期刊论文详细信息
BMC Veterinary Research
Growth performance, serum biochemical profile, jejunal morphology, and the expression of nutrients transporter genes in deoxynivalenol (DON)- challenged growing pigs
Tiejun Li1  Jielin Duan1  Jie Yin1  Wenkai Ren1  Liuqin He1  Peng Liao1  Li Wu1 
[1] Institute of Subtropical Agriculture, Chinese Academy of Sciences, 644# Yuandaer Road, Changsha 410125, China
关键词: Nutrition;    Growing pigs;    Amino acid transporter;    Deoxynivalenol (DON);   
Others  :  1219094
DOI  :  10.1186/s12917-015-0449-y
 received in 2014-12-03, accepted in 2015-06-02,  发布年份 2015
PDF
【 摘 要 】

Background

Fusarium infection with concurrent production of deoxynivalenol (DON) causes an increasing safety concern with feed worldwide. This study was conducted to determine the effects of varying levels of DON in diets on growth performance, serum biochemical profile, jejunal morphology, and the differential expression of nutrients transporter genes in growing pigs.

Results

A total of twenty-four 60-day-old healthy growing pigs (initial body weight = 16.3 ± 1.5 kg SE) were individually housed and randomly assigned to receive one of four diets containing 0, 3, 6 or 12 mg DON/kg feed for 21 days. Differences were observed between control and the 12 mg/kg DON treatment group with regards to average daily gain (ADG), although the value for average daily feed intake (ADFI) in the 3 mg/kg DON treatment group was slightly higher than that in control (P<0.01). The relative liver weight in the 12 mg/kg DON treatment group was significantly greater than that in the control (P<0.01), but there were no significant differences in other organs. With regard to serum biochemistry, the values of blood urea nitrogen (BUN), alkaline phosphatase (ALP), alanine aminotransferase (ALT) and aspartate amino transferase (AST) in the 3 treatment groups were higher than those in the control, and the serum concentrations of L-valine, glycine, L-serine, and L-glutamine were significantly reduced in the 3 treatment groups, especially in the 12 mg/kg DON group (P<0.01). Serum total superoxide dismutase (T-SOD), glutathione peroxidase (GSH-Px) were markedly decreased after exposure to DON contaminated feeds (P<0.01). The villi height was markedly decreased and the lymphocyte cell numbers markedly increased in the 3 DON contaminated feeds (P<0.01). The mRNA expression levels of excitatory amino acid transporter-3 (EAAC-3), sodium-glucose transporter-1 (SGLT-1), dipeptide transporter-1 (PepT-1), cationic amino acid transporter-1 (CAT-1) and y + L-type amino acid transporter-1 (LAT-1) in control were slightly or markedly higher than those in the 3 DON treatment groups.

Conclusions

These results showed that feeds containing DON cause a wide range of effects in a dose-dependent manner. Such effects includes weight loss, live injury and oxidation stress, and malabsorption of nutrients as a result of selective regulation of nutrient transporter genes such as EAAC-3, SGLT-1, PepT-1, CAT-1 and LAT-1.

【 授权许可】

   
2015 Wu et al.

【 预 览 】
附件列表
Files Size Format View
20150715040722106.pdf 1151KB PDF download
Fig. 1. 128KB Image download
【 图 表 】

Fig. 1.

