Proteomic changes in chicken primary hepatocytes exposed ...

15
Proteomics 2013, 13, 3175–3188 3175 DOI 10.1002/pmic.201300015 RESEARCH ARTICLE Proteomic changes in chicken primary hepatocytes exposed to T-2 toxin are associated with oxidative stress and mitochondrial enhancement Peiqiang Mu 1, Ming Xu 1, Lei Zhang 2 , Kaixin Wu 1 , Jun Wu 1 , Jun Jiang 1 , Qingmei Chen 1 , Lijuan Wang 1 , Xianqing Tang 1 and Yiqun Deng 1 1 Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong, P. R. China 2 Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China T-2 toxin is a mycotoxin that is toxic to plants, animals, and humans. However, its molecular mechanism remains unclear, especially in chickens. In this study, using 2D electrophoresis with MALDI-TOF/TOF-MS, 53 proteins were identified as up- or downregulated by T-2 toxin in chicken primary hepatocytes. Functional network analysis by ingenuity pathway analysis showed that the top network altered by T-2 toxin is associated with neurological disease, cancer, organismal injury, and abnormalities. Most of the identified proteins were associated with one of eight functional classes, including cell redox homeostasis, transcriptional or translational regulation, cell cycle or cell proliferation, stress response, lipid metabolism, transport, carbo- hydrate metabolism, and protein degradation. Subcellular location categorization showed that the identified proteins were predominantly located in the mitochondrion (34%) and interest- ingly, the expression of all the identified mitochondrial proteins was increased. Further cellular analysis showed that T-2 toxin was able to induce the ROS accumulation and could lead to an increase in mitochondrial mass and adenosine 5 -triphosphate content, which indicated that oxidative stress and mitochondrial enhancement occurred in T-2 toxin-treated cells. Overall, these results characterize the global proteomic response of chicken primary hepatocytes to T-2 toxin, which may lead to a better understanding of the molecular mechanisms underlying its toxicity. Keywords: Chicken / Mitochondrial enhancement / Oxidative stress / Proteome / T-2 toxin Received: January 10, 2013 Revised: August 5, 2013 Accepted: August 9, 2013 Additional supporting information may be found in the online version of this article at the publisher’s web-site Correspondence: Professor Yiqun Deng, Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, P. R. China E-mail: [email protected] Fax: +86-20-38604967 Abbreviations: ATP, adenosine 5 -triphosphate; DCFH-DA,2 ,7 - dichlorofluorescin diacetate; MTT, 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide; IPA, ingenuity pathway analysis; PI, propidium iodide 1 Introduction T-2 toxin is a mycotoxin that belongs to the group of type A tri- chothecenes. It is produced by several fungal genera that can infect crops in the field or during storage and subsequently contaminate animal feed and human food [1,2]. Many reports worldwide have described an association between T-2 toxin and agricultural damage and adverse effects on animals [3–6]. These authors contributed equally to this work. Colour Online: See the article online to view Figs. 1, 2, and 4 in colour. C 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

Transcript of Proteomic changes in chicken primary hepatocytes exposed ...

Proteomics 2013, 13, 3175–3188 3175DOI 10.1002/pmic.201300015

RESEARCH ARTICLE

Proteomic changes in chicken primary hepatocytes

exposed to T-2 toxin are associated with oxidative

stress and mitochondrial enhancement

Peiqiang Mu1∗, Ming Xu1∗, Lei Zhang2, Kaixin Wu1, Jun Wu1, Jun Jiang1, Qingmei Chen1,Lijuan Wang1, Xianqing Tang1 and Yiqun Deng1

1 Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College ofLife Sciences, South China Agricultural University, Guangzhou, Guangdong, P. R. China

2 Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou,Guangdong, P. R. China

T-2 toxin is a mycotoxin that is toxic to plants, animals, and humans. However, its molecularmechanism remains unclear, especially in chickens. In this study, using 2D electrophoresiswith MALDI-TOF/TOF-MS, 53 proteins were identified as up- or downregulated by T-2 toxinin chicken primary hepatocytes. Functional network analysis by ingenuity pathway analysisshowed that the top network altered by T-2 toxin is associated with neurological disease, cancer,organismal injury, and abnormalities. Most of the identified proteins were associated with oneof eight functional classes, including cell redox homeostasis, transcriptional or translationalregulation, cell cycle or cell proliferation, stress response, lipid metabolism, transport, carbo-hydrate metabolism, and protein degradation. Subcellular location categorization showed thatthe identified proteins were predominantly located in the mitochondrion (34%) and interest-ingly, the expression of all the identified mitochondrial proteins was increased. Further cellularanalysis showed that T-2 toxin was able to induce the ROS accumulation and could lead to anincrease in mitochondrial mass and adenosine 5′-triphosphate content, which indicated thatoxidative stress and mitochondrial enhancement occurred in T-2 toxin-treated cells. Overall,these results characterize the global proteomic response of chicken primary hepatocytes to T-2toxin, which may lead to a better understanding of the molecular mechanisms underlying itstoxicity.

Keywords:

Chicken / Mitochondrial enhancement / Oxidative stress / Proteome / T-2 toxin

Received: January 10, 2013Revised: August 5, 2013

Accepted: August 9, 2013

� Additional supporting information may be found in the online version of this article atthe publisher’s web-site

Correspondence: Professor Yiqun Deng, Guangdong ProvincialKey Laboratory of Protein Function and Regulation in AgriculturalOrganisms, College of Life Sciences, South China AgriculturalUniversity, Guangzhou, Guangdong 510642, P. R. ChinaE-mail: [email protected]: +86-20-38604967

Abbreviations: ATP, adenosine 5′-triphosphate; DCFH-DA, 2′, 7′-dichlorofluorescin diacetate; MTT, 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide; IPA, ingenuity pathway analysis;PI, propidium iodide

1 Introduction

T-2 toxin is a mycotoxin that belongs to the group of type A tri-chothecenes. It is produced by several fungal genera that caninfect crops in the field or during storage and subsequentlycontaminate animal feed and human food [1,2]. Many reportsworldwide have described an association between T-2 toxinand agricultural damage and adverse effects on animals [3–6].

∗These authors contributed equally to this work.

Colour Online: See the article online to view Figs. 1, 2, and 4 incolour.

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

3176 P. Mu et al. Proteomics 2013, 13, 3175–3188

The consumption of T-2 toxin can lead to different toxiceffects, such as fatal alimentary toxic aleukia disease in hu-mans and vomiting in pigs [7]; therefore, the assessment ofT-2 toxin levels in foodstuffs, especially in chicken, whichserve as a major human food source, is important. Chick-ens are at particular risk of T-2 toxin exposure because manychicken feed mainly contain cereal grains. Previous reportsshowed that the average T-2 toxin in cereal grains rangesbetween 0.03 mg/kg and 0.155 mg/kg [8, 9]. The intake ofT-2 toxin-contaminated feed can lead to decreased weightgain and egg production in chickens and egg hatching isalso impaired [10–13]. T-2 toxin can also cause feather alter-ations in chickens [14]. At the molecular level, exposure to T-2toxin causes a significant reduction in serum total protein andcholesterol levels and an increase in serum uric acid and lac-tate dehydrogenase levels [15]. Furthermore, DNA fragmenta-tion has been observed in chicken spleen leukocytes, and sup-plementation with nucleotides was able to reduce the degreeof this damage [16]. Cell viability, alkaline phosphatase activ-ity, and glutathione content in primary cultures of chickentibial growth plate chondrocytes were all markedly decreasedupon exposure to T-2 toxin [17]. A T-2 toxin-containing dietwas associated with significant decreases in antioxidant con-centrations [18]. However, the exact molecular mechanism ofT-2 toxicity in chickens is unknown.

Several experiments to investigate the toxicity of T-2toxin have already been performed in various cell lines,especially cancer cells, and a variety of mechanisms havebeen proposed. The inhibition of elongation of proteinbiosynthesis in eukaryotic cells and apoptosis are generallyconsidered the main cytotoxic effects of T-2 toxin [1, 3]. T-2toxin-induced apoptosis has been observed in various celltypes, such as HL60, Jurkat, U937 [19], Vero cells [20], andhuman hepatoma cells [21]. Apoptosis has also been reportedin vivo in thymic and splenic lymphocytes, bone marrow,and intestinal epithelial cells [22] and in the skin [23], kid-ney [24], and brain [25] in mice. In addition to the inhi-bition of protein biosynthesis and apoptosis, the inhibitionof DNA and RNA synthesis by T-2 toxin has also been ob-served in several cell lines [26–28]. Other activities of T-2toxin have also been observed, such as inducing single-strandbreaks in the DNA of lymphoid cells [29], impairing antibodyproduction [30], altering membrane functions [31], reducinglymphocyte proliferation [32], and altering the maturation ofdendritic cells [33]. However, the in vitro–in vivo and inter-species extrapolation of the action of T-2 toxin has not beenclearly clarified.

