Elsevier

Gene

Volume 452, Issue 2, 1 March 2010, Pages 45-53
Gene

Comparative genomics identifies new alpha class genes within the avian glutathione S-transferase gene cluster

https://doi.org/10.1016/j.gene.2009.11.001Get rights and content

Abstract

Glutathione S-transferases (GSTs: EC2.5.1.18) are a superfamily of multifunctional dimeric enzymes that catalyze the conjugation of glutathione (GSH) to electrophilic chemicals. In most animals and in humans, GSTs are the principal enzymes responsible for detoxifying the mycotoxin aflatoxin B1 (AFB1) and GST dysfunction is a known risk factor for susceptibility towards AFB1. Turkeys are one of the most susceptible animals known to AFB1, which is a common contaminant of poultry feeds. The extreme susceptibility of turkeys is associated with hepatic GSTs unable to detoxify the highly reactive and electrophilic metabolite exo-AFB1-8,9-epoxide (AFBO). In this study, comparative genomic approaches were used to amplify and identify the α-class tGST genes (tGSTA1.1, tGSTA1.2, tGSTA1.3, tGSTA2, tGSTA3 and tGSTA4) from turkey liver. The conserved GST domains and four α-class signature motifs in turkey GSTs (with the exception of tGSTA1.1 which lacked one motif) confirm the presence of hepatic α-class GSTs in the turkey. Four signature motifs and conserved residues found in α-class tGSTs are (1) xMExxxWLLAAAGVE, (2) YGKDxKERAxIDMYVxG, (3) PVxEKVLKxHGxxxL and (4) PxIKKFLXPGSxxKPxxx. A BAC clone containing the α-class GST gene cluster was isolated and sequenced. The turkey α-class GTS genes genetically map to chromosome MGA2 with synteny between turkey and human α-class GSTs and flanking genes. This study identifies the α-class tGST gene cluster and genetic markers (SNPs, single nucleotide polymorphisms) that can be used to further examine AFB1 susceptibility and resistance in turkeys. Functional characterization of heterologously expressed proteins from these genes is currently underway.

Introduction

Glutathione S-transferases (GSTs; E.C.2.5.1.18), a superfamily of multifunctional dimeric proteins, are important phase II biotransformation enzymes involved in cellular detoxification and excretion of a variety of xenobiotic substances (Eaton and Bammler, 1999, Frova, 2006). Carcinogens, environmental toxins and products of oxidative stress are detoxified by GSTs which principally catalyze the conjugation of reactive, electrophilic atoms with reduced glutathione (GSH) (Konishi et al., 2005, Salinas and Wong, 1999). Because of their importance in disease resistance, cancer susceptibility, and responsiveness to drug therapy, mammalian GSTs have been intensively studied. GSTs are primarily cytosolic enzymes, but microsomal forms also exist (Kelner et al., 1996). Cytosolic GSTs exist as dimeric subunits of 23–30 k Da with an average length of 199–244 amino acids (Hayes and Pulford, 1995, Mannervik and Danielson, 1988). Each subunit is composed of two spatially distinct domains. The N-terminal domain I has an α/β structure consisting of four β-strands and three α-helices. Domain II contains a larger α domain with five to six α-helices. There are two ligand-binding sites per subunit: a specific GSH-binding site (G-site) and the hydrophobic substrate binding site (H-site) (Frova, 2006, Sun et al., 1998).

Cytosolic GSTs from human, rat, and mouse have been well studied and are assigned to one of seven classes [alpha (α), mu (μ), pi (π), theta (τ), sigma (σ), zeta (ζ), omega (ο)] based on amino acid similarities (Frova, 2006, Hayes et al., 2005). Human GSTs are diverse and most abundantly expressed in the liver. Members of each class tend to have high sequence identity (> 60%)(Board, 1998) and individual genes for each human GST class are clustered together on the same chromosome (Board and Webb, 1987). Human α-class GSTs (hGSTA) are well documented with five functional genes (hGSTA1-hGSTA5) and seven pseudogenes on chromosome 6p12.1-6p12.2 (Coles and Kadlubar, 2005, Morel et al., 2002).