【 参考文献 】
  • [1]Pestka JJ. Deoxynivalenol: mechanisms of action, human exposure, and toxicological relevance. Arch Toxicol. 2010; 84(9):663-79.
  • [2]Pestka JJ. Mechanisms of deoxynivalenol-induced gene expression and apoptosis. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 2008; 25(9):1128-40.
  • [3]Rotter BA, Prelusky DB, Pestka JJ. Toxicology of deoxynivalenol (vomitoxin). J Toxicol Environ Health. 1996; 48(1):1-34.
  • [4]Maresca M. From the gut to the brain: journey and pathophysiological effects of the food-associated trichothecene mycotoxin deoxynivalenol. Toxins (Basel). 2013; 5(4):784-820.
  • [5]Ferrari L, Cantoni AM, Borghetti P, De Angelis E, Corradi A. Cellular immune response and immunotoxicity induced by DON (deoxynivalenol) in piglets. Vet Res Commun. 2009; 33 Suppl 1:133-5.
  • [6]Kinser S, Jia Q, Li M, Laughter A, Cornwell P, Corton JC et al.. Gene expression profiling in spleens of deoxynivalenol-exposed mice: immediate early genes as primary targets. J Toxicol Environ Health A. 2004; 67(18):1423-41.
  • [7]Chaytor AC, See MT, Hansen JA, de Souza AL, Middleton TF, Kim SW. Effects of chronic exposure of diets with reduced concentrations of aflatoxin and deoxynivalenol on growth and immune status of pigs. J Anim Sci. 2011; 89(1):124-35.
  • [8]Pestka JJ, Lin WS, Miller ER. Emetic activity of the trichothecene 15-acetyldeoxynivalenol in swine. Food Chem Toxicol. 1987; 25(11):855-8.
  • [9]Hunder G, Schumann K, Strugala G, Gropp J, Fichtl B, Forth W. Influence of subchronic exposure to low dietary deoxynivalenol, a trichothecene mycotoxin, on intestinal absorption of nutrients in mice. Food Chem Toxicol. 1991; 29(12):809-14.
  • [10]Awad WA, Razzazi-Fazeli E, Bohm J, Zentek J. Effects of B-trichothecenes on luminal glucose transport across the isolated jejunal epithelium of broiler chickens. J Anim Physiol Anim Nutr (Berl). 2008; 92(3):225-30.
  • [11]Maresca M, Mahfoud R, Garmy N, Fantini J. The mycotoxin deoxynivalenol affects nutrient absorption in human intestinal epithelial cells. J Nutr. 2002; 132(9):2723-31.
  • [12]Sergent T, Parys M, Garsou S, Pussemier L, Schneider YJ, Larondelle Y. Deoxynivalenol transport across human intestinal Caco-2 cells and its effects on cellular metabolism at realistic intestinal concentrations. Toxicol Lett. 2006; 164(2):167-76.
  • [13]Pinton P, Accensi F, Beauchamp E, Cossalter AM, Callu P, Grosjean F et al.. Ingestion of deoxynivalenol (DON) contaminated feed alters the pig vaccinal immune responses. Toxicol Lett. 2008; 177(3):215-22.
  • [14]Kasuga F, Hara-Kudo Y, Saito N, Kumagai S, Sugita-Konishi Y. In vitro effect of deoxynivalenol on the differentiation of human colonic cell lines Caco-2 and T84. Mycopathologia. 1998; 142(3):161-7.
  • [15]Awad WA, Ghareeb K, Zentek J. Mechanisms underlying the inhibitory effect of the feed contaminant deoxynivalenol on glucose absorption in broiler chickens. Vet J. 2014; 1:198-190.
  • [16]Awad WA, Aschenbach JR, Setyabudi FM, Razzazi-Fazeli E, Bohm J, Zentek J. In vitro effects of deoxynivalenol on small intestinal D-glucose uptake and absorption of deoxynivalenol across the isolated jejunal epithelium of laying hens. Poult Sci. 2007; 86(1):15-20.
  • [17]Yin J, Ren W, Duan J, Wu L, Chen S, Li T et al.. Dietary arginine supplementation enhances intestinal expression of SLC7A7 and SLC7A1 and ameliorates growth depression in mycotoxin-challenged pigs. Amino Acids. 2014; 46(4):883-92.