Understanding the mechanism of action of T-2 toxin isessential for predicting its potential deleterious effects anddeveloping countermeasures. Therefore, this study was in-tended to evaluate the actions of T-2 toxin in chickens andexplore its potential molecular mechanisms at the proteomelevel. Primary hepatocytes have been successfully used inmany toxicity studies because they reflect the in vivo re-sponse to some degree and are easily manipulated [34, 35].In this study, we investigated the proteomic changes caused

by T-2 toxin exposure in primary cultures of chicken hepato-cytes and analyzed T-2 toxicity at the cellular and molecularlevels.

2 Materials and methods

2.1 Chemicals and reagents

T-2 toxin was purchased from Sigma-Aldrich (St. Louis, MO,USA). Williams’ E medium, Medium 199, Dulbecco’s modi-fied Eagle’s medium and FBS were obtained from Invitrogen(Carlsbad, CA, USA). All other chemicals including CHAPS,DTT, and PMSF, were obtained from GE Healthcare (Upp-sala, Sweden) or Merck (Darmstadt, Germany), unless oth-erwise indicated. All primers were synthesized by InvitrogenBiotechnology, Guangzhou (Guangzhou, China). All otherchemicals and reagents were of the highest analytical gradeavailable.

2.2 Chicken primary hepatocytes isolation, culture,

and T-2 toxin treatment

Eighteen-day-old chicken embryos were purchased fromthe Institute of Animal Science, Guangdong Academyof Agricultural Sciences (Guangzhou, China). Hepatocyteswere isolated as described previously [36]. Chicken em-bryo primary hepatocytes were cultured in Williams’ Emedium (Sigma-Aldrich) containing 8% FBS, 100 U/mLpenicillin/streptomycin, 10 nM insulin, and 10 nMdexamethasone (Sigma-Aldrich) for 5 h and then cultured inMedium 199 (Gibco BRL, Carlsbad, CA, USA) containing 8%FBS, 100 U/mL penicillin/streptomycin, 10 �g/mL insulin,1 �g/mL dexamethasone, and 2 mM L-glutamate (Invitro-gen) for 30 h. The medium was then changed to medium199 containing 8% FBS, 100 U/mL penicillin/streptomycin,10 �g/mL insulin, 1 �g/mL dexamethasone, 2 mML-glutamate, and various concentrations of T-2 toxin andcultured as described in the following experimental pro-cedures, including 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, Giemsa staining, 2DE,quantitative real-time RT-PCR, flow cytometry assay, singlecell gel electrophoresis, mitochondrial mass determination,and adenosine 5′-triphosphate (ATP) content determination.All cells were maintained in a humidified incubator at 37�Cwith 5% CO2. All experimental procedures were approved bythe Institute of Animal Care and Use Committee of SouthChina Agricultural University, and adhered to the ChineseGuidelines for the Proper Conduct of Animal Experimentsfor the use of laboratory animals.

2.3 MTT assay and Giemsa staining

Cell viability was measured using the MTT assay [37].Chicken primary hepatocytes were seeded onto 96-well plates

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

Proteomics 2013, 13, 3175–3188 3177

(1 × 105 cells per well) and treated for 48 h with different con-centrations of T-2 toxin (0–320 ng/mL), referred to the MTTassay of porcine primary hepatocytes under T-2 toxin treat-ment [34]. Subsequently, 0.5 mg/mL MTT (Sigma-Aldrich)was added to each well, and the plates were incubated at 37�Cfor an additional 4 h. The medium containing MTT was thenremoved, and 200 �L DMSO (Sigma-Aldrich) was added toeach well to dissolve the formazan crystals. The absorbanceat 570 nm was measured using a microplate reader (Bio-Radlaboratories, Hercules, CA, USA). All experiments were per-formed in triplicate.

For Giemsa staining, chicken primary hepatocytes weretreated with 0.05, 0.1, or 0.2 �g/mL T-2 toxin for 48 h. Thecells were subsequently washed in Dulbecco’s PBS, fixed with50% methanol for 2 min, fixed with absolute methanol for10 min, stained with Giemsa solution for 2 min and thenstained with 1/10 Giemsa solution for 2 min. Finally, thecells were washed with ultra-pure water and observed by lightmicroscopy (Olympus IX70-S1F, Tokyo, Japan).

2.4 Two-dimensional gel electrophoresis

The chicken primary hepatocytes from the control and0.05 �g/mL T-2 toxin treatment groups were homogenizedin 0.1 mL of lysis buffer (7 M urea, 2 M thiourea, 4% CHAPS,30 mM Tris, 40 mM DTT, 2% IPG buffer, and 1 mM PMSF).The homogenates were centrifuged at 20 000 × g for 20 minat 4�C. The 2D Clean-Up Kit (GE Healthcare) was used toremove ionic interfering components from the protein ex-traction. The protein concentration in the urea-containingprotein samples was quantified using a 2D Quant Kit (GEHealthcare). A 250 �g sample of protein was diluted to450 �L with rehydration solution (8 M urea, 2% CHAPS,40 mM DTT, and 0.5% IPG buffer) and applied to immobi-lized dry IPG strips (linear pH gradient from pH 4–7, 24 cm)and rehydrated passively for 14 h. The samples were thenfocused for 30 min at 100 V, 30 min at 200 V, 1 h at 500 V,30 min gradient at 1000 V, and 4 h gradient at 8000 V, for atotal of 50 000 V h, using the IPGphor III system (AmershamBiosciences, Piscataway, NJ, USA). Focused IPG strips wereequilibrated in equilibration solution I containing 6 M urea,2% SDS, 30% glycerol, 50 mM Tris–HCl, pH 6.8, and 1% DTTfor 15 min and then equilibrated for an additional 15 min inequilibration solution II that was identical to equilibrationsolution I, except that the DTT was replaced by 2.5% iodoac-etamide. After equilibration, 12% SDS-PAGE was performedon an Ettan DALT II system (Amersham Biosciences). The2DE gel was then stained with silver nitrate [38]. The 2DE gelof the control and T-2 toxin-treated groups were replicatedthree times biologically.

2.5 2DE image analysis

The stained 2DE gels were scanned with MagicScan soft-ware on an Imagescanner (Amersham Biosciences) and an-

alyzed using ImageMaster V 5.0 software (GE Healthcare,Piscataway, NJ, USA) according to the manufacturer’s pro-tocol. Spot intensities were calculated by the spot volume(after normalizing the image by the total spot volume nor-malization method) multiplying the total area of all the spots.The change index was described as the ratio between thespot percentages relative to the spot volumes in the controland T-2 toxin-treated chicken primary hepatocytes. Proteinswere classified as being differentially expressed between thecontrol and T-2 toxin-treated groups when the spot intensityshowed a ≥1.3-fold difference between the control and T-2toxin-treated groups. Differences in protein expression levelswere evaluated with the Student’s t-test and considered to besignificant when p < 0.05.

2.6 Protein identification by MALDI-TOF/TOF

analysis

The spots of interest were manually excised from the silver-stained gels and subjected to in-gel trypsin digestion. Afterdigestion, the peptides were then extracted twice using 0.1%TFA in 50% ACN. The extracts were pooled and dried com-pletely using a Speed Vac vacuum concentrator. The mix-tures of peptides were redissolved in 0.1% TFA. Then, 0.4 �Lof matrix (CHCA in 30% ACN, 0.1% TFA) was added into0.8 �L of peptide solution before spotting on the target plate.

Protein identification was performed on an AB SCIEXMALDI TOF-TOFTM 5800 Analyzer (AB SCIEX, Foster,CA, USA) equipped with a neodymium: yttrium-aluminum-garnet laser (laser wavelength was of 349 nm) according to ourpreviously described method [34] and listed in the supportinginformation.

2.7 Bioinformatics analysis of differentially

expressed proteins

The functional networks were identified by the ingenuitypathway analysis (IPA) according to the biological functionsand/or diseases (Ingenuity Systems, Redwood, CA, USA).The statistically significance score for each network were gen-erated based on the fit of the network to the set of genesdefined by the user. The score is derived from the nega-tive log of p-value, which indicates the likelihood that thefocus genes would be found together in a network by ran-dom chance. If the score of a functional network is 20 orhigher, the network is not being generated by random chancealone with at least 99% confidence. In addition, to obtain amore detailed and precise view of the biological significanceof the altered proteins, these differently expressed proteinswere further categorized according to their main molecu-lar functions and subcellular localizations by searching theGene Ontology (http://www.geneontology.org/) and PubMed(http://www.ncbi.nlm.nih.gov) databases.