Avian GSTs comprise a complex isoenzyme system that has received much less attention (Yeung and Gidari, 1980). According to electrophoretic mobility on SDS/PAGE, five groups of GST subunits (designated CL1–CL5) have been identified in the cytosolic fraction of Leghorn chick livers (Chang et al., 1990). Searches of Expressed Sequence Tag (EST) databases have isolated α (Chang et al., 1990, Chang et al., 1992, Liu et al., 1993), μ (Liu and Tam, 1991, Sun et al., 1998), τ (Hsiao et al., 1995) and σ (Thomson et al., 1998) classes from cDNA sequences of the domestic chicken. Full-length cDNA of α-class GSTs was isolated and heterologously expressed in baculovirus and Escherichia coli system using GST-specific substrates (Liu et al., 1997, Liu et al., 1993). Nine class-alpha isozymes with distinctive molecular masses were affinity purified from chicken livers and partially cloned and characterized (Hsieh et al., 1999); clustering of chicken ESTs accessioned in Genbank suggests expression of six separate α-class transcripts. The nomenclature of chicken α-class GSTs was recently re-named (GenBank accession nos.) as cGSTA1 (NM_001001777), cGSTA2 (NM_001001776), cGSTA3 (NM_204818), and cGSTA-CL3 (M38219) based on subunit nomenclature proposed by (Mannervik et al., 1992).

In nearly all animals studied, GSTs are the principal detoxification enzymes for aflatoxin B1 (AFB1), a ubiquitous food and feed-borne mycotoxin that is a potent animal and human hepatotoxin and carcinogen (Coulombe, 1993, Newberne and Butler, 1969). To exert its toxic and carcinogenic effects, AFB1 requires metabolic activation to the highly reactive electrophilic and carcinogenic intermediate, the exo-AFB1-8,9-epoxide (AFBO), catalyzed by hepatic cytochromes P450 (CYPs) (Hayes et al., 1991b, Swenson et al., 1975). When functional, GSTs catalyze the conjugation of GSH to AFBO, thereby rendering it non-toxic and easily excretable. We recently amplified and cloned from turkey CYP1A5 and CYP3A37, high-affinity enzymes mostly responsible for AFB1 bioactivation in turkey liver (Rawal et al., 2009, Yip and Coulombe, 2006).

In rodents, α-class GSTs are the most efficient isozymes in detoxifying the AFBO (Hayes et al., 1991a, Hayes et al., 1991b). Mouse liver cytosol almost exclusively conjugates the exo-AFBO through the activity of its α-class GST (Raney et al., 1992), designated Gsta3, which has a high affinity toward AFBO, has been shown to be critical in the relative resistance of mice toward AFB1 (Ramsdell and Eaton, 1990). Rat constitutively express only small amount of α-class GST with high AFBO activity (rGSTA5-5) and thus are sensitive to AFB1-induced hepatocarcinogenesis (Hayes and Pulford, 1995, Wang et al., 2002). In contrast to rodents, constitutively expressed human hepatic α-class GSTs has little or no AFBO detoxifying activity(Raney et al., 1992, Slone et al., 1995).

Turkeys are one of the most susceptible animals known to AFB1 (Giambrone et al., 1985, Hamilton et al., 1972). Even small amounts in the diet cause severe hepatotoxicosis and reduction in growth rate, feed efficiency and hatchability, acute hepatic necrosis, and increased susceptibility to bacterial and viral diseases(Kubena et al., 1995, Pier et al., 1980).

There is currently little information available on the nature of GSTs in turkeys. Using prototype substrates we have demonstrated that turkey liver possesses active GST activity, but none with measurable GST-mediated detoxification of AFB1 (Klein et al., 2000, Klein et al., 2002, Klein et al., 2003). The purpose of this study was to fully characterize the α-class GSTs of the turkey. We amplified six α-class GSTs (tGSTs) from turkey liver mRNA by RACE and genetically mapped them to turkey chromosome MGA2. The CHORI-260 turkey BAC library was screened to identify clones containing the α-class gene cluster and one clone was fully sequenced and assembled. The six α-class GST genes were annotated according to gene structure, sequence similarity and synteny with chicken and human α-class GSTs. This study provides the complete sequence of the α-class genes and genetic markers that will be important in future studies of AFB1 susceptibility and resistance in turkeys.