  • [18]Wu L, Wang W, Yao K, Zhou T, Yin J, Li T et al.. Effects of dietary arginine and glutamine on alleviating the impairment induced by deoxynivalenol stress and immune relevant cytokines in growing pigs. PLoS One. 2013; 8(7): Article ID e69502
  • [19]Xiao H, Tan BE, Wu MM, Yin YL, Li TJ, Yuan DX et al.. Effects of composite antimicrobial peptides in weanling piglets challenged with deoxynivalenol: II. Intestinal morphology and function. J Anim Sci. 2013; 91(10):4750-56.
  • [20]Weaver AC, See MT, Hansen JA, Kim YB, De Souza AL, Middleton TF et al.. The use of feed additives to reduce the effects of aflatoxin and deoxynivalenol on pig growth, organ health and immune status during chronic exposure. Toxins (Basel). 2013; 5(7):1261-81.
  • [21]Shi Y, Pestka JJ. Attenuation of mycotoxin-induced IgA nephropathy by eicosapentaenoic acid in the mouse: dose response and relation to IL-6 expression. J Nutr Biochem. 2006; 17(10):697-706.
  • [22]Awad WA, Razzazi-Fazeli E, Bohm J, Zentek J. Influence of deoxynivalenol on the D-glucose transport across the isolated epithelium of different intestinal segments of laying hens. J Anim Physiol Anim Nutr (Berl). 2007; 91(5–6):175-80.
  • [23]Danicke S, Valenta H, Klobasa F, Doll S, Ganter M, Flachowsky G. Effects of graded levels of Fusarium toxin contaminated wheat in diets for fattening pigs on growth performance, nutrient digestibility, deoxynivalenol balance and clinical serum characteristics. Arch Anim Nutr. 2004; 58(1):1-17.
  • [24]Diesing AK, Nossol C, Danicke S, Walk N, Post A, Kahlert S et al.. Vulnerability of polarised intestinal porcine epithelial cells to mycotoxin deoxynivalenol depends on the route of application. PLoS One. 2011; 6(2): Article ID e17472
  • [25]Nejdfors P, Ekelund M, Jeppsson B, Westrom BR. Mucosal in vitro permeability in the intestinal tract of the pig, the rat, and man: species- and region-related differences. Scand J Gastroenterol. 2000; 35(5):501-7.
  • [26]Xiao H, Wu MM, Tan BE, Yin YL, Li TJ, Xiao DF et al.. Effects of composite antimicrobial peptides in weanling piglets challenged with deoxynivalenol: I. Growth performance, immune function and antioxidation capacity. J Anim Sci. 2013; 91(10):4772-80.
  • [27]Danicke S, Brosig B, Kersten S, Kluess J, Kahlert S, Panther P et al.. The Fusarium toxin deoxynivalenol (DON) modulates the LPS induced acute phase reaction in pigs. Toxicol Lett. 2013; 220(2):172-80.
  • [28]Rohweder D, Kersten S, Valenta H, Sondermann S, Schollenberger M, Drochner W et al.. Bioavailability of the Fusarium toxin deoxynivalenol (DON) from wheat straw and chaff in pigs. Arch Anim Nutr. 2013; 67(1):37-47.
  • [29]Moore CJ, Blaney BJ, Spencer RA, Dodman RL. Rejection by pigs of mouldy grain containing deoxynivalenol. Aust Vet J. 1985; 62(2):60-2.
  • [30]Gutzwiller A. Effects of deoxynivalenol (DON) in the lactation diet on the feed intake and fertility of sows. Mycotoxin Res. 2010; 26(3):211-5.
  • [31]Tiemann U, Danicke S. In vivo and in vitro effects of the mycotoxins zearalenone and deoxynivalenol on different non-reproductive and reproductive organs in female pigs: a review. Food Addit Contam. 2007; 24(3):306-14.
  • [32]Muller G, Kielstein P, Rosner H, Berndt A, Heller M, Kohler H. Studies on the influence of combined administration of ochratoxin A, fumonisin B1, deoxynivalenol and T2 toxin on immune and defence reactions in weaner pigs. Mycoses. 1999; 42(7–8):485-93.
  • [33]Prelusky DB, Gerdes RG, Underhill KL, Rotter BA, Jui PY, Trenholm HL. Effects of low-level dietary deoxynivalenol on haematological and clinical parameters of the pig. Nat Toxins. 1994; 2(3):97-104.