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

3178 P. Mu et al. Proteomics 2013, 13, 3175–3188

2.8 Quantitative real-time RT-PCR

Total RNA was isolated from 0.05 �g/mL T-2 toxin-treatedchicken primary hepatocytes or controls using Trizol reagent(Invitrogen) according to the manufacturer’s instruction.RNA samples (3 �g) were reverse transcribed by M-MLVreverse transcriptase (Promega, Madison, WI, USA) withOligo d (T) (MBI Fermentas, Newington, NH, USA) andrandom primers. PCR primers specific for the genes encod-ing the proteins identified in MALDI-TOF/TOF-MS were de-signed based on expressed sequence tags from Gallus gallus(Supporting Information Table S2). All primers were synthe-sized by Invitrogen (Guangzhou, China). The GAPDH waschosen as an internal control and used for normalization.Each RT-PCR assay was replicated three times. Fold differ-ences in expression levels were calculated using the 2−��Ct

method [39].

2.9 Flow cytometry assay

The chicken primary hepatocyte cells were exposed to0.05 �g/mL of T-2 toxin for 12, 24, 48, or 72 h. Then, theROS content and level of apoptosis were examined with BDFACSCaliburTM flow cytometer (BD Biosciences, FranklinLakes, NJ, USA) using CellQuest 3.1 (BD Biosciences) andFlowJo software (Tree Star, Ashland, OR, USA).

Intracellular ROS were detected using an oxidation-sensitive fluorescent probe 2′, 7′-dichlorofluorescin diacetate(DCFH-DA) (Beyotime, Shanghai, China). After treatmentwith T-2 toxin (0.05 �g/mL for 12, 24, 48, and 72 h),chicken primary hepatocytes cells were washed twice withPBS and incubated with 10 �M DCFH-DA solution at 37�C for20 min. The fluorescence intensity was then measuredby BD FACSCaliburTM flow cytometry at an excitationwavelength of 488 nm and emission wavelength of535 nm.

To examine apoptosis, an apoptosis detection kit (KeyGEN,Nanjing, China) based on annexin V-FITC was used. AnnexinV specifically binds to phosphatidylserine, which is exposedto the plasma membrane surface during the early stages ofcell apoptosis, and propidium iodide (PI) is a vital dye la-beling the nucleus in dying cells. Therefore, the viable cellsare not stained by annexin V or PI, and the early apoptoticcells are stained with annexin V-FITC only, whereas the lateapoptotic cells and necrotic cells are double-stained with bothannexin V-FITC and PI. Cells were treated with or without0.05 �g/mL T-2 toxin for 12, 24, 48, and 72 h, resuspended inbinding buffer at a final cell concentration of 1 × 106 cells/mL,and then incubated with both annexin V-FITC and PI for10 min in the dark. The fluorescence intensity was analyzedby BD FACSCaliburTM flow cytometry at an excitation wave-length of 425 nm, FITC fluorescence detection wavelengthof 525 nm, and PI fluorescence detection wavelength of630 nm.

2.10 Single cell gel electrophoresis

Oxidative damage to the DNA of chicken primary hepatocyteswas determined by the single cell gel electrophoresis (cometassay) according to Olive [40]. T-2 toxin-treated or -untreatedcells were harvested by trypsin digestion, and the cell densitywas adjusted to 1 × 106 cells/mL before electrophoresis. Theelectrophoresis slides were precoated in 0.5% normal-meltingpoint agarose by dipping; then, a 10 �L cell sample was mixedwith 70 �L of 0.7% low-melting point agarose and layeredon the precoated slide. Finally, the cell layer was covered by75 �L of 0.7% low-melting point agarose and lysed at 4�C forat least 1 h in lysing solution (2.5 M NaCl, 100 mM EDTA,10 mM Tris-base, 1% Triton X-100, and 10% DMSO) andthen washed in PBS twice. The lysed slides were then dippedinto alkaline buffer (300 mM NaOH, 1 mM EDTA, pH>13)for 30 min to allow the unwinding of the DNA and the ex-pression of alkali-labile damage. Single cell electrophoresiswas carried out in a prechilled alkaline solution at 25 V and300 mA for 30 min. After electrophoresis, the cells were neu-tralized in 0.4 mM Tris-HCl, pH 7.5 for 15 min at 4�C; theneutralization procedure was repeated three times. Then, thecells were stained with PI (50 �g/mL) for 10 min. To visualizethe DNA damage, the PI-stained DNA was observed using a20× objective lens on a fluorescence microscope (OlympusIX70-S1F). A minimum of 100 comets on each slide was an-alyzed using CASP CometScore software. The tail moment([percent of DNA in the tail]×[tail length]) was used to indicatethe degree of DNA damage.

2.11 Mitochondrial mass and ATP content

determination

The ATP content in the samples was analyzed using theATP bioluminescent somatic cell assay (FLASC) Kit (Sigma-Aldrich), which is based on the production of light throughthe reaction of ATP with added luciferase and luciferin, asrecommended in the kit manual. Briefly, 0.1 mL of ATP As-say Mix Working Solution was added to a 1.5 mL Eppendorftube and allowed to stand at room temperature for 3 min.Then, the ATP of T-2 toxin-treated or -controlled cells (1 ×106 cells/mL) was released by adding 0.1 mL of 1× somaticcell ATP releasing reagent with 0.05 mL ATP standard so-lution (0.5 �g/mL) in another new tube. Finally, 0.1 mL ofthe ATP-released cell samples were mixed with the preparedATP Assay Mix Working Solution, and the light emitted bythe reaction of ATP with luciferase and luciferin was im-mediately measured with a GLOMAX 20/20 luminometer(Promega).

The mitochondria mass was determined by using Mi-toTracker red (Molecular Probes, Eugene, OR, USA),a mitochondria-selective membrane potential-independentdye. The cells was grown on 12 mm diameter coverslips andincubated with 0.05 �g/mL T-2 toxin for 48 h. And then,the cells were incubated with 200 nM MitoTracker red for

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

Proteomics 2013, 13, 3175–3188 3179

Control 0.05 μg/ml T-2

0

20

40

60

80

100

120

0.00 0.10 0.20 0.30 0.40

T-2 concentration (μg/ml)

IC50=0.06 μg/ml

A B

Cel

l via

bilit

y (c

ontr

ol %

)

0.1 μg/ml T-2 0.2 μg/ml T-2

Figure 1. Viability of T-2 toxin-treated primary hepatocytes. (A) Cell viability was assessed using the MTT assay, measuring absorbanceat 570 nm. The viability of T-2 toxin-treated cell is expressed as a percentage of the value in control cells. Each point represents themean ± SD of three duplicate experiments. (B) Representative light micrographs of Giemsa staining under an optical microscope areshown for each treatment group.

30 min, washed three times with 1 × PBS, fixed by 5%paraformalclehyde for 10 min and then stained the nucleus by4′, 6-diamidino-2-phenylindole for 2 min. The red and bluefluorescence intensity was examined under a Zeiss AxioObserver D1 fluorescence microscope (Zeiss, Gottingen,Germany) using 63× magnification and was counted [41].

3 Results

3.1 The cytotoxicity of T-2 toxin in chicken primary

hepatocytes

To assess the toxicity of T-2 toxin in chicken primary hep-atocytes, the cell viability, and cell morphology of the T-2toxin-treated cells were analyzed. As expected, the viability ofthe chicken primary hepatocyte cells was inversely related tothe concentration of T-2 toxin after a 48-h treatment. The cal-culated IC50 of T-2 toxin was 0.062 �g/mL (Fig. 1A), demon-strating its obvious toxicity in chicken primary hepatocytes.The toxicity of T-2 toxin was further analyzed by Giemsastaining for morphological changes, which demonstrated thatthe T-2 toxin-treated cells rounded up and decreased in size,exhibiting increased intercellular spaces (Fig. 1B). The celldensity decreased distinctly with increasing of T-2 toxin con-centration (Fig. 1B). These results indicated that T-2 toxin hasobvious toxicity in chicken primary hepatocytes.

3.2 Proteomic profiles of chicken primary

hepatocytes regulated by T-2 toxin

To further understand the toxicity of T-2 toxin in primarychicken hepatocytes from a molecular perspective, the pro-

teomic profile changes induced by T-2 toxin were investigatedby comparative 2DE proteomic analysis. A total of 92 pro-tein spots from chicken primary hepatocytes that exhibited agreater than 1.3-fold difference in spot intensity after treat-ment with 0.05 �g/mL of T-2 toxin for 48 h were detectedin three replicate gels from three independent experiments(marked with numbers and crosses with lines in Fig. 2A). Ofthese 92 proteins, 53 were identified by MALDI-TOF/TOF-MS analysis, including 34 that were upregulated (≥1.3-fold)and 19 that were downregulated (≥1.3-fold) by T-2 toxin. Theaccession number, protein score, experimental mass, andpI, sequence coverage, fold change, molecular function, andsubcellular location of each differentially regulated proteinare shown in Table 1. The full name and enlarged spots ofthe identified proteins were shown in Supporting Informa-tion Table S1. Furthermore, the genes encoding 23 of theseproteins were randomly selected for further analysis by qRT-PCR, and 20 of them showed changes in expression that wereconsistent with the 2DE analysis (Supporting InformationFig. S1).