Section snippets

RNA extraction and Rapid Amplification of cDNA Ends (RACE)

Male day-old turkey poults (Nicholas commercial strain) were obtained from Moroni Feed Co. (Moroni, UT). Freshly isolated turkey livers were stored in RNAlater (Ambion). Samples were homogenized using a Polytron (Brinkman) and mRNA was extracted using Oligotex Direct mRNA kit (Qiagen). The first strand cDNA was synthesized using MMLV reverse transcriptase (Clontech) and each 5′-CDS primer and 3′-CDS primer provided in SMART™ RACE cDNA Amplification Kit (Clontech), respectively, to carry out

Amplification of turkey α-class GST genes

The full-length cDNAs of six α-class GST genes were isolated and amplified from turkey liver using 5′- and 3′-RACE. Availability of the extensive chicken EST database which is genetically close to turkey (http://www.ncbi.nlm.nih.gov/genome/guide/chicken/ enabled the design of gene-specific primers to amplify turkey α-class GST genes (Table 1, Table 2). Primers for cGSTA1 amplified the three related A1 genes, tGSTA1.1, tGSTA1.2 and tGSTA1.3. Predicted open reading frames (tGSTA1.1, 743 bp with

Conclusions

The extreme sensitivity of turkeys is strongly associated with unresponsiveness of hepatic GSTs toward AFBO. We have shown that while turkey livers contain catalytically-active GSTs, none of which possess affinity toward AFBO (Klein et al., 2000, Klein et al., 2002, Klein et al., 2003), a situation similar in humans where constitutively expressed hepatic α-class GSTs have little or no AFBO detoxifying activity (Raney et al., 1992, Slone et al., 1995). Despite the large impact of this mycotoxin

Acknowledgements

The authors thank L.D. Chaves for assistance in BAC screening and sequence annotation. We also thank Dr. Lynn Bagley of the Moroni Feed Cooperative for generously providing turkeys and feed for this study. This research was supported in part by NRI competitive grant 2004-35205-14217 from the USDA-CSREES, by competitive grant 2007-35205-17880 from the USDA CSREES, Animal Genome program, and support from the Minnesota, and Utah and Agricultural Experiment Stations, where this is published as

References (59)

  • KonishiT. et al.

    Molecular cloning and characterization of alpha-class glutathione S-transferase genes from the hepatopancreas of red sea bream, Pagrus major

    Comp. Biochem. Physiol. C Toxicol. Pharmacol.

    (2005)
  • KubenaL.F. et al.

    Effects of feeding fumonisin B1 present in Fusarium moniliforme culture material and aflatoxin singly and in combination to turkey poults

    Poult. Sci.

    (1995)
  • LiuL.F. et al.

    Nucleotide sequence of a class mu glutathione S-transferase from chicken liver

    Biochim. Biophys. Acta

    (1991)
  • LiuL.F. et al.

    Nucleotide sequence of class-alpha glutathione S-transferases from chicken liver

    Biochim. Biophys. Acta

    (1993)
  • MannervikB. et al.

    Nomenclature for mammalian soluble glutathione transferases

    Methods Enzymol.

    (2005)
  • NuccetelliM. et al.

    Shifting substrate specificity of human glutathione transferase (from class Pi to class alpha) by a single point mutation

    Biochem. Biophys. Res. Commun.

    (1998)
  • RamsdellH.S. et al.

    Mouse liver glutathione S-transferase isoenzyme activity toward aflatoxin B1-8,9-epoxide and Benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide

    Toxicol. Appl. Pharm.

    (1990)
  • SchiothH.B. et al.

    Remarkable synteny conservation of melanocortin receptors in chicken, human, and other vertebrates

    Genomics

    (2003)
  • SunY.J. et al.

    The three-dimensional structure of an avian class-mu glutathione S-transferase, cGSTM1-1 at 1.94 A resolution

    J. Mol. Biol.

    (1998)
  • YeungT.C. et al.

    Purification and properties of a chicken liver glutathione S-transferase

    Arch. Biochem. Biophys.

    (1980)
  • AllardyceC.S. et al.

    The role of tyrosine-9 and the C-terminal helix in the catalytic mechanism of Alpha-class glutathione S-transferases

    Biochem. J.

    (1999)
  • AnderssonL. et al.

    Comparative genome organization of vertebrates. The First International Workshop on Comparative Genome Organization

    Mamm. Genome

    (1996)
  • BoardP.G.

    Identification of cDNAs encoding two human alpha class glutathione transferases (GSTA3 and GSTA4) and the heterologous expression of GSTA4-4

    Biochem. J.

    (1998)
  • BoardP.G. et al.

    Isolation of a cDNA clone and localization of human glutathione S-transferase 2 genes to chromosome band 6p12

    Proc. Natl. Acad. Sci. U. S. A.

    (1987)
  • ChangL.H. et al.

    Characterization of glutathione S-transferases from day-old chick livers

    Biochem.

    (1990)
  • ChangL.H. et al.

