury and organ

    sphingolipid alterations in cattle. Forexample, renal lesions in FB

    1-treated calves

    consisted of vacuolar change, apoptosis,caryomegaly, and proliferation of proximalrenal tubular cells, as well as dilation ofproximal renal tubules, which containedcellular debris and protein (Mathur et al.,2001). Probably there is species-specificityin susceptibility to FB

    1. For example, FB

    1was

    investigated in four groups of growing ducks,each receiving 0, 5, 15 or 45 mg/kg FB

    1by

    daily oral administration over 12 days. Nosign of apoptosis was present in the liver orin peripheral blood lymphocytes and onlymoderate oxidative damage was noted (Baillyet al.,2001).

    The apoptosis-stimulating effect of FB1has

    been clearly demonstrated; howevermolecular mechanisms of this stimulation arenot clear at present. Nevertheless, stimulationof lipid peroxidation by FB

    1and decreased

    antioxidant concentrations including GSH intissues could lead to changes in redox statusof the cell and trigger a cascade of apoptotic

    changes.

    OCHRATOXIN A

    Lipid peroxidation due to OTA consumptionmay be implicated in DNA damage. When aspecially designed oxidation system was used,OTA was found to facilitate single-strand

    05-Surai.p65 19/12/2004, 23:24103

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    12/46

    104 Effects of mycotoxins on antioxidant status and immunity

    cleavage of supercoiled plasma DNA throughROS production (Gillman et al.,1999). Ingeneral, OTA is implicated in DNA-adductformation (Grosse et al., 1997). OTAtreatment of rats resulted in a 5-fold increasein the expression of the protein haemoxygenase-1 generated during oxidative stress(Gautier et al.,2001). Changes in redox statusof the cell due to the pro-oxidant action ofOTA could trigger cell apoptosis. Forexample, OTA at a dose of 20 mg/kg causednuclear changes characteristic of apoptosis inhamster kidney (HaK) and HeLa cells

    (Seegers et al., 1994). Furthermore, theresults indicated that OTA might activatedifferent cellular processes involved in thedegradation of DNA in HaK and HeLa cells.Exposure of human proximal tubule-derivedcells to 30 nmol/L or more OTA led to DNAfragmentation and chromatin condensation(Schwerdt et al.,1999). It was concluded thatexposure to low OTA concentrations can leadto direct or indirect caspase-3 activation andsubsequently to apoptosis in cultured humanproximal tubule cells and in other renalepithelial cell lines of different origins.

    Administration of OTA twice a week forone or two weeks resulted in apoptosis inthe liver of mice (Atroshi et al., 2000). Inparticular, the presence of intracellularapoptosis bodies was detected two weeksafter toxin treatment. Light microscopicexamination demonstrated the presence ofeosinophilic globules, often containingapoptotic bodies. They were found within thecytoplasm of intact hepatic cells. The numberof apoptotic bodies was further enhanced attwo weeks, resulting in an 8-fold increase inliver over the control values (Atroshi et al.,

    2000). Wistar rats were treated with a lowdose (5 mM and 1 mg/kg, respectively) or ahigh dose (12.5 mM and 10 mg/kg,respectively) of OTA for 24 or 72 hrs. Markedtreatment-specific transcriptional changeswere detected for genes involved in DNAdamage response and apoptosis (Luhe et al.,2003). Because of OTA consumption, therewas a striking increase in the counts of

    eosinophils and of apoptotic phagocytes inweaner pigs in a dose-dependent manner(Muller et al.,1999).

    Nanomolar concentrations of OTApromote apoptosis in a cell-type specificfashion. For example, OTA, at noncytotoxicdoses, was able to detach collagen- andfibronectin-adherent cells from immobilizedsubstratum causing apoptosis as measured bycaspase-3 activation (Scibelli et al., 2003).Relatively low OTA concentrations (547.2,752.5 and 930.3 ng OTA/g kidney tissue)have activated apoptotic processes and

    oxidative damage in kidney cells (Petrik etal.,2003). In human monocyte/macrophageline THP-1 metabolic activity, cellproliferation, cell membrane integrity, celldifferentiation, phagocytic behaviour, NOsynthesis and cell surface markers werelargely suppressed by OTA at concentrationsbetween 10 and 1000 ng/ml (Mulleret al.,2003). OTA greatly induced apoptosis inhuman myc-transfected cell line (Horvath etal.,2002).

    Exposure to low OTA concentrations (5-30 nmol/L) can lead to time- and

    concentration-dependent direct or indirectcaspase-3 activation and subsequently toapoptosis in cultured human proximal tubulecells and in other renal epithelial cell linesof different origins (Schwerdt et al.,1999).Additional evidence was provided indicatingthat OTA interacts in a cell type-specific waywith distinct members of the mitogen-activated protein kinase family providinginduction of apoptosis via the c-jun aminoterminal kinase pathway (Gekle et al.,2000).

    OTHER MYCOTOXINS

    When thein vitro effect of deoxynivalenol(DON) on apoptosis in specific T- and B-cellsubsets within thymus, spleen and Peyerspatch cultures was studied (Pestka et al.,1994) it was shown that depending onlymphocyte subset, tissue source andglucocorticoid induction, DON could either

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    13/46

    P.F. Surai and J.E. Dvorska 105

    enhance or inhibit apoptosis. Combinedtreatment of mice with DON andlipopolysaccharide significantly increased theamount of apoptotic thymic and splenic tissue(Zhou et al., 2000). It was suggested thatinhibition of protein synthesis and inductionof apoptosis are the main mechanisms ofDON toxicity in intestinal cells (Maresca etal.,2002).