  • [34]Overnes G, Matre T, Sivertsen T, Larsen HJ, Langseth W, Reitan LJ et al.. Effects of diets with graded levels of naturally deoxynivalenol-contaminated oats on immune response in growing pigs. Zentralbl Veterinarmed A. 1997; 44(9–10):539-50.
  • [35]Swamy HV, Smith TK, MacDonald EJ, Boermans HJ, Squires EJ. Effects of feeding a blend of grains naturally contaminated with Fusarium mycotoxins on swine performance, brain regional neurochemistry, and serum chemistry and the efficacy of a polymeric glucomannan mycotoxin adsorbent. J Anim Sci. 2002; 80(12):3257-67.
  • [36]Bergsjo B, Langseth W, Nafstad I, Jansen JH, Larsen HJ. The effects of naturally deoxynivalenol-contaminated oats on the clinical condition, blood parameters, performance and carcass composition of growing pigs. Vet Res Commun. 1993; 17(4):283-94.
  • [37]Smith TK, McMillan EG, Castillo JB. Effect of feeding blends of Fusarium mycotoxin-contaminated grains containing deoxynivalenol and fusaric acid on growth and feed consumption of immature swine. J Anim Sci. 1997; 75(8):2184-91.
  • [38]Young LG, McGirr L, Valli VE, Lumsden JH, Lun A. Vomitoxin in corn fed to young pigs. J Anim Sci. 1983; 57(3):655-64.
  • [39]Rotter BA, Thompson BK, Lessard M, Trenholm HL, Tryphonas H. Influence of low-level exposure to Fusarium mycotoxins on selected immunological and hematological parameters in young swine. Fundam Appl Toxicol. 1994; 23(1):117-24.
  • [40]Swamy HV, Smith TK, MacDonald EJ, Karrow NA, Woodward B, Boermans HJ. Effects of feeding a blend of grains naturally contaminated with Fusarium mycotoxins on growth and immunological measurements of starter pigs, and the efficacy of a polymeric glucomannan mycotoxin adsorbent. J Anim Sci. 2003; 81(11):2792-803.
  • [41]Goyarts T, Grove N, Danicke S. Effects of the Fusarium toxin deoxynivalenol from naturally contaminated wheat given subchronically or as one single dose on the in vivo protein synthesis of peripheral blood lymphocytes and plasma proteins in the pig. Food Chem Toxicol. 2006; 44(12):1953-65.
  • [42]Nyblom H, Berggren U, Balldin J, Olsson R. High AST/ALT ratio may indicate advanced alcoholic liver disease rather than heavy drinking. Alcohol Alcohol. 2004; 39(4):336-9.
  • [43]Wu G. Amino acids: metabolism, functions, and nutrition. Amino Acids. 2009; 37(1):1-17.
  • [44]Wang W, Wu Z, Dai Z, Yang Y, Wang J, Wu G. Glycine metabolism in animals and humans: implications for nutrition and health. Amino Acids. 2013; 45(3):463-77.
  • [45]Wu G. Functional amino acids in growth, reproduction, and health. Adv Nutr. 2010; 1(1):31-7.
  • [46]Yin J, Ren W, Liu G, Duan J, Yang G, Wu L et al.. Birth oxidative stress and the development of an antioxidant system in newborn piglets. Free Radic Res. 2013; 47(12):1027-35.
  • [47]Robert L. Serum haptoglobin in clinical biochemistry: change of a paradigm. Pathol Biol (Paris). 2013; 61(6):277-9.
  • [48]Jiang SZ, Yang ZB, Yang WR, Gao J, Liu FX, Broomhead J et al.. Effects of purified zearalenone on growth performance, organ size, serum metabolites, and oxidative stress in postweaning gilts. J Anim Sci. 2011; 89(10):3008-15.
  • [49]Dinu D, Bodea GO, Ceapa CD, Munteanu MC, Roming FI, Serban AI et al.. Adapted response of the antioxidant defense system to oxidative stress induced by deoxynivalenol in Hek-293 cells. Toxicon. 2011; 57(7–8):1023-32.
  • [50]Mary VS, Theumer MG, Arias SL, Rubinstein HR. Reactive oxygen species sources and biomolecular oxidative damage induced by aflatoxin B1 and fumonisin B1 in rat spleen mononuclear cells. Toxicology. 2012; 302(2–3):299-307.