    Cloning and expression of a chick liver glutathione S-transferase CL 3 subunit with the use of a baculovirus expression system

    Biochem. J.

    (1992)
  • ChavesL.D. et al.

    Genome-wide genetic diversity of ‘Nici’, the DNA source for the CHORI-260 turkey BAC library and candidate for whole genome sequencing

    Anim. Genet.

    (2009)
  • DirrH. et al.

    X-ray crystal structures of cytosolic glutathione S-transferases. Implications for protein architecture, substrate recognition and catalytic function

    Eur. J. Biochem.

    (1994)
  • EatonD.L. et al.

    Concise review of the glutathione S-transferases and their significance to toxicology

    Toxicol. Sci.

    (1999)
  • Cited by (18)

    • Rice protein suppresses ROS generation and stimulates antioxidant gene expression via Nrf2 activation in adult rats

      2016, Gene
      Citation Excerpt :

      ARE-mediated transcriptional activation is primarily dependent on the transcription factor, Nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2). Nrf2 induces and up-regulates the expression of genes encoding antioxidant and phase II detoxification, including glutamate cysteine ligase (GCL), glutathione S-transferase (GST), heme oxygenase 1 (HO-1), NAD(P)H:quinone oxidoreductase 1 (NQO1), etc (Li and Kong, 2009; Kim et al., 2010; Niture et al., 2014). Thus, Nrf2 pathway is regarded as playing a key role in protecting against oxidative stress through activation and up-regulation of ARE-driven detoxification and antioxidant genes (Nguyen et al., 2009).

    • Antioxidant capacity responsible for a hypocholesterolemia is independent of dietary cholesterol in adult rats fed rice protein

      2014, Gene
      Citation Excerpt :

      Currently, we have limited knowledge to explain this contradiction, but the regulatory effect of rice protein on glutathione metabolism to induce an antioxidant response to oxidative stress should be taken into account. As for the most frequently suggested antioxidative mechanism, glutathione homeostasis is primarily maintained by glutathione redox cycling (Kim et al., 2010; Schwaab et al., 1995; Wu et al., 2004), direct uptake and de novo synthesis (Forman et al., 2009). In this study, the view that rice protein can improve glutathione metabolism to control oxidative stress is strongly supported by the observations that rice protein can modulate glutathione synthesis (by γ-GCS), glutathione oxidation (by GPx), and the GSSG reduction to GSH (by GR) during adult period.

    • Heterologous expression and functional characterization of avian mu-class glutathione S-transferases

      2013, Comparative Biochemistry and Physiology - C Toxicology and Pharmacology
      Citation Excerpt :

      We have also shown that liver cytosols from certain subspecies of wild turkeys possess significant GST-mediated AFBO detoxification, reflecting the relative resistance of wild turkeys to aflatoxicosis compared to their domestic counterparts (Quist et al., 2000; Kim et al., 2010). AFB1 detoxification in turkeys also closely resembles that in humans because they also lack hepatic alpha-class GSTs with activity toward AFB1 seen in mice and rats (Klein et al., 2000a, 2000b; Kim et al., 2010; Kim et al., 2010) suggesting that turkeys may represent a better model to study AFB1 toxicology than either of these rodent species (Rawal and Coulombe, 2011). While the GSTA genes in mice and rats are primarily involved in AFBO detoxification, they do not appear to be similarly involved in humans and non-human primates, where mu-class hGSTM1 and theta-class hGSTT1 (Eaton and Bammler, 1999), and GSTM2, respectively have been hypothesized as important candidates.

    • Metabolism of aflatoxin B1 in Turkey liver microsomes: The relative roles of cytochromes P450 1A5 and 3A37

      2011, Toxicology and Applied Pharmacology
      Citation Excerpt :

      Deficient or complete lack of functional GSTs with affinity toward exo-AFBO appears to be a major reason that turkeys are extremely susceptible to AFB1 (Klein et al., 2000). Our laboratory amplified, sequenced and mapped new Alpha-class genes within the GST gene cluster from turkey liver, which consists of six subunits A1.1, A1.2, A1.3, A2, A3, and A4 (Kim et al., 2010). Clarifying the role of these and other genes in AFB1 susceptibility will be greatly assisted by parallel efforts to sequence the turkey genome (Dalloul et al., 2010).

    • Developing mechanism-based and exposure biomarkers for mycotoxins in animals

      2011, Determining Mycotoxins and Mycotoxigenic Fungi in Food and Feed
    View all citing articles on Scopus
    View full text