    It seems likely that DON can interact withother inducers of apoptosis to increaseapoptotic action. As demonstrated by DNAfragmentation and flow cytometric analysis,

    apoptosis in thymus, Peyers patches, andbone marrow was marked in mice 12 hrs afteradministering Escherichia coliLPS (0.1 mg/kg BW i.p.) concurrently with DON (12.5 mg/kg BW p.o.), whereas apoptosis in controlmice or mice treated with either toxin alonewas minimal (Islam et al.,2002). Therefore,lipopolysaccharide can interact with DON inmice to induce the glucocorticoid-drivenapoptotic loss of immature thymocytes andcytotoxic T-lymphocytes in thymus, mature-B-lymphocytes in Peyers patch, and pro/pre-B-lymphocytes and mature B-lymphocytes in

    bone marrow in mice (Islam et al.,2003).Dead lymphocytes in the thymus, spleen

    and Peyers patches showed ultrastructuralcharacteristics of apoptosis. DON andaflatoxin G

    1 (AFG

    1) could induce and

    accelerate apoptosis in human peripheralblood lymphocytes. For example, a typicalsub-diploid apoptosis peak was demonstratedin lymphocytes treated with DON and AFG

    1

    (Wang et al.,1999). In particular, a significantdose response effect and time effectcorrelation was found between apoptosis ratesand mycotoxin concentrations and the treated

    time.In an experiment with male rats, AFB

    1was

    administered i.p. or intratracheally at 2-8 mg/kg BW and animals were killed after 26 hrs(Raj et al.,2001). Dose-dependent inductionof micronuclei in bone marrow and lung cellswas observed. Furthermore, apoptotic bodieswere found in lung cells of rats given AFB

    1,

    which was correlated with micronucleiinduction. Clearly, AFB

    1induced micronuclei

    and apoptosis in lung and bone marrow cells.This stimulation could be associated with thestimulation of the phosphatidylinositol (PI)cycle and activation of PI kinase by AFB

    1

    (Pasupathy et al.,1999). Apoptotic rates inrat liver were significantly reduced whenmelatonin was co-administered with AFB

    1(El-

    Gibaly et al.,2003). Rats were treated withAFB

    1(50 mg/kg BW) for 8 weeks. The levels

    of caspase-3 activity in the AFB1group were

    significantly higher than in controls and the

    apoptosis was associated with degenerativeand necrotic changes in the hepatocytes(Meki et al.,2001a). In the study, caspase-3activity was positively correlated with MDAwhile negatively correlated with GSH, GSH-Px and GSH-reductase in rat livers treatedwith AFB

    1. It is interesting that melatonin

    treatment of rats could enhance hepaticantioxidant/detoxification system, whichconsequently reduced the apoptotic rate andthe necrobiotic changes in the liver.

    It seems likely that apoptosis is theprincipal mechanism contributing to germ

    cell depletion and testicular atrophy followingzearalenone (ZEA) exposure. In particular,Kim et al. (2003) demonstrated that a singledose of ZEA (5 mg/kg) induced testiculargerm cell apoptosis in a time-dependent andstage-specific pattern. In another study, ZEA(10, 20 and 40 mM) induced concentration-dependent DNA fragmentation in three celllines (Vero, Caco-2 and DOK) resulting inDNA laddering patterns on agarose gelelectrophoresis. This observation wasconsistent with apoptosis, which wasconfirmed by observations of formation of

    apoptotic bodies. Furthermore, ZEA inducedcell cycle arrest in the three cell linescharacterised by an increase in the numberof cells in the G2/M phase of the cell cycle(Abid-Essefi et al.,2003). It is interesting thatvitamin E (25 mM) added simultaneously withZEA partially reduced DNA fragmentation andapoptotic body formation after 24 hrsincubation.

    05-Surai.p65 19/12/2004, 23:24105

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    14/46

    106 Effects of mycotoxins on antioxidant status and immunity

    Mycotoxins and the immune system

    All animals protect themselves from invasionby microorganisms, parasites, fungi, viruses andany foreign molecules. This protective capacityrequires an effective immune system, which isan important determinant of animal health andwell being. In that sense, the remarkable abilityof the immune system to distinguish betweenself and non-self is a great achievement ofevolution. Commercial animal production isbased on balanced feed, providing nutrientrequirements and optimised environmental

    conditions. However, it is very difficult to avoidvarious nutritional or environmental stresses,which are responsible for immunosuppressionand increased susceptibility to various diseasesand consequently decreased productive andreproductive performance of farm animals. Inthis respect mycotoxins are one of the mostimmunosupressive factors in animal diets.

    There are two major types of immunefunction: natural and acquired immunity (Figure2). Natural immunity, called the innate immunesystem, includes physical barriers (e.g. skin,mucus layer of the gastrointestinal tract), specific

    molecules such as agglutinins, precipitins, acutephase proteins and lysozyme, phagocyticdestruction of pathogens by macrophages andneutrophils, and lysing activity by lymphocytescalled natural killer (NK) cells.

    Macrophages perform a range of functions,including phagocytosis of foreign particles,destruction of bacterial or tumor cells,secretion of prostaglandins and cytokines,and as a result regulating activity oflymphocytes and other macrophages(Qureshi, 1998). In fact, phagocytosis is themajor mechanism by which microbes are

    removed from the body and is especiallyimportant for defence against extracellularmicrobes. As a result of stimulation (forexample by microbes) monocytesdifferentiate into macrophages that are morepowerful in mediating host defence (Klasing,1998a).

    Macrophage activation and phagocytosisof foreign particles are regularly accompaniedby a respiratory burst, an increase in theproduction of reactive oxygen species (ROS),exerted by the enzyme complex NADPHoxidase. Therefore macrophages as well as

    Immunity

    Natural (innate) Aquired (adaptive, specific)

    Physical

    barriers Phagocytes

    Specific

    moleculeHumoral Cell-mediated

    Macrophages Neutrophils

    NK cells Mastcells

    Dendritic

    cells

    ROS, RNS,Small peptides

    Elcosanoids

    Cytokines, Complement

    Acute phase proteins

    Kill pathogen

    B-lymphocytes T-lymphocytes

    Immunoglobulins

    (antibodies)

    Direct contact

    with target cells

    Pathogen removal and resistance

    to pathogens (immunologic memory)

    Inflammatory

    response

    Figure 2. General overview of the immune system.

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    15/46

    P.F. Surai and J.E. Dvorska 107

    other phagocytic leukocytes (e.g.,neutrophils, monocytes and eosinophils) cansynthesize toxic oxygen metabolites such assuperoxide anion (O

    2-), hydroxyl radical

    (OH), singlet oxygen (1O2), hydrogen

    peroxide (H2O

    2), nitric oxide (NO) and

    peroxinitrite (ONOO-) during the respiratoryburst (Zhao et al., 1998). For example, abacterium coming into contact with theplasma membrane is enclosed in a plasmamembrane vesicle containing NADPHoxidase, and exposed to an intensive flow ofsuperoxide radical (Giller and Sigler, 1995).

    Superoxide radical can dismutate to H2O2,which penetrates the bacterium withproduction of hydroxyl radical, which isultimately deadly to biological molecules.In general, the production of ROS andreactive nitrogen species (RNS) is acharacteristic of both mammalian and avianmacrophages (Qureshi et al.,1998a).