3.3 Functional networks, molecular functions, and

subcellular localizations of T-2 toxin-regulated

proteins

To further interpret these data in a biological context, theproteins from Table 1 were analyzed using IPA tool. Thethree top networks of proteins that were up- or downregu-lated by T-2 toxin treatment in chicken primary hepatocytes(Table 2, Supporting Information Fig. S2–S4) were: (i) neuro-logical disease, cancer, organismal injury, and abnormalities,

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

3180 P. Mu et al. Proteomics 2013, 13, 3175–3188

pH4 7MW (KDa) 4 7

200116

97.266.444.3

29.0

20.114.3

6.5

Control T-2 toxin

Transcriptional or translational

regulation13%

Cell cycle or cell proliferation

15%

Cell redoxhomeostasis

15%

Others14%

Protein degradation

7%

Carbohydrate metabolism

5%Transport

9%Lipid

metabolism11%

Stress response

11%

A

Cytoplasm24%

Mitochondrion34%

100% up

0% down

Golgi apparatus

1%

Peroxisome3%Extracellular

space6%

Plasma membrane

6%

Cytosol7%

Nucleus15%

Ribosomes4%

B C

Figure 2. Representative 2DE gel of T-2 toxin-treated or untreated chicken primary hepatocytes, molecular functions, and subcellularlocalizations of identified proteins. (A) Representative 2DE gel. Numbers and crosses with lines indicate protein spots that changed bymore than twofold compared to the control. (B) The annotated molecular functions of the identified proteins. (C) The subcellular localizationsof the identified proteins.

(ii) protein trafficking, auditory disease, cellular compromise,and (iii) amino acid metabolism, molecular transport, smallmolecule biochemistry. The top functional network regulatedby T-2 toxin indicates that chickens may have a high risk ofneurological disease when exposed to T-2 toxin.

Analysis of the molecular function annotations of the pro-teins regulated by T-2 toxin revealed an enrichment in cellredox homeostasis (15%), cell cycle or cell proliferation reg-ulation (15%), transcriptional or translational (13%), stressresponse (11%), lipid metabolism (11%), transport (9%), pro-tein degradation (7%), and carbohydrate metabolism (5%)(Fig. 2B). The greatest number of identified proteins wereinvolved in cell redox homeostasis, which suggested that T-2toxin might trigger an oxidative stress response in chickenprimary hepatocytes.

Subcellular localization analysis indicated that 34% of theaffected proteins were mitochondrial, 24% were cytoplasmic,15% were nuclear, 7% were cytosolic, 6% localized to theplasma membrane, 6% localized to the extracellular space,4% localized to the ribosome, 3% localized to the peroxisome

and 1% localized to the Golgi apparatus (Fig. 2C). Theseresults suggest that the T-2 toxin may induce mitochondrialchanges in chicken primary hepatocytes.

3.4 T-2 toxin induces oxidative stress and

mitochondrial enhancement

To examine whether T-2 toxin induces oxidative stress andmitochondrial changes in chicken primary hepatocytes at thecellular level, the ROS content, mitochondrial mass, and cel-lular ATP levels of T-2 toxin-treated or -untreated chickenprimary hepatocytes were determined. As expected, the pro-portion of ROS-positive cells and intensity of the ROS signalin 0.05 �g/mL T-2 toxin-treated cells increased over time,compared to the untreated cells (Fig. 3). After treatmentfor 72 h, the proportion of ROS-positive cells increased to85% (Fig. 3). Interestingly, the mitochondrial mass and cel-lular ATP level increased significantly after T-2 toxin treat-ment. After a 48-h treatment with 0.05 �g/mL T-2 toxin, the

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

Proteomics 2013, 13, 3175–3188 3181Ta

ble

1.

Pro

tein

sd

iffer

enti

ally

exp

ress

edu

po

nT

-2to

xin

trea

tmen

t(1

.3-f

old

chan

ge

or

gre

ater

,p<

0.05

)

Sp

ota)

No

.Pr

ote

inn

ame

Acc

.No

.b)

Th

eo.;

Ob

s.M

r/p

Ic)Pr

ote

insc

ore

d)

Seq

cov.

(%)e)

Pep

tid

esf)

Fold

g)

Mo

lecu

lar

fun

ctio

nh

)Lo

cati

on

i)

Cel

lred

oxh

om

eost

asis

9G

LRX

5g

i|566

0595

817

528/

7.85

;12

351/

5.28

210

409

3.1

±0.

1Ir

on

-su

lfu

rcl

ust

erb

ind

ing

Mit

och

on

dri

on

,cyt

op

lasm

17N

DU

FS8

gi|1

1809

0950

2437

9/5.

84;2

023

3/5.

1053

956

102.

0.2

Ele

ctro

ntr

ansp

ort

,oxi

dat

ion

red

uct

ion

Mit

och

on

dri

on

35H

IBA

DH

gi|5

7530

712

3566

0/8.

6;29

317/

5.79

303

5212

0.4

3-H

ydro

xyis

ob

uty

rate

deh

ydro

gen

ase

acti

vity

Mit

och

on

dri

on

47A

KR

1B1

gi|3

2691

2359

3595

8/6.

84;3

835

5/7.

5125

919

89

±1.

3A

ldo

-ket

ore

du

ctas

e(N

AD

P)

acti

vity

lyso

som

e

52G

RH

PR

gi|1

1810

4639

3507

9/6.

9;40

271/

7.68

340

258

4.8

±0.

3H

ydro

xyp

yru

vate

/gly

oxyl

ate

red

uct

ase

acti

vity

Cyt

op

lasm

60N

DU

FB10

gi|5

0755

667

2076

9/5.

98;1

730

1/6.

3534

058

1131

.1±

4.5

NA

DH

deh

ydro

gen

ase

(ub

iqu

ino

ne)

acti

vity

Mit

och

on

dri

on

67C

RY

Mg

i|118

0983

4833

302/

5.75

;30

630/

5.63

338

387

−3.3

±0.

3O

xid

ore

du

ctas

eac

tivi

tyC

yto

pla

sm,n

ucl

eus,

mit

och

on

dri

on

,pla

sma

mem

bra

ne

80T

XN

L1g

i|508

0660

849

955/

8.72

;40

849/

5.11

476

296

−2.1

±0.

3Pr

ote

ind

isu

lfid

eox

ido

red

uct

ase

acti

vity

Cyt

op

lasm

,nu

cleu

s

Cel

lcyc

leo

rce

llp

rolif

erat

ion

5N

PM

1g

i|345

3078

8038

128/

4.87

;17

136/

5.36

162

184

−2.8

±0.

3H

isto

ne

bin

din

g,R

NA

bin

din

gN

ucl

eolu

s

18A

RH

GD

IBg

i|507

2856

822

929/

5.08

;21

162/

5.07

176

263

−4.8

±0.

4G

TPa

seac

tiva

tor

acti

vity

Cyt

op

lasm

,cyt

oso

l25

CD

V3

gi|7

1896

619

2160

0/4.

95;5

318

4/5.

1213

835

6−4

.2±

0.3

Cel

lpro

lifer

atio

nC

yto

pla

sm,n

ucl

eus

58S

EP

T2

gi|3

2692

9511

3938

0/6.

24;5

105

8/7.

6310

936

6−2

.5±

0.3

GT

Pb

ind

ing

Cyt

op

lasm

62P

SM

B7

gi|2

2414

4619

2803

2/5.

51;1

793

1/7.

7941

866

16−2

.5±

0.3

Pro

tein

po

lyu

biq

uit

inat

ion

Cyt

oso

l77

RA

NG

AP

1g

i|575

2516

016

424/

5.63

;34

454/

5.12

168

5716

−3.6

±0.

3G

TPa

seac

tiva

tor

acti

vity

Cyt

op

lasm

,nu

cleu

s78

RA

NB

P1

gi|4

3142

223

739/

5.15

;26

838/

5.10

156

163

−2.1

±0.

1G

TPa

seac

tivi

tyC

yto

pla

sm,n

ucl

eus

Tran

scri

pti

on

alo

rtr

ansl

atio

nal

reg

ula

tio

n7

NP

M3

gi|1

1809

2933

1474

6/4.

29;1

715

8/5.

2820

940

8−2

.8±

0.3

Nu

clei

cac

idb

ind

ing

Nu

cleo

lus

16S

ET

gi|3

9536

1724

348/

4.97

;20

996/

5.10

187

397

−4.8

±0.