  • [51]Boisclair YR, Rhoads RP, Ueki I, Wang J, Ooi GT. The acid-labile subunit (ALS) of the 150 kDa IGF-binding protein complex: an important but forgotten component of the circulating IGF system. J Endocrinol. 2001; 170(1):63-70.
  • [52]Kobayashi-Hattori K, Amuzie CJ, Flannery BM, Pestka JJ. Body composition and hormonal effects following exposure to mycotoxin deoxynivalenol in the high-fat diet-induced obese mouse. Mol Nutr Food Res. 2011; 55(7):1070-8.
  • [53]Amuzie CJ, Pestka JJ. Suppression of insulin-like growth factor acid-labile subunit expression–a novel mechanism for deoxynivalenol-induced growth retardation. Toxicol Sci. 2010; 113(2):412-21.
  • [54]Amuzie CJ, Shinozuka J, Pestka JJ. Induction of suppressors of cytokine signaling by the trichothecene deoxynivalenol in the mouse. Toxicol Sci. 2009; 111(2):277-87.
  • [55]Kolf-Clauw M, Castellote J, Joly B, Bourges-Abella N, Raymond-Letron I, Pinton P et al.. Development of a pig jejunal explant culture for studying the gastrointestinal toxicity of the mycotoxin deoxynivalenol: histopathological analysis. Toxicol In Vitro. 2009; 23(8):1580-4.
  • [56]Awad WA, Bohm J, Razzazi-Fazeli E, Zentek J. Effects of feeding deoxynivalenol contaminated wheat on growth performance, organ weights and histological parameters of the intestine of broiler chickens. J Anim Physiol Anim Nutr (Berl). 2006; 90(1–2):32-7.
  • [57]Piva A, Casadei G, Pagliuca G, Cabassi E, Galvano F, Solfrizzo M et al.. Activated carbon does not prevent the toxicity of culture material containing fumonisin B1 when fed to weanling piglets. J Anim Sci. 2005; 83(8):1939-47.
  • [58]Brown TP, Rottinghaus GE, Williams ME. Fumonisin mycotoxicosis in broilers: performance and pathology. Avian Dis. 1992; 36(2):450-4.
  • [59]Bouhet S, Oswald IP. The intestine as a possible target for fumonisin toxicity. Mol Nutr Food Res. 2007; 51(8):925-31.
  • [60]Bracarense AP, Lucioli J, Grenier B, Drociunas Pacheco G, Moll WD, Schatzmayr G et al.. Chronic ingestion of deoxynivalenol and fumonisin, alone or in interaction, induces morphological and immunological changes in the intestine of piglets. Br J Nutr. 2012; 107(12):1776-86.
  • [61]Wu G. Functional amino acids in nutrition and health. Amino Acids. 2013; 45(3):407-11.
  • [62]Zhang J, Yin Y, Shu XG, Li T, Li F, Tan B et al.. Oral administration of MSG increases expression of glutamate receptors and transporters in the gastrointestinal tract of young piglets. Amino Acids. 2013; 45(5):1169-77.
  • [63]Robinson MB. The family of sodium-dependent glutamate transporters: a focus on the GLT-1/EAAT2 subtype. Neurochem Int. 1998; 33(6):479-91.
  • [64]Hwang ES, Hirayama BA, Wright EM. Distribution of the SGLT1 Na+/glucose cotransporter and mRNA along the crypt-villus axis of rabbit small intestine. Biochem Biophys Res Commun. 1991; 181(3):1208-17.
  • [65]Wright EM, Loo DD, Turk E, Hirayama BA. Sodium cotransporters. Curr Opin Cell Biol. 1996; 8(4):468-73.
  • [66]Daniel H. Molecular and integrative physiology of intestinal peptide transport. Annu Rev Physiol. 2004; 66:361-84.
  • [67]Broer S. Amino acid transport across mammalian intestinal and renal epithelia. Physiol Rev. 2008; 88(1):249-86.
  • [68]Ruan Z, Lv Y, Fu X, He Q, Deng Z, Liu W et al.. Metabolomic analysis of amino acid metabolism in colitic rats supplemented with lactosucrose. Amino Acids. 2013; 45(4):877-87.
  • [69]Wang J, Chen L, Li P, Li X, Zhou H, Wang F et al.. Gene expression is altered in piglet small intestine by weaning and dietary glutamine supplementation. J Nutr. 2008; 138(6):1025-32.
  文献评价指标  
  下载次数:7次 浏览次数:22次