    In general ROS, RNS and eicosanoids (e.g.leukotrienes and prostaglandins) haverecently received substantial attention asmajor metabolites produced by macrophages(Dietert and Golemboski, 1998). Because of

    this powerful weapon, macrophages bind,internalize, and degrade foreign antigens(e.g. bacteria) quite quickly. It takes only 15minutes for chicken macrophages to kill morethan 80% of the internalized Salmonella(Qureshi et al., 1998a). Therefore naturalimmunity works rapidly, and gives rise to theacute inflammatory response. Macrophagescontain various substances (includingenzymes producing free radicals and smallpeptides with antibiotic activity) involved inmicrobial killing. They also have receptorsfor chemoattractive factors released from

    microbes. In addition to ROS, RNS andeicosanoids mentioned above, macrophagesalso synthesize and secrete a great numberof cellular communication molecules such ascytokines, including the pro-inflammatorycytokines interleukin 1 (IL-1), interleukin 6(IL-6), and TNF-. They also produce cytokineinhibitors, hormones and neurotransmitters(Klasing, 1998a), which regulate specific

    immunity (initiating and directing theimmune and inflammatory responses) andmany other related physiological responses.Therefore macrophages are importantamplifiers of the immune response, both bycytokine production and by serving to presentparasite derived peptides to T-cells.

    Acquired or specific immunity consists ofhumoral and cell-mediated immunity. Thereare two major types of lymphocytes, B-cellsand T-cells. Humoral immunity is mediatedby antibodies that are released by B-cells intothe bloodstream. The bursa of Fabricius is the

    site of B-lymphocyte development anddifferentiation in birds. This immunity isbased on the production of immunoglobulins.They are responsible for recognition andelimination of specific antigens: they bindand remove from the host invading organisms/substances.

    Cell-mediated immunity is based onspecific antigen recognition by thymus-derived T-lymphocytes. Due to this immunitycells infected with a foreign agent, forexample virus, are destroyed via directcontact between an activated T-cell and the

    target (infected) cell (Qureshi et al.,1998).Cell-mediated immunity is responsible fordelayed-type hypersensitivity (DTH)reactions, foreign graft rejections, resistanceto many pathogenic microorganisms andtumor immunosurveillance (Wu andMaydani, 1998).

    In birds, both T-cell and B-cell precursorsoriginate in the bone marrow. Actualdevelopment of T-cells takes place in thethymus and B-cells develop in the bursa ofFabricius in birds and in bone marrow ofmammals (Lahtala, 1998). Interactions

    between T- and B-cells, as well as antigenpresenting cells, are responsible for thedevelopment of specific immunity. Thedefence mechanisms of specific immunity areinduced or stimulated by exposure to foreignsubstances, are specific for distinctmacromolecules and increase in magnitudewith each successive exposure to a particularmacromolecule (Miles and Calder, 1998). In

    05-Surai.p65 19/12/2004, 23:24107

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    16/46

    108 Effects of mycotoxins on antioxidant status and immunity

    comparison to natural immunity, specificimmunity takes longer to develop, but ishighly specific for antigens and has memory.These two parts of the immune system worktogether through direct cell contact andthrough interactions involving such chemicalmediators as cytokines and chemokines.Therefore, the animal immune systemrequires the co-operation of macrophages,bursa-derived B-lymphocytes and thymus-derived T-lymphocytes with various othertypes of cells.

    The immune response involves cellular

    proliferation (T-lymphocytes), enhancedprotein synthesis (including immunoglobulinsynthesis by B-lymphocytes and acute phaseprotein synthesis in the liver) andinflammatory mediator production.Physiological changes resulting fromstimulation of the immune system includefever, anorexia and loss of tissue components(Grimble, 1997).

    When analysing data related toimmunomodulation careful attention must bepaid to methods used to assess immunologicalfunctions. For example, in vivomethods of

    immune function assessment are based on twomain approaches: antibody response tovaccine or delayed-type hypersensitivity(DTH) reactions. In the first method,immunisation with appropriate antigens (viralor bacterial) can elicit serum antibodies. Thehaemagglutination (HA) assay measuresserum antibody concentration (titre) againstantigens, which are often sheep red bloodcells (SRBC). This assay provides informationabout humoral immunity (B-cellresponsiveness) and its association with cell-mediated immunity (T-cell co-operation). The

    second (DTH) method is used to assess cell-mediated immune function.

    In vitro indices of immune functioninclude (Wu and Meydani, 1998):

    Lymphocyte proliferation assay. This assayprovides information about cell-mediatedimmune response and consists ofmeasuring the number of cells in culture

    with or without addition of a stimulatoryagent (mitogen). In this assay, isolatedlymphocytes are incubated with mitogens,which activate division of either T-or B-lymphocytes. Various mitogens are usedin such assays, but most often they includeconcanavalin A (con A, a T-cell mitogen),phytohemagglutin (PHA, a T-cellmitogen), lipopolysaccharide (LPS, a B-cell mitogen) and pokeweed mitogen(PWM, T- and B-cell mitogen) (Hayek etal., 1996). Decreased proliferation mayindicate impaired cell-mediated

    immunity. Cytokine production. T-cells produce a

    range of protein mediators calledcytokines, which regulate cell activation,growth, differentiation, inflammation andimmunity.

    Cytotoxicity assay. This assay assessesactivity of cytotoxic T-lymphocytes (agroup of T-cells that kill other cells byrecognising their cell-surface antigens)and NK cells (a group of non-T and non-Blymphocytes that kill virus-infected andtumour cells).

    Flow cytometric analysis. This assayidentifies cells with different surfacemarkers. The results can be used forunderstanding the cellular basis ofimmune response.

    Plaque-forming cell (PFC) test. This showsthe number of antibody-producing cells.

    The impact of the avian immune system inmodern production cannot be overestimated.The banning of feed grade antibiotics in Europehas made immunocompetence the major factordetermining efficiency of poultry production.

    Molecular immunology is developing veryquickly; and mechanisms and factors affectingavian immunocompetence have recentlyreceived substantial attention (McCorkle, 1998;Saif and Swayne, 1998) and nutritionalmodulation of resistance to infectious diseasesin poultry (Klasing, 1998) is a frontline for futureresearch. It must be emphasised that cellularintegrity is very important for receiving and

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    17/46

    P.F. Surai and J.E. Dvorska 109

    responding to the messages needed to co-ordinate an immune response (Latshaw, 1991).Therefore, the antioxidant/pro-oxidant balanceof the host is a critical consideration in optimalimmune system function.