4Pr

ote

inp

ho

sph

atas

ein

hib

ito

rac

tivi

tyE

nd

op

lasm

icre

ticu

lum

,n

ucl

eus

23W

DR

61g

i|945

3681

933

426/

5.24

;35

503/

5.08

279

5112

−4.2

±0.

3H

isto

ne

H3-

K4

trim

ethy

lati

on

Cyt

op

lasm

,nu

cleu

s

26H

NR

NP

Cg

i|620

8863

422

044/

9.44

;50

635/

5.12

193

53

−2.5

±0.

3m

RN

Ab

ind

ing

Nu

cleu

s61

MR

PS

23g

i|507

5841

422

130/

6.66

;18

056/

9.01

222

205

−2.5

±0.

3S

tru

ctu

ralc

on

stit

uen

to

fri

bo

som

eM

ito

cho

nd

rio

n

74M

RP

L12

gi|1

1809

9877

2094

0/9.

66;1

593

3/5.

5727

535

6−3

.6±

0.3

Str

uct

ura

lco

nst

itu

ent

of

rib

oso

me

Mit

och

on

dri

on

90H

NR

PD

Lg

i|718

9674

131

590/

9.01

;42

858/

9.86

149

255

−2.1

±0.

1R

NA

bin

din

gN

ucl

eus,

cyto

pla

sm92

TS

FMg

i|622

8664

523

940/

9.33

;37

054/

5.67

286

5212

−2.8

±0.

3Tr

ansl

atio

nel

on

gati

on

fact

or

acti

vity

Intr

acel

lula

r,m

ito

cho

nd

rio

n,n

ucl

eus

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

3182 P. Mu et al. Proteomics 2013, 13, 3175–3188

Ta

ble

1.

Co

nti

nu

ed

Sp

ota)

No

.Pr

ote

inn

ame

Acc

.No

.b)

Th

eo.;

Ob

s.M

r/p

Ic)Pr

ote

insc

ore

d)

Seq

cov.

(%)e)

Pep

tid

esf)

Fold

g)

Mo

lecu

lar

fun

ctio

nh

)Lo

cati

on

i)

Tran

spo

rt4

TIM

M8A

gi|5

0745

652

1090

7/5.

13;1

110

5/5.

1530

068

158.

0.8

Pro

tein

targ

etto

mit

och

on

dri

on

Mit

och

on

dri

on

14P

RB

P4

gi|1

3240

321

3975

.44;

1651

3/5.

5223

271

164

±0.

4Tr

ansp

ort

erac

tivi

ty,

reti

no

l-b

ind

ing

pro

tein

,E

xtra

cellu

lar

reg

ion

19S

NA

P23

gi|3

1385

1076

2387

4/4.

9;22

602/

5.15

102

63

4.1

±0.

2Pr

ote

intr

ansp

ort

Cel

lula

rm

emb

ran

e31

NA

PG

gi|5

0736

711

3538

7/5.

48;4

495

3/5.

4917

341

102.

0.1

Intr

acel

lula

rp

rote

intr

ansp

ort

Mit

och

on

dri

on

66C

LIC

4g

i|118

1015

8735

857/

5.61

;25

194/

5.38

345

304

−4.1

±0.

2Vo

ltag

e-ga

ted

chlo

rid

ech

ann

elac

tivi

tyC

yto

pla

sm,i

ntr

acel

lula

r,m

ito

cho

nd

rio

n,n

ucl

eus,

cyto

sol

Str

ess

resp

on

se32

CN

PY

3g

i|363

7314

5325

232/

7.78

;40

483/

5.63

128

399

2.2

±0.

1In

nat

eim

mu

ne

resp

on

seE

nd

op

lasm

icre

ticu

lum

36M

PS

Tg

i|507

2878

833

486/

5.80

;33

062/

5.85

598

6718

0.3

Th

iosu

lfat

esu

lfu

rtra

nsf

eras

eac

tivi

tyC

yto

pla

sm,m

ito

cho

nd

rio

n

48A

NX

A2

gi|4

5382

533

3890

1/6.

92;3

744

5/8.

3832

151

122.

0.3

Ph

osp

ho

lipas

ein

hib

ito

rac

tivi

tyE

xtri

nsi

cto

pla

sma

mem

bra

ne

54G

LUL

gi|4

5382

781

4274

6/6.

38;4

934

5/9.

8061

636

72.

0.2

Glu

tam

ate-

amm

on

ialig

ase

acti

vity

Mit

och

on

dri

on

,Go

lgi

app

arat

us

56G

LUL

gi|4

5382

781

4274

6/6.

38;5

067

7/8.

7028

429

72.

0.3

Glu

tam

ate-

amm

on

ialig

ase

acti

vity

Mit

och

on

dri

on

65H

SP

25g

i|580

3755

819

086/

6.31

;21

282/

5.37

238

318

0.5

Mo

lecu

lar

chap

ero

nes

Peri

nu

clea

rre

gio

no

fcy

top

lasm

Lip

idm

etab

olis

m37

EC

H1

gi|3

6374

7010

2281

3/11

.0;2

616

8/7.

0725

59

42.

0.1

Iso

mer

ase

acti

vity

Mit

och

on

dri

on

38E

CH

1g

i|363

7470

1022

813/

11.0

;25

161/

6.91

122

43

3.7

±0.

3Is

om

eras

eac

tivi

tyM

ito

cho

nd

rio

n43

EC

HS

1g

i|326

9232

5723

924/

5.28

;24

025/

9.24

463

4915

6.5

±0.

3E

no

yl-C

oA

hyd

rata

seac

tivi

tyM

ito

cho

nd

rio

n

45E

CH

DC

2g

i|507

5161

630

801/

9.21

;24

704/

8.59

207

6515

3.1

±0.

5Ly

ase

acti

vity

Mit

och

on

dri

on

46E

CI2

gi |5

0734

079

3391

4/9.

03;2

580

5/8.

9220

746

123.

0.3

Iso

mer

ase

acti

vity

Mit

och

on

dri

on

64A

PO

A1

gi|2

2701

628

790/

5.45

;22

870/

5.39

410

7216

2.3

±0.

2P

ho

sph

olip

idb

ind

ing

Ext

race

llula

rre

gio

n,p

lasm

am

emb

ran

eC

arb

ohy

dra

tem

etab

olis

m44

PG

AM

1g

i|718

9598

529

051/

7.03

;24

309/

8.79

403

6217

2.4

±0.

3B

isp

ho

sph

og

lyce

rate

mu

tase

acti

vity

Cyt

oso

l

51R

NF1

9Ag

i|118

1018

0138

672/

6.41

;39

583/

8.74

348

5010

1.3

UD

P-g

luco

se4-

epim

eras

eac

tivi

tyC

yto

sol

64A

PO

A1

gi|2

2701

628

790/

5.45

;22

870/

5.39

410

7216

2.3

±0.

3P

ho

sph

olip

idb

ind

ing

Ext

race

llula

r,p

lasm

am

emb

ran

e

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

Proteomics 2013, 13, 3175–3188 3183

Ta

ble

1.

Co

nti

nu

ed

Sp

ota)

No

.Pr

ote

inn

ame

Acc

.No

.b)

Th

eo.;

Ob

s.M

r/p

Ic)Pr

ote

insc

ore

d)

Seq

cov.

(%)e)

Pep

tid

esf)

Fold

g)

Mo

lecu

lar

fun

ctio

nh

)Lo

cati

on

i)

Pro

tein

deg

rad

atio

n20

PS

MF1

gi|7

1896

121

2935

2/5.

19;2

693

2/5.

1319

147

10−2

.3±

0.2

Pro

teas

om

ein

hib

ito

rC

yto

pla

sm62

PS

MB

7g

i|224

1446

1928

032/

5.51

;17

744/

8.76

418

6616

−2.5

±0.

1Pr

ote

inp

oly

ub

iqu

itin

atio

nC

yto

sol

70C

LPP

gi|5

6269

518

2307

8/8.

76;2

230

8/6.

1029

636

62.

0.1

Ser

ine-

typ

een

do

pep

tid

ase

acti

vity

Mit

och

on

dri

on

75C

TS

Bg

i|461

9545

538

475/

5.74

;20

374/

5.11

350

344

−2.7

±0.

1C

yste

ine-

typ

een

do

pep

tid

ase

acti

vity

Ext

race

llula

rsp

ace

Oth

er22

EFH

D1

gi|7

2535

161

2696

7/5.

07;2

675

6/5.

0722

549

104.

0.3

Neu

ron

pro

ject

ion

dev

elo

pm

ent

Mit

och

on

dri

alin

ner

mem

bra

ne

27V

IMg

i|572

4009

030

092/

4.75

;60

913/

5.13

336

6815

-21

±2.