    The clinical toxicologic syndromes causedby ingestion of mycotoxins have beencharacterized in domestic animals, poultryand laboratory animals and range from acutemortality to decreased performance andimmunosuppression. In fact, consumption ofcertain mycotoxins, at levels that do notcause overt clinical mycotoxicosis, suppresses

    immune function and decrease resistance toinfectious disease (Corrier, 1991). The effectsof several mycotoxins on immune responseshave been investigated (Tables 5-9);however, most data were obtained withlaboratory animals. In some instances, farmanimals and cells derived from livestockspecies have been employed to evaluate theimmunotoxicity of mycotoxins (Sharma,1993; Bondy and Pestka, 2000).

    Immunosupressive potency of variousmycotoxins differs substantially. Effects of DON,3-acetyldeoxynivalenol, fusarenon-X, T-2 toxin,

    ZEA, zearalenol, -zearalenol and NIV onT- and B-cells were studied using a proliferationassay and antibody-dependent cellular cytotoxicNK cell activity on human peripheral bloodmononuclear cells (Berek et al., 2001). Themycotoxin concentrations used in theexperiments were comparable to those foundin normal human peripheral blood (0.2-1800ng/ml). Among the mycotoxins tested, T-2toxin, fusarenon-X, NIV and DON showed thehighest immunosuppressing effects in vitro. Inparticular, mycotoxin-induced immuno-suppression was related to depressed T- or

    B-lymphocyte activity. Furthermore, they alsoinhibited NK cell activity (Berek et al.,2001).

    As can be seem from Table 5,immunosuppression caused by AFB

    1has been

    demonstrated in various livestock species(e.g., chickens, turkeys, pigs and lambs) andalso in laboratory animals (mice and rats) andin various in vitro systems. Aflatoxin is animmunomodulating agent that acts primarily

    on cell-mediated immunity and phagocyticcell function. Therefore, AFB

    1 mainly

    decreases lymphocyte activity and may affectmacrophages assisting lymphocyte function.For example, in macrophages exposed toAFB

    1, NO production stimulated by LPS

    significantly decreased (Moon et al.,1998).Furthermore, AFB

    1 decreased pro-

    inflammatory and increased anti-inflammatory cytokine mRNA expression inweanling piglets (Marin et al.,2002). AFB

    1

    can be transferred from chicken to the eggand further to the developing embryo.

    Therefore, the progeny of hens consumingan AFB

    1-contaminated diet may be

    increasingly susceptible to disease owing tosuppression of humoral and cellularimmunity. For example, anti-Brucella abortusantibody production was compromised andROS production by macrophages from AFB

    1

    progeny decreased (Qureshi et al., 1998).Long-term immune depression of macrophage-mediated functions can occur followingembryonic exposure to AFB

    1(Neldon-Ortiz and

    Qureshi, 1992). Furthermore, depressedcomplement and interferon production could

    also result from AFB1 exposure. Therefore,acquired immunity from vaccination programsmay be substantially suppressed; and in suchcases the signs of disease observed are thoseof the infectious process rather than those ofthe aflatoxin that predisposed the animal toinfection. Mixtures of aflatoxin with othermycotoxins can result in greatly augmentedbiological responses in terms of weight gaindepression, lethality, and immune reactivity(Pier, 1992).

    The immunomodulatory effects ofochratoxins have also been considered for

    many years and a summary is shown in Table6. OTA has been shown to affect mainlyhumoral immune function at the level ofantibody synthesis in chickens, rats and mice.However, number and phagocytic activity ofmacrophages were also decresed in growinggilts receiving OTA for 35 days (Harvey et al.,1992). IL production was also compromised. Ithas been demonstrated that exposure of purified

    05-Surai.p65 19/12/2004, 23:24109

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    18/46

    110 Effects of mycotoxins on antioxidant status and immunity

    Table 5. Effects of aflatoxin B1on immunity.

    Species Dose Effects on immune system Reference

    Broiler chicks 1 mg/kg, 7-49 days Titres to NK Shivachandra et al.,2003Broiler chicks 2.5 mg/kg diet for Peripheral T-lymphocyte counts , Celik et al.,2000

    21 days Splenic plasma cell counts Broiler chicks 2.5 mg/kg diet for Percentage/mean phagocytic Ibrahim et al.,2000

    21 days activity ; Immune responsemeasured by HI test

    Broiler breeder 0.2; 1; 5 or In progeny, anti-Brucella abortus Qureshi et al.,1998 hens 10 mg/kg titres ; Phagocytosis of SRBC

    and ROS production bymacrophages in 5 mg AFB

    1/kg

    Broiler chicks 5 mg/kg Secondary antibodies Okotie-Eboh et al.,1997against IBD

    Broiler chicks 400 g/kg AFB1

    Cell-mediated immunity Giambrone et al.,1985a+AFB

    2for 5 weeks measured by DTH skin test ;

    Humoral immunityunchanged;Acquired immunity to ND orfowl cholera vaccinationunchanged

    Broiler chickens 2.5 g/g of feed Total complement activity Stewart et al.,1985for 42 days

    Chickens 2.5 g/g diet from Cell-mediated immunity (graft- Giambrone et al.,1978hatching to 4 vs. host reaction) ; DTHweeks of age reactions to tuberculin ;

    Humoral immunity (to rabbitRBC) unchanged; Serum IgG and

    IgA; IgMunchangedChickens 0.3 mg/kg from Cell mediated immune response Kadian et al.,19880 to 6 weeks (DTH reaction ) suppressed at

    30, 45 and 60 days of age

    14-day-old 200 g/kg Cell-mediated immunity (DTH skin Giambrone et al.,1985b turkeys and test) ; Humoral immunity

    unchanged; Acquired immunity toND or fowl cholera vaccinationunchanged

    4-month-old 100-400 g/kg Lymphoblast transformation Quist et al.,2000 wild turkeys feed for 2 wk

    Chick embryo 1.09-17.4 g/g Mitotic index B-cells; Mitotic Potchinsky and Bloom,embryo at day 18 index T-cells unchanged; Sister 1993of incubation chromatid exchanges in B-

    lymphocytes ; Sister chromatidexchanges in T-lymphocytes

    6-day chick 0.1, 0.5, and 1 Macrophages recruited in the Neldon-Ortiz and Qureshi, embryos g/per embryo peritoneal cavity after i.p. 1992

    Sephadex elicitation ; Substrateadherence potential of peritonealexudate cells at 1 g AFB

    1;

    Macrophage phagocytic potential at 0.5-1.0 g AFB

    1

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    19/46

    P.F. Surai and J.E. Dvorska 111

    Table 5. Continued.Species Dose Effects on immune system Reference

    6-day chick 0.1 g Incidence of sister chromatid Dietert et al.,1985 embryos exchanges in blood cells (5-fold);

    Cell-mediated immunity (graft vshost and cutaneous basophilhypersensitivity reactions)

    Weanling 140-280 g/kg, AFB1decreased proinflammatory Marin et al.,2002

    piglets 4 wks (IL-1, TNF-) and increasedanti-inflammatory (IL-10) cytokinemRNA expression

    Weaned pigs 140, or 280 g/kg, Skin thickness (PHA test) van Heugten et al.,1994for 3 wks linearly with dietary AFB

    1

    Pigs 300 or 500 mg/kg Humoral immune response Panangala et al.,1986of feed to Erysipelothrix rhusiopathiae,measured by ELISA unchanged;Proliferative responses to mitogensunchanged;Complement titers;Serum IgG and M values

    Female lambs 2 mg/kg for 37 d Response to intradermal Fernandez et al.,2000injection of PHA

    White-tailed 0.8 mg/kg for 8 Lymphocyte proliferation and Quist et al.,1997deer wks DTH reactionsunchangedWeanling rats 60, 300 and Cell mediated immunity, Raisuddin et al.,1993

    600 g/kg BW measured by DTH responseassay, at the 300 and 600 gdose levels

    Weanling rats 350 and Population and phagocytic Raisuddin et al.,1990700 g/kgBW capacity of macrophages

    Rats and mice Aerosol inhalation Alveolar macrophage Jakab et al.,1994or intratracheal phagocytosis at 16.8 g/kginstillation to AFB

    1with the effect persisting for~2 weeks

    Mice 0.03, 0.145 or [3H]thymidine uptake in Reddy et al.,19870.70 mg/kg orally lymphocyte cultures; Synthesisevery other day for of DNA in mixed lymphocyte ;2 weeks Primary antibody production by

    splenic cells from animalschallenged with SRBC; DTHreaction to hemocyanin

    Murine 10 or 50 mM NO production stimulated Moon et al.,1998 peritoneal by LPS , mediated by the macrophages reduction of iNOS activity,

    mRNA, and protein

    Human 0.1-1 pg/ml Phagocytosis and microbicidal Cusumano et al.,1996 monocytes activity , Intrinsic antiviral

    activity or superoxideproduction unchanged

    Rat Kupffer 0.01 pg/ml Phagocytosis , intracellular Cusumano et al.,1995 cells in vitro killing of Candida albicans ,

    intrinsic anti-Herpes virus activity

    05-Surai.p65 19/12/2004, 23:24111

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    20/46

    112 Effects of mycotoxins on antioxidant status and immunity

    Table 6. Effect of Ochratoxin A on immunity.Species Dose Effects on immune system Reference

    Broiler chicks 2 mg/kg Vaccination titres to NK , Santin et al.,2002Mitotic cells in the bursa

    Chicks 5 mg/kg Weight of lymphoid organs , Stoev et al.,2002Humoral immune response

    Broiler chicks 130, 305 and HI titers to ND vaccination Stoev et al.,2000790 mg/kg

    Broiler chicks Up to 4 mg/kg for Ig-containing cells in Dwivedi and Burns, 198420 days from hatch lymphoid organs ; Total Ig

    levels at 2-4 ppmBroiler chickens 0.5-8 g/g from Total circulating lymphocytes ; Chang et al.,1979

    day-old to 3 weeks Monocytes at 2.0 g/g;

    of age Circulating heterophils unchangedSalmonella- 3.0 mg/kg PHA- and ConA-stimulated Elissalde et al.,1994 challenged blastogenesis ; S.typhimurium broiler chicks alone had no effect on the

    variables measured

    Growing gilts 2.5 mg/kg of feed Cutaneous basophil hyper- Harvey et al.,1992for 35 days sensitivity response to PHA ,

    DTH to tuberculin , Stimulationindex for lymphoblastogenesis ,Il-2 production when lymphocyteswere stimulated with ConA ,Number and phagocytic activity ofmacrophages

    Rats Gavage with 0, Thickening of the basement Dortant et al.,2001

    0.07, 0.34 or membrane and reduction in1.68 mg OTA/kg splenic T-cell fraction. IgG atBW for 4 wks 0.34-1.68 mg/kg OTA

    Rat Dams exposure to The proliferative response of Thuvander et al.,1996ba single dose of splenocytes to ConA in pupsOTA (0, 10, 50 or from dams given 10 or 50 g250 mg/kg BW) on OTA/kg BW; Proliferation ofday 11 of lactation thymocytes in response to

    ConA in pups from damsexposed to 50 g OTA/kg BW

    Mice Dams, OTA, In offspring on days 14 and 28 Thuvander et al.,1996200 g/kg before postpartum percentages of splenicand during gestation CD4+ and CD8+ cells

    Mice Dams exposure to Proliferation of splenic and Thuvander et al.,1996a

    OTA (500 g/kg BW) thymic lymphocytes in responseon day 16 of to mitogens in pups at 15gestation days of age ; percentages of

    mature CD4+ cells andpercentages of immature, double-positive (CD4/CD8+) cells inthe exposed pups

    Mice Dams exposure to Proliferative responsiveness of Thuvander et al.,1996aOTA (500 g/kg lymphocytes in the offspringBW) on day 10 when stimulated with B- or T-cellpostpartum mitogens 3 days after exposure

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    21/46

    P.F. Surai and J.E. Dvorska 113

    human lymphocyte populations andsubpopulations to the toxin will abrogate the

    cells ability to respond to activating stimuli invitro (Lea et al., 1989). Thus, both IL-2production and IL-2 receptor expression ofactivated T-lymphocytes were severelyimpaired. The results strongly suggest that thetoxin caused immunosuppression throughinterference with essential processes in cellmetabolism (Lea et al., 1989). In particular,OTA appears to suppress NK cell activity byinhibiting production of basal interferon. (Lusteret al.,1987).