3Pr

ote

inb

ind

ing

,tr

ansc

rip

tio

nfa

cto

rP

lasm

am

emb

ran

e,cy

toso

l

28P

PA2

gi|1

1809

0079

5218

0/8.

91;4

529

1/5.

5821

65

44.

0.4

Ino

rgan

icd

iph

osp

hat

ase

acti

vity

Mit

och

on

dri

on

,cyt

op

lasm

30S

TOM

L2g

i|118

1036

3932

061/

6.14

;53

317/

5.55

259

4716

3.7

±0.

3R

ecep

tor

bin

din

gM

ito

cho

nd

rio

n,c

yto

pla

sm34

SE

PH

S1

gi|2

5576

0030

4340

8/5.

64;4

314

8/5.

6412

820

51.

0.2

Sel

enid

e,w

ater

dik

inas

eac

tivi

tyPe

roxi

som

e

50E

SD

gi|3

2691

4131

3200

1/6.

14;3

870

2/8.

8024

928

83.

0.4

S-f

orm

ylg

luta

thio

ne

hyd

rola

seac

tivi

tyC

yto

pla

sm,n

ucl

eus

63V

IMg

i|572

4009

030

092/

4.75

;62

194/

5.10

357

7218

−8.5

±1.

1Pr

ote

inb

ind

ing

,tr

ansc

rip

tio

nfa

cto

rP

lasm

am

emb

ran

e,cy

toso

l

71N

CL

gi|1

1959

1368

5857

6/4.

57;5

809

0/6.

2734

427

4−5

.8±

1.3

Nu

clei

cac

idb

ind

ing

Cyt

op

lasm

,nu

cleu

s

a)T

he

po

siti

on

so

fth

ese

spo

tsar

ed

isp

laye

din

the

gel

sin

Fig

.2.

b)

Th

eG

enb

ank

iden

tifi

cati

on

nu

mb

ero

fth

eid

enti

fied

pro

tein

s.c)

Th

eth

eore

tica

lan

do

bse

rved

rela

tive

mo

lecu

lar

mas

san

dis

oel

ectr

icp

oin

to

fid

enti

fied

pro

tein

s.d

)T

he

MA

SC

OT

sco

reo

fid

enti

fied

pro

tein

s.e)

Th

ese

qu

ence

cove

rag

eo

fp

rote

ins

inp

erce

nta

ge

ob

tain

edb

yM

ALD

I-TO

F/TO

F-M

Sid

enti

fica

tio

n.

f)N

um

ber

of

un

iqu

ep

epti

des

mat

ched

toth

ep

rote

inse

qu

ence

.g

)Po

siti

veva

lues

rep

rese

nt

up

reg

ula

tio

naf

ter

T-2

toxi

ntr

eatm

ent;

neg

ativ

eva

lues

rep

rese

nt

do

wn

reg

ula

tio

naf

ter

T-2

toxi

ntr

eatm

ent.

h)

Th

eid

enti

fied

pro

tein

sar

eg

rou

ped

acco

rdin

gto

thei

rfu

nct

ion

sb

yse

arch

ing

the

Gen

eO

nto

log

y(h

ttp

://w

ww

.gen

eon

tolo

gy.

org

/)an

dPu

bM

ed(h

ttp

://w

ww

.ncb

i.nlm

.nih

.gov

)d

atab

ases

.i)

Th

esu

bce

llula

rlo

caliz

atio

ns

are

cate

go

rize

db

yse

arch

ing

the

Gen

eO

nto

log

y(h

ttp

://w

ww

.gen

eon

tolo

gy.

org

/).

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

3184 P. Mu et al. Proteomics 2013, 13, 3175–3188

Table 2. Functional networks generated by IPA analysisa)

Associated network functions Molecules in network Score

Neurological disease, cancer,organismal injury, andabnormalities

Actin, AKR1B1, Alpha catenin, ANXA2, APOA1, ARHGDIB, caspase, caveolin, CD3,Ck2, CLIC4, creatine kinase, CRYAB, CTSB, GLUL, histone h3, HNRNPC, LDL,Mapk, NAPG, NCL, NFkB (complex), NPM1, NPM3, P38 MAPK, PGAM1, Pkc(s),PSMF1, RANBP1, Ras, RBP4, SNAP23, TCR, Vegf, VIM

47

Protein trafficking, auditorydisease, cellular compromise

ADRM1, C11orf58, CDV3, ECH1, EFHD1, ESD, GRHPR, HIBADH, IDE, JOSD1, MPST,MRPS23, MSTN, PPA2 (includes EG:27068), PSMB7, PSME1, RNF19A, SEPHS1,SNCAIP, STOML2, SUFU, TIMM9, TIMM10, TIMM44, TIMM23B, TIMM8A,TIMM8B, TOMM7, TOMM22, UBC, UBE2L3, UBE2L6, UCKL1, USP30, WDR61

37

Amino acid metabolism,molecular transport, smallmolecule biochemistry

26s Proteasome, Akt, APP, BSG (includes EG:12215), C18-ceramide, CNPY3, CRYM,ECHS1, ERK1/2, GLRX5, GPI, Hsp27, HTT, IDE, IL6, MRAS, MRPL12, NADHdehydrogenase, NADH2 dehydrogenase (ubiquinone), ND2 (includesEG:140532), NDUFB10, NDUFS8, PARK7, PDGF-AA, PSME1, PTEN, SEPT2, SET,SYN1, taurine, trehalose, TSC1, TXNL1, UTP14A, VHL

20

a) The double or single underline in the table indicates up- or downregulation by T-2 toxin, respectively.

mitochondrial mass and ATP levels of the chicken primaryhepatocytes increased by threefold, compared to untreatedcells (Fig. 4), which might indicate mitochondrial functionwas enhanced in the T-2 toxin-treated chicken primary hepa-tocytes.

Because high ROS content always causes DNA damageand induces cell apoptosis [35], and increasing ATP content isalso associated with apoptosis [42, 43], the DNA damage wasexamined and the apoptosis was examined. The comet assayshowed that the DNA tail length and DNA content in thetail increased with time after T-2 toxin treatment, suggestingan increase in DNA breakage due to T-2 toxin treatment(Supporting Information Fig. S5). PI-FITC double stainingrevealed that T-2 toxin induced cell apoptosis in chickenprimary hepatocytes, and the proportion of late apoptotic andnecrotic cells increased over time after 0.05 �g/mL T-2 toxintreatment (Supporting Information Fig. S6). In summary, T-2toxin induced oxidative stress and mitochondrial enhance-ment in chicken primary hepatocytes, which may be causedby the alteration of proteins related to cell redox homeostasisand significant changes in the mitochondrion proteome.

4 Discussion

The trichothecene mycotoxins produced by Fusarium speciesare frequent contaminants of cereal grains and products in-tended for animal consumption. T-2 toxin is the most toxictrichothecene; animals suffer from several pathological con-ditions after consuming of foodstuffs contaminated with tri-chothecenes [4].

Proteomic analyses of the effects of T-2 toxin have been per-formed previously to study the metabolic pathways in porcineprimary hepatocytes [34]. Here, we mainly focused on the toxi-city of T-2 toxin in chicken using proteomic analysis. Through2DE and MALDI-TOF/TOF-MS, we identified 53 T-2 toxin-regulated proteins, which were mainly involved in cell redoxhomeostasis, cell cycle or cell proliferation, transcriptional ortranslational regulation, stress response, lipid metabolism,

transport, carbohydrate metabolism, or protein degradation.The proteins in these eight groups are generally consistentwith the previously reported toxic effects of T-2 toxin, such asoxidative stress and the inhibition of protein synthesis. Theinduction of oxidative stress by T-2 toxin has been reportedin many cell types, including rat ovarian granulosa cells [44],THP-1 monocytes [45], and chicken growth plate chondro-cytes [17]. These results indicate that oxidative stress may bea common effect of T-2 toxin. Another well-known effect ofT-2 toxin is the inhibition of protein synthesis through bind-ing to peptidyl transferase, which is an integral component ofthe 60S ribosomal subunit [7,46]. In this study, another threetranslation-related enzymes, TSFM, MRPS23, and MRPL12,were also found to be regulated by T-2 toxin (Table 1). Thesethree enzymes may also be related to the inhibition of proteinsynthesis by T-2 toxin. Apart from translation, three proteinsrelated to transcription, including SET, HNRNPC, and NPM3were identified (Table 1), which may be related to the inhibi-tion of RNA synthesis by T-2 toxin.