    It is important to note that subchronic oralexposure to OTA affects certain immune

    functions in mice at exposure levels that maybe found in contaminated food products(Thuvander et al.,1995). In the mouse modelOTA had a non-selective suppressive effecton various immune reactions. They includeweight depression, lymphopenia, neutrophiliaand eosinophilia. Furthermore, antibody-producing cells, antibody titres in bloodserum and phagocytosis of E. coliby bloodphagocytes become suppressed. Immunizedanimals also showed a lower survival rateafter experimental infection with Pasteurellamultocidaas well as an increase in oxygen

    radicals in blood cells (Muller et al.,1995).Similarly, in Salmonella-challenged broilerchicks PHA- and ConA-stimulatedblastogenesis was depressed as a result ofOTA consumption, while Salmonellaalonehad no effect on these parameters (Elissaldeet al.,1994).

    In weaner pigs subtoxic amounts of OTAproduced immunomodulation in a dose-dependent mode (Muller et al., 1999). It

    included increased counts of total leukocytesand neutrophils in the blood and reduced

    lymphocyte levels. There was also asubstantial increase in the counts ofeosinophils and of apoptotic phagocytes.Reduced phagocytosis and reducedexpression of a swine-specific surface markeron lymphocytes were also observed.

    It is interesting that the time of exposuresignificantly influences the immunotoxiceffects of OTA on the developing immunesystem in rodents (Thuvander et al.,1996a).Recent studies have examined immunefunction in the offspring of rats and miceexposed to OTA pre- and perinatally (Bondy

    and Pestka, 2000). It has been shown thatthe decrease in proliferation and antibodyproduction in young animals resulted fromprenatal modulation of the immune system.In particular, prenatal exposure to relativelylow doses of OTA may induce immuno-suppression in the progeny. In contrast,short-term exposure of suckling pups to OTAvia the milk stimulated the proliferativeresponses of lymphocytes to polyclonalactivation.

    Fumonisin toxicity has been characterizedrelatively recently in comparison to aflatoxin

    and ochratoxin (Table 7), and fumonisin-induced immunotoxicity is an area of activeresearch (Bondy and Pestka, 2000). In fact,FB

    1 has diverse effects on the immune

    system, causing both stimulation andsuppression of responses to foreign antigens,and apparently inducing an antigenic responseto FB

    1. For example, in chickens FB

    1caused

    a decrease in total immunoglobulins, in IgGand macrophage phagocytic activity (Qureshi

    Table 6. Continued.

    Species Dose Effects on immune system Reference

    Mice 250 or 2600 mg/kg After 28 days, AB production Thuvander et al.,1995diet, 28 days and plaque-forming cells ;

    Decrease in thymocyte cellcounts in the 250-g/kggroup; After 90 days, proportionof mature CD4+ or CD8+ cells

    Mice 0.005 g/kg BW Immune response to SRBC Haubeck et al.,1981

    05-Surai.p65 19/12/2004, 23:24113

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    22/46

    114 Effects of mycotoxins on antioxidant status and immunity

    Table 7. Effects of Fumonisins on immunity.Species Dose Effects on immune system Reference

    White Leghorn 6.1, 10.5, and Splenic, thymic, and liver Qureshi et al.,1995 chicks 42.7 mg/kg from weights ; Bursa of Fabricius

    day 7 to 42 unchanged; Total Ig andIgG; Macrophage phagocyticactivity ; NK cell activityunchanged

    Turkey poults 75 mg/kg feed and Primary immune response to Weibking et al.,1994AB

    1, 200 g/kg SRBC (combination of FB

    1and

    feed, from day AFB1); PHA responseunchanged

    1 to 21

    Weaned pigs 1-100 mg/animal/d Titres unchanged; Lymphocyte Tornyos et al.,2003stimulation by PHA, ConA, LPSunchanged

    Rat Injected at 7.5 or Thymus weight , Serum Bondy et al.,199510 mg/kg BW/day IgM; Circulating phagocyticfor 4 days cell numbers

    Mice 50 (LD), FM1+ After challenge with T. cruzi, Dresden et al.,2002FM

    2, 6 weeks NO production by MGafter

    14 days

    Mice Five daily s.c. Relative spleen and thymus Johnson and Sharma,injections of weights in F , No effect on organ 20012.25 mg/kg/d FB

    1weights in M; Splenic cellularityand the basal rate of lymphocyteproliferation in F ; PHA-inducedT-lymphocyte and LPS-inducedB-lymphocyte proliferation in F ;Expression of IL-2 mRNA insplenocytes in F

    Mice I.p. with SRBC, Number of plaque-forming cells Martinova et al.,1995at 5-100 g produced against SRBC

    Mice I.p daily, at 1 to 4 to 12-fold increase in the Martinova et al.,199550 g number of of plaque-forming

    cells after SRBC injection

    Macrophage 1-10 M Membrane fluidity ; Membrane Ferrante et al.,2002 cell line peroxidative damage

    Murine 1-100 M LPS-induced production of NO Meli et al.,2000 macrophage and PGE

    2at 0.1-10 M

    cell line

    Murine 1, 10, and 100 M LPS-induced NO production Rotter and Oh, 1996 macrophage

    cellRat splenic 1-100 g/ml NO production ; ConA-induced Dombrink-Kurtzman et al., macrophages proliferation of splenic cells in 2000 and lymphocytes the presence of NO synthase

    inhibitor

    et al.,1995). However, in turkey poults acombination of FB

    1and AFB

    1was responsible

    for increased primary immune response toSRBC (Weibking et al., 1994). In weaned

    pigs, FB1(up to 100 mg/animal/day) did not

    affect immunity (Tornyos et al.,2003). Effectsof FB

    1on immunocompetence of laboratory

    animals also vary substantially (Table 7).

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    23/46

    P.F. Surai and J.E. Dvorska 115

    Immunomodulatory properties of FB1probably depend on its effect on lipidmetabolism, antioxidant/pro-oxidant balanceand interactions with other factors. Forexample, FB

    1 decreased CD3 receptor

    expression on the surface of thymus cells invitroand in vivo, which is consistent withthe sharp decrease in the ceramide contentin this organ (Martynova et al.,1995).

    Similar to FB1, the trichothecenes can both

    suppress and stimulate immune function(Table 8). Most of the research on T-2 toxineffects on immunity was perfomed with

    laboratory animals and there are only a fewpublications addressing immunomodulatingproperties of T-2 toxin in farm animals.Futhermore, the molecular basis of immunemodulation by mycotoxins remains mostlyunknown and continues to be an active areaof research (Bondy and Pestka, 2000). Itappears that trichothecenes are potentimmunosuppressive agents that directly affectimmune cells and modify immune responsesbecause of tissue damage elsewhere. Forexample, sheep and calves treated withFusarium T-2 toxin develop leukopenia and

    decreased function of peripheral lymphocytes(Sharma, 1993). In fact, exposure ofexperimental animals and humans to T-2 toxinhas been shown to have a variety ofimmunosuppressive effects, including alteredparameters of humoral immunity. It is wellestablished that T-2 toxin is cytotoxic tolymphocytic cellsin vitro, however, limitedinformation is presently available regardingthe contribution of such a mechanism toimmunosuppression in vivo, or to potentialimmune cell targets. It seems likely thatlymphocyte progenitors, in contrast to

    thymocytes, represent highly sensitive targetsof T-2 toxin exposure, responsible for thymicatrophy (Holladay et al.,1993). For example,subchronic T-2 toxin treatment of timed-pregnant B6C3F1 mice resulted in significantand selective depletion of fetal liver cellsexpressing low levels of surface CD44 andCD45 antigens, suggestive of possiblelymphoid progenitor cell sensitivity to this

    agent (Holladay et al.,1995). The authorsshowed that the precursors of B-cells mightrepresent highly sensitive targets of T-2 toxinexposure. Impaired resistance to pathogenicmicroorganisms occurs after exposure to thetrichothecenes T-2 toxin and DON. This maypredispose food animals to infectious diseaseand could result in decreased productivity aswell as increased animal-to-humantransmission of pathogens such as Salmonellaand Listeria (Pestka and Bondy, 1990). Forexample, mice challenged with S.typhimuriumand then treated with T-2 toxin

    every other day for 10 days had markedlylarger and more bacteria-related lesions in thespleen, kidney, and liver than animalschallenged with S. typhimuriumalone (Taiand Pestka, 1990). In great contrast, in miceexposed to T-2 toxin at 3 mg/kg BW bygavage, virulence of both E. coli and S.auerus decreased. Apparently there arespecies-specific features in sensitivity totrichothecenes. For example, recently it hasbeen shown that T-2 toxin (up to 1 ppm) inthe poult diet did not affect antibodyproduction stimulated by various antigens

    (Sklan et al.,2003).Immunomodulating properties of DON

    have been studied mainly with rodents (Table9). The evidence is quickly accumulating toshow that DON can be immunosuppressiveor immunostimulatory, depending upon thedose and duration of exposure. Whileimmunosuppression is probably related to theinhibition of translation, immunostimulationcan be a result of interference with variouscellular regulatory mechanisms (Rotter et al.,1996). For example, in vivoDON suppressesnormal immune response to pathogens and

    simultaneously induces autoimmune-likeeffects. Characteristic effects of DON alsoinclude superinduction of cytokineproduction by T-helper cells (in vitro) andactivation of macrophages and T-cells toproduce proinflammatory cytokines. Althoughthese effects have been largely characterisedin the mouse, several investigations withDON suggest that immunotoxic effects are

    05-Surai.p65 19/12/2004, 23:24115

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    24/46

    116 Effects of mycotoxins on antioxidant status and immunity

    Table 8. Effects of T-2 toxin on immunity.

    Species Dose Effects on immune system Reference

    Young poults Up to 1 mg/kg for Antibody titres unchanged Sklan et al.,200332 days

    7-week-old pigs 0.5, 1.0, 2.0 or Leukocyte count ; Proportion of Rafai et al.,19953.0 mg/kg diet for T-lymphocytes ; Antibody3 weeks formation ; Blastogenic

    transformation of lymphocytes

    9-10-week- Aerosol, 390 g/L Lymphocyte count , Neutrophil Pang et al.,1988 old pigs count ; ConA, PHA, and PWM

    blastogenic responses and H1 titersto SRBC

    9-10-week- 15 mg/kg Blastogenic responses to ConA, Pang et al.,1987 old pigs PHA and PWM at early (3 to

    5 days) and late (20 to 28 days)postdosing intervals; HA titer to SRBCunchanged

    Calves 0.6 mg/kg/day Lymphocyte responses to Buening et al.,1982mitogens ; Chemotaxic migrationof neutrophils

    Calves 0.6 mg/kg per day IgM and IgA ; IgG Mann et al., 1982for 43 days

    Monkeys Gastric intubation Leucocyte countsat the end of Jagadeesan et al.,1982at 100 g/kg BW/ wk 4; Bactericidal activity ofday for 4-5 wk neutrophils , T-cell number ;

    Lymphocyte transformation ,

    B-cell number ; IgG and IgMlevels

    Rats 2 mg/kg In progeny, the cortex was Lafarge-Frayssinet et al.,atrophic while the medulla 1990was proliferative. In thespleen, both B- and T-cells wereimpaired and responsivenessto PHA and LPS decreased

    Mice 3 mg/kg BW by Virulence of both E. coliand Cooray and Jonsson,gavage S. aureus 1990

    Mice 1 mg/kg every Markedly larger and more Tai and Pestka, 1990 challenged other day for 10 d bacteria-related lesions in thewith spleen, kidney, and liver than S. typhimurium animals challenged with

    S. typhimuriumaloneMice 20 ng Immune response to sheep RBC Holt and DeLoach,1988

    Mice 1.5 or 3.0 ppm T-lymphocyte-dependent Schiefer et al.,1987in the diet for humoral immunity unaffected16 months

    Mice exposed 4.0 mg/ kg of BW Growth of Listeria ; Mortality Corrier and Ziprin, to Listeria 1986

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    25/46

    P.F. Surai and J.E. Dvorska 117

    Table 9. Effects of DON on immune system.