In addition to these proteins that may be related with thosewidely considered action of T-2 toxin, two subunits of mito-chondrial complex I, NDUFB10 and NDUFS8, were foundto be regulated by T-2 toxin in chicken primary hepatocytes(Table 1). In particular, NDUFB10 was upregulated morethan 20-fold by T-2 toxin (Table 1). Similarly, some mem-bers of mitochondrial complex I are regulated by T-2 toxinin porcine primary hepatocytes [34]. Mitochondrial complexI has been shown to be related to many neurological dis-eases in humans, such as Parkinson’s disease and Leighsyndrome [47, 48]. In the present study, the top functionalnetworks of T-2 toxin-regulated proteins generated by IPAanalysis were neurological disease, cancer, organismal in-jury, and abnormalities (Table 2, Supporting InformationFig. S2). Hence, chicken feed containing T-2 toxin may bea high risk factor for neurological disease. However, the hy-pothesis that the correlation of the function of proteins iden-tified in chicken primary hepatocytes with the action of T-2toxin, such as inhibiting protein, DNA or RNA synthesis andneurological disease requires further information.

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

Proteomics 2013, 13, 3175–3188 3185

Figure 3. ROS production in T-2 toxin-treated or untreated chicken primary hepatocytes. Chicken primary hepatocytes were untreated(Control) or treated with 0.05 �g/mL of T-2 toxin for 12, 24, 48, and 72 h and exposed to DCFH-DA, followed by flow cytometry analysis.The data shown are representative dot-plot graphs of different groups of cells from three independent experiments. The M1 and M2 zonesindicate the fluorescence intensity of ROS-negative and -positive cells, respectively. Data are expressed as the mean% ± SEM of fluorescentcells in different groups from three independent experiments.

Most of the identified proteins under T-2 toxin treatmentwere localized to the mitochondrion, and all the identified mi-tochondrial proteins were upregulated by T-2 toxin (Fig. 2C,Table 1) that inspired our interest in the function of themitochondria in T-2 toxin-treated cells. The major role ofmitochondrion is to supply power to the cell through theaerobic metabolism of acetyl-CoA by the tricarboxylic acid cy-cle and oxidative phosphorylation, which produces ATP [49].ATP is used for many biological processes, such as mus-cle contraction, the synthesis, and degradation of biologicalmolecules, membrane transport, and apoptosis [50]. Interest-ingly, the mitochondrial mass and ATP content of chicken pri-mary hepatocytes increased significantly after treatment with0.05 �g/mL T-2 toxin (Fig. 4). It was reported that oxida-tive stress could induce mitochondrion mass increase, whichsuggested being a feedback mechanism that compensatedfor defects in mitochondria harboring mutated mitochon-drial DNA and a defective respiratory system [51]. In thisstudy, the T-2 toxin can induce oxidative stress to chicken pri-mary hepatocytes. Hence, we propose that the mitochondrionmass and ATP increase may be also a compensational mech-anism for the defects, such as DNA damage which brought

by T-2 toxin. Changes in mitochondrial protein expressionhave been proposed to alter the coupling efficiency of mito-chondrial oxidative phosphorylation, which influences ATPgeneration, and ROS generation [52]. The ROS content of T-2toxin-treated cells increased significantly compared to the un-treated cells (Fig. 3), which may also be partly due to the largevariation in mitochondria protein expression. The other ma-jor role of mitochondrion is maintaining oxidation–reduction(redox) balance [49]. Mitochondria are the major sites of cel-lular ROS production, which occurs through the reduction ofmolecular oxygen to superoxide by Complexes I and III of theelectron transport chain [53, 54]. Two subunits of Complex Iwere upregulated by T-2 toxin in chicken primary hepatocytes(Table 1), which may trigger ROS production and thereby in-duce ROS accumulation.

In summary, the toxicity of T-2 toxin in chicken pri-mary hepatocytes was investigated by proteomic analysis. Itwas found that T-2 toxin-induced oxidative stress and mi-tochondrion enhancement in chicken primary hepatocytes,which may be caused by changes in the expression of pro-teins related to cell oxidative stress and the mitochondrialproteome.

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

3186 P. Mu et al. Proteomics 2013, 13, 3175–3188

Figure 4. Mitochondrial mass and ATP content changes inducedby T-2 toxin. (A) Representative MitoTracker (red) and nuclear(blue) staining of untreated (Control) and 48 h treated chicken pri-mary hepatocytes by 0.05 �g/mL T-2 toxin (T-2 toxin). (B) Ratio ofthe intensity of MitoTracker/nuclear staining in chicken primaryhepatocytes treated with 0.05 �g/mL T-2 toxin for 48 h. (C) ATPcontent of 0.05 �g/mL T-2 toxin treated chicken primary hepato-cytes relative to control.

This work was supported by the National Basic Research Pro-gram of China (973 Program) [Grant 2009CB118802]; the Na-tional Natural Science Foundation of China [Grant 31172087];the Specialized Research Fund for the Doctoral Program of HigherEducation of China [Grant 20114404110010]; the Guang-dong Natural Science Foundation [Grant S2012040007589]; theChina Postdoctoral Science Foundation [Grant 2012M511577];

The authors have declared no conflict of interest.

5 References

[1] Li, Y., Wang, Z., Beier, R. C., Shen, J. et al., T-2 toxin, atrichothecene mycotoxin: review of toxicity, metabolism,and analytical methods. J. Agric. Food Chem. 2011, 59,3441–3453.

[2] Desjardins, A. E., Hohn, T. M., McCormick, S. P., Tri-chothecene biosynthesis in Fusarium species: chemistry, ge-netics, and significance. Microbiol. Rev. 1993, 57, 595–604.

[3] Kalantari, H., Moosavi, M. Review on T-2 toxin. JJNPP 2010,5, 26–38.

[4] Paterson, R. R., Lima, N., Toxicology of mycotoxins. EXS2010, 100, 31–63.

[5] Adejumo, T. O., Hettwer, U., Karlovsky, P., Occurrence ofFusarium species and trichothecenes in Nigerian maize. Int.J. Food Microbiol. 2007, 116, 350–357.

[6] Curtui, V., Usleber, E., Dietrich, R., Lepschy, J., Martlbauer,E., A survey on the occurrence of mycotoxins in wheat andmaize from western Romania. Mycopathologia 1998, 143,97–103.

[7] Bennett, J. W., Klich, M., Mycotoxins. Clin. Microbiol. Rev.2003, 16, 497–516.

[8] Sokolovic, M., Garaj-Vrhovac, V., Simpraga, B., T-2 toxin:incidence and toxicity in poultry. Arh. Hig. Rada Toksikol.2008, 59, 43–52.

[9] Schollenberger, M., Muller, H. M., Rufle, M., Suchy, S. et al.,Natural occurrence of 16 Fusarium toxins in grains and feed-stuffs of plant origin from Germany. Mycopathologia 2006,161, 43–52.

[10] Tobias, S., Rajic, I., Vanyi, A., Effect of T-2 toxin on egg pro-duction and hatchability in laying hens. Acta Vet. Hung. 1992,40, 47–54.

[11] Diaz, G. J., Squires, E. J., Julian, R. J., Boermans, H. J., Indi-vidual and combined effects of T-2 toxin and DAS in layinghens. Br. Poult. Sci. 1994, 35, 393–405.

[12] Wyatt, R. D., Doerr, J. A., Hamilton, P. B., Burmeister, H. R.,Egg production, shell thickness, and other physiological pa-rameters of laying hens affected by T-2 toxin. Appl. Micro-biol. 1975, 29, 641–645.

[13] Lee, J. T., Jessen, K. A., Beltran, R., Starkl, V. et al., Effects ofmycotoxin-contaminated diets and deactivating compoundin laying hens: 2. Effects on white shell egg quality and char-acteristics. Poult. Sci. 2012, 91, 2096–2104.

[14] Wyatt, R. D., Hamilton, P. B., Burmeister, H. R., Altered feath-ering of chicks caused by T-2 toxin. Poult. Sci. 1975, 54,1042–1045.

[15] Pande, V. V., Kurkure, N. V., Bhandarkar, A. G., Effect of T-2toxin on growth, performance and haematobiochemical al-terations in broilers. Indian J. Exp. Biol. 2006, 44, 86–88.

[16] Frankic, T., Pajk, T., Rezar, V., Levart, A., Salobir, J., The roleof dietary nucleotides in reduction of DNA damage inducedby T-2 toxin and deoxynivalenol in chicken leukocytes. FoodChem. Toxicol. 2006, 44, 1838–1844.

[17] He, S. J., Hou, J. F., Dai, Y. Y., Zhou, Z. L., Deng, Y. F., N-acetyl-cysteine protects chicken growth plate chondrocytes fromT-2 toxin-induced oxidative stress. J. Appl. Toxicol. 2011, 32,980–985.

[18] Dvorska, J. E., Pappas, A. C., Karadas, F., Speake, B. K., Surai,P. F., Protective effect of modified glucomannans and or-ganic selenium against antioxidant depletion in the chickenliver due to T-2 toxin-contaminated feed consumption.Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2007, 145,582–587.

[19] Huang, P., Akagawa, K., Yokoyama, Y., Nohara, K. et al., T-2toxin initially activates caspase-2 and induces apoptosis inU937 cells. Toxicol. Lett. 2007, 170, 1–10.

[20] Bouaziz, C., Abid-Essefi, S., Bouslimi, A., El Golli, E., Bacha,H., Cytotoxicity and related effects of T-2 toxin on culturedVero cells. Toxicon 2006, 48, 343–352.

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

Proteomics 2013, 13, 3175–3188 3187

[21] Bouaziz, C., Sharaf El Dein, O., El Golli, E., Abid-Essefi, S.et al., Different apoptotic pathways induced by zearalenone,T-2 toxin and ochratoxin A in human hepatoma cells. Toxi-cology 2008, 254, 19–28.

[22] Shinozuka, J., Suzuki, M., Noguchi, N., Sugimoto, T. et al., T-2toxin-induced apoptosis in hematopoietic tissues of mice.Toxicol. Pathol. 1998, 26, 674–681.

[23] Albarenque, S. M., 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.

[24] Grizzle, J. M., Kersten, D. B., McCracken, M. D., Houston,A. E., Saxton, A. M., Determination of the acute 50% lethaldose T-2 toxin in adult bobwhite quail: additional studieson the effect of T-2 mycotoxin on blood chemistry andthe morphology of internal organs. Avian Dis. 2004, 48,392–399.

[25] Sehata, S., Kiyosawa, N., Makino, T., Atsumi, F. et al., Mor-phological and microarray analysis of T-2 toxin-inducedrat fetal brain lesion. Food Chem. Toxicol. 2004, 42,1727–1736.

[26] Dugyala, R. R., Kim, Y. W., Sharma, R. P., Effects of aflatoxinB1 and T-2 toxin on the granulocyte-macrophage progenitorcells in mouse bone marrow cultures. Immunopharmacol-ogy 1994, 27, 57–65.

[27] Lautraite, S., Parent-Massin, D., Rio, B., Hoellinger, H., Com-parison of toxicity induced by T-2 toxin on human and ratgranulo-monocytic progenitors with an in vitro model. Hum.Exp. Toxicol. 1995, 14, 672–678.

[28] Parent-Massin, D., Haematotoxicity of trichothecenes. Toxi-col. Lett. 2004, 153, 75–81.

[29] Lafarge-Frayssinet, C., Decloitre, F., Mousset, S., Martin, M.,Frayssinet, C., Induction of DNA single-strand breaks by T-2toxin, a trichothecene metabolite of Fusarium: effect on lym-phoid organs and liver. Muta. Res. /Genet. Toxicol. 1981, 88,115–123.

[30] Li, M., Harkema, J. R., Islam, Z., Cuff, C. F., Pestka, J. J.,T-2 toxin impairs murine immune response to respiratoryreovirus and exacerbates viral bronchiolitis. Toxicol. Appl.Pharmacol. 2006, 217, 76–85.

[31] Bunner, D. L., Morris, E. R., Alteration of multiple cell mem-brane functions in L-6 myoblasts by T-2 toxin: an importantmechanism of action. Toxicol. Appl. Pharmacol. 1988, 92,113–121.

[32] Kamalavenkatesh, P., Vairamuthu, S., Balachandran, C.,Manohar, B. M., Raj, G. D., Immunopathological effect ofthe mycotoxins cyclopiazonic acid and T-2 toxin on broilerchicken. Mycopathologia 2005, 159, 273–279.

[33] Hymery, N., Sibiril, Y., Parent-Massin, D., In vitro effects oftrichothecenes on human dendritic cells. Toxicol. In Vitro2006, 20, 899–909.

[34] Wang, J., Jiang, J., Zhang, H., Cai, H. et al., Integrated tran-scriptional and proteomic analysis with in vitro biochemicalassay reveal the important role of CYP3A46 in T-2 toxin hy-droxylation in porcine primary hepatocytes. Mol. Cell. Pro-teomics 2011, 10, M111. 008748.

[35] Liu, J., Ouyang, M., Jiang, J., Mu, P. et al., Mequindox in-

duced cellular DNA damage via generation of reactive oxy-gen species. Mutat. Res. 2012, 741, 70–75.

[36] Hamer, M. J., Dickson, A. J., Control of glycolysis in culturedchick embryo hepatocytes. Fructose 2,6-bisphosphate con-tent and phosphofructokinase-1 activity are stimulated byinsulin and epidermal growth factor. Biochem. J. 1990, 269,685–690.

[37] Mosmann, T., Rapid colorimetric assay for cellular growthand survival: application to proliferation and cytotoxicity as-says. J. Immunol. Methods 1983, 65, 55–63.

[38] Yan, J. X., Wait, R., Berkelman, T., Harry, R. A. et al., A modi-fied silver staining protocol for visualization of proteins com-patible with matrix-assisted laser desorption/ionization andelectrospray ionization-mass spectrometry. Electrophoresis2000, 21, 3666–3672.

[39] Livak, K. J., Schmittgen, T. D., Analysis of relativegene expression data using real-time quantitative PCRand the 2(-Delta Delta C(T)) method. Methods 2001, 25,402–408.

[40] Olive, P. L., Wlodek, D., Durand, R. E., Banath, J. P., Fac-tors influencing DNA migration from individual cells sub-jected to gel electrophoresis. Exp. Cell Res. 1992, 198,259–267.

[41] Parra, V., Eisner, V., Chiong, M., Criollo, A. et al., Changesin mitochondrial dynamics during ceramide-induced car-diomyocyte early apoptosis. Cardiovasc. Res. 2008, 77,387–397.

[42] Richter, C., Schweizer, M., Cossarizza, A., Franceschi, C., Con-trol of apoptosis by the cellular ATP level. FEBS Lett. 1996,378, 107–110.

[43] Zamaraeva, M. V., Sabirov, R. Z., Maeno, E., Ando-Akatsuka,Y. et al., Cells die with increased cytosolic ATP during apop-tosis: a bioluminescence study with intracellular luciferase.Cell Death Differ. 2005, 12, 1390–1397.

[44] Wu, J., Jing, L., Yuan, H., Peng, S. Q., T-2 toxin induces apop-tosis in ovarian granulosa cells of rats through reactive oxy-gen species-mediated mitochondrial pathway. Toxicol. Lett.2011, 202, 168–177.

[45] Rakkestad, K. E., Skaar, I., Ansteinsson, V. E., Solhaug, A.et al., DNA damage and DNA damage responses in THP-1monocytes after exposure to spores of either Stachybotryschartarum or Aspergillus versicolor or to T-2 toxin. Toxicol.Sci. 2010, 115, 140–155.

[46] Shifrin, V. I., Anderson, P., Trichothecene mycotoxins triggera ribotoxic stress response that activates c-Jun N-terminal ki-nase and p38 mitogen-activated protein kinase and inducesapoptosis. J. Bio. Chem. 1999, 274, 13985–13992.

[47] Smeitink, J., van den Heuvel, L., Human mitochondrial com-plex I in health and disease. Am. J. Hum. Genet. 1999, 64,1505–1510.

[48] Navarro, A., Boveris, A., Brain mitochondrial dysfunctionand oxidative damage in Parkinson’s disease. J. Bioenerg.Biomembr. 2009, 41, 517–521.

[49] Morris, E. M., Rector, R. S., Thyfault, J. P., Ibdah, J. A., Mi-tochondria and redox signaling in steatohepatitis. Antioxid.Redox Signal. 2011, 15, 485–504.

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

3188 P. Mu et al. Proteomics 2013, 13, 3175–3188

[50] Brookes, P. S., Yoon, Y., Robotham, J. L., Anders, M.W., Sheu, S. S., Calcium, ATP, and ROS: a mito-chondrial love-hate triangle. Am .J. Physiol. 2004, 287,C817–833.

[51] Lee, H. C., Yin, P. H., Lu, C. Y., Chi, C. W., Wei, Y. H., In-crease of mitochondria and mitochondrial DNA in responseto oxidative stress in human cells. Biochem. J. 2000, 348,425–432.

[52] Wallace, D. C., Fan, W., Procaccio, V., Mitochondrial en-

ergetics and therapeutics. Ann. Rre. Pathol. 2010, 5,297–348.

[53] Kushnareva, Y., Murphy, A. N., Andreyev, A., Complex I-mediated reactive oxygen species generation: modulationby cytochrome C and NAD(P)+ oxidation-reduction state.Biochem. J. 2002, 368, 545–553.

[54] Pessayre, D., Fromenty, B., Mansouri, A., Mitochondrial in-jury in steatohepatitis. Eur. J. Gastroenterol. Hepatol. 2004,16, 1095–1105.

C© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.com

本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

提供一站式文献检索和下载服务”的24 小时在线不限IP

图书馆。

图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

图书馆导航:

图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具