    Species Dose Effects on immune system Reference

    Growing pigs 0.6, 1.8 and Secondary antibody response Overnes et al.,19974.7 mg/kg to tetanus toxoid dose-

    dependentlyYoung pigs 0.75- 3.00 mg/kg -globulin levels, and Rotter et al.,1994

    diet for 28 days antibody titers to SRBCMice 5 or 25 mg/kg BW mRNAs for IL-1, IL-6, and Zhou et al.,1997

    TNF-; the T-helper 1 cytokinesIFN-and IL-2; and the T helper2 cytokines IL-4 and IL-10

    Mice 25 mg/kg for 8 or Serum IgA and microhematuria Dong and Pestka, 199324 ws index after 4 to 8 wks and

    continued to increase with furtherexposure

    Mice 25 ppm for 4 and Total IgA ; Total IgG and IgM Rasooly ands Pestka, 19928 wks

    Mice 10-100 mg/kg in Serum levels of anti-SRBC Robbana-Barnat et al.,

    the diet antibodies ; Stimulation of B- 1988and T-cells by mitogens

    Weanling mice 0.5- 25.0 mg/kg Total circulating white blood Forsell et al.,1986over 8 wks cells ;PMN neutrophils ; Serum

    IgM ; IgA Weanling mice 0.75, 2.5 or 7.5 Serum IgM to SRBC ; Plaque- Tryphonas et al.,1984

    mg/kg BW by forming cell numbers gavage

    Murine 0.1 ng/ml-1 g/ml Phagocytosis ; Microbiocidal Ayral et al.,1992 peritoneal activity ; Superoxide anion macrophages production at 1 ng/ml;

    Phagosome-lysozome fusion at 100 ng/nl

    Lymphocytes 0.005-100 ng/ml PHA-induced lymphocyte Miller and Atkinson, 1986 in vitro proliferation at 0.005- 0.5 ng

    DON/ml; at 50-100 ng/mllymphocyte proliferation

    Human 216 ng/ml 50% inhibition in cell Meky et al.,2001 lymphocytes proliferation

    Murine 25-100 ng/ml Production of H2O

    2after LPS Ji et al.,1998

    macrophage stumulation , but at 250 ng/m; cell line NO production at 25-250 ng/ml

    Table 8. Continued.

    Species Dose Effects on immune system Reference

    Mice 20 ppm for Spleen proliferative responses Friend et al.,19831- 4 weeks to ConA ;

    Human 1 -5 ng/ml 50% inhibition in cell Meky et al.,2001 lymphocytes proliferation

    05-Surai.p65 19/12/2004, 23:24117

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    26/46

    118 Effects of mycotoxins on antioxidant status and immunity

    also likely in domestic animals (Rotter et al.,1996). Therefore, DON can selectively andconcurrently upregulate or downregulatecritical functions associated with activatedmacrophages. For example, a single DONexposure rapidly induces gene expression invivo for a wide range of cytokines withapparently complete recovery occurring after24 hrs. Elevated cytokine expression mayplay an important role in the patho-physiological effects of DON and othertrichothecenes (Zhou et al., 1997). Malesappeared more susceptible than female mice

    to DON-induced IgA dysregulation and IgAnephropathy in terms of latency, thresholddose, and severity (Greene et al.,1994). It isinteresting that experimental dysregulationof IgA production and IgA nephropathypersisted up to four months after a discreteperiod of dietary DON exposure (Dong andPestka, 1993).

    Mycotoxin-induced immunosuppression isrelated to both natural and adaptiveimmunity:

    depressed T- or B-lymphocyte activity

    depressed NK cell activity suppressed immunoglobulin and antibody

    production reduced complement or interferon activity impaired macrophage functions

    The sensitivity of the immune system tomycotoxin-induced immunosuppression arisesfrom the vulnerability of the continuallyproliferating and differentiating cells thatparticipate in immunomediated activities andregulate the complex communication networkbetween cellular and humoral components

    (Corrier, 1991). In fact, high levels ofpolyunsaturated fatty acids in the immune cellsand presence of sensitive receptors on cellsurfaces make them an important target for freeradical attack (Surai, 2002). As mentionedabove, oxidative stress caused by mycotoxinconsumption could be responsible for breakingeffective communication between immunecells, which ultimately would misregulate the

    immune system and result in immuno-suppression. Although the molecular basis formany of the specific immunosuppressive effectsof mycotoxins is presently unclear, inhibitionof DNA, RNA and protein synthesis via a varietyof different mechanisms appears to be directlyor indirectly responsible for theimmunosuppressive action of many mycotoxins(Corrier, 1991). Furthermore, detrimental effectson antioxidant defences, stimulation of lipidperoxidation and apoptosis and damage toimmune cell receptors appear to be involvedin the immunomodulation properties of

    mycotoxins. Consequences of oxidative stressfor immunocompetence are shown in Figure3.

    Protective effect of antioxidants

    against mycotoxicoses

    The wide range of mycotoxins that cancontaminate animal feed and their differingchemical compositions makes protectionagainst mycotoxin-related toxicity a difficult

    task. There are various approaches to controlor combat mycotoxin problems. The simpleststrategy is based on the prevention ofmycotoxin formation in feeds by specialmanagement programmes including storageat low moisture levels and prevention ofgrain damage during processing (Dawson,2001). However, modern agronomictechnology cannot eliminate pre-harvestinfection of susceptible crops by fungi (Wood,1992). Therefore, this strategy can be onlypartially effective; and in countries withwarm and humid conditions, this strategy can

    be quite costly.Other strategies based on microbial orthermal inactivation of toxins, physicalseparation of contaminated feedstuffs,irradiation, ammoniation and ozonedegradation have not been developed to anindustrial level because they are either time-consuming or comparatively expensive(Dawson, 2001a). In recent years, nutritionalmanipulation has been actively used to

  • 7/26/2019 Chapter Mycotoxine Immunity- Blue Book 2005

    27/46

    P.F. Surai and J.E. Dvorska 119

    improve animal defence against mycotoxinsor to decrease the detrimental consequencesof mycotoxin consumption.

    Since lipid peroxidation plays an importantrole in mycotoxin toxicity, a protective effectof antioxidants is expected (Galvano et al.,2001). Indeed, in several experiments withvarious animal species, protective effects ofantioxidants against the toxic effects ofmycotoxins were observed. For example,

    vitamin E supplementation ameliorated the pro-oxidative effects of OTA in the chicken(Hoehler and Maraquardt, 1996) and mouse(Grosse et al.,1997). Trolox C, a water-solubleform of vitamin E, prevented the genotoxiceffects of OTA in E. coli (Malavielle et al.,1994). Protective effects of vitamin E were alsoobvious in aflatoxicosis (Shen et al.,1994; Choiet al.,1995; Souza et al.,1999; Verma andNair, 2001), DON or T-2 toxicoses (Segal etal.,1983; Tsuchida et al.,1984; Rizzo et al.,1994) in rats; T-2 toxicosis in mice in vivo(Atroshi et al.,1997) or in an in vitrosystem