You are on page 1of 10

Wageningen Academic

World Mycotoxin Journal, February 2008; 1(1): 3-12 P u b l i s h e r s

The ‘omics’ tools: genomics, proteomics, metabolomics and their potential for solving
the aflatoxin contamination problem

D. Bhatnagar1, K. Rajasekaran1, G.A. Payne2, R.L. Brown1, J. Yu1 and T.E. Cleveland1

1United States Department of Agriculture, Agricultural Research Service, Southern Regional Research Center, 1100
Robert E. Lee Blvd., New Orleans, LA 70124, USA; 2North Carolina State University, Raleigh, North Carolina, USA;
dbhatnag@srrc.ars.usda.gov

Received: 12 December 2007 / Accepted: 6 January 2008


© 2008 Wageningen Academic Publishers

Abstract

Aflatoxins are highly carcinogenic secondary metabolites produced primarily by the fungi Aspergillus flavus and
Aspergillus parasiticus. Aflatoxin contamination of food and feed is an age old problem of particular concern over
the last four decades. Now, for the first time control measures for this problem appear within reach. For practical and
sustainable control of aflatoxin contamination to be realised, however, additional information is needed rather rapidly,
particularly for understanding the specific molecular factors (both in the plant and the fungus) involved during host
plant-fungus interaction. The information derived from the use of novel tools such as genomics, proteomics and
metabolomics provides us with the best and the quickest opportunity to achieve a clear understanding of the survival
of toxigenic fungi in the field, the ability of the fungus to invade crops, and the process of toxin contamination under
various environmental conditions. Significant progress has been made recently in understanding the genomic make-
up of the most significant aflatoxin producing field fungus, namely Aspergillus flavus. Progress also has been made
in the study of host crop resistance to fungal invasion through the use of proteomics. The information available on
production of aflatoxin and other metabolites by Aspergillus flavus is reasonably extensive, although the application
of metabolomics as a tool in this study is relatively new. In this review there is a discussion of the use of genomics,
proteomics and metabolomics in deriving the requisite information for developing effective strategies to interrupt
the machinery in the fungus for production of these toxins, as well as to assist in the development of host-resistance
against fungal invasion and aflatoxin contamination of crops.

Keywords: Aspergillus flavus, aflatoxins, genomics, proteomics, metabolomics

1. Introduction in the field and during storage (Payne and Brown, 1998).
Aflatoxin contamination continues to be a serious problem
Aflatoxins are highly toxic and carcinogenic secondary in many parts of the world (Richard and Payne, 2003).
metabolites produced mainly by three anamorphic species
of the genus Aspergillus: A. flavus, A. parasiticus and A. Initially studied because of their negative impact on human
nomius (Ehrlich et al., 2003). They are the most potent, and animal health (Eaton and Gallagher, 1994), aflatoxins
naturally-occurring carcinogens known and have been are perhaps the most well-known class of mycotoxins,
linked to liver cancer and several other maladies in animals serving as a model system for the study of the genetics
and humans (Turner et al., 2003; Valdivia et al., 2001; Otim of mycotoxin biosynthesis and secondary metabolism
et al., 2005). Among the aflatoxin-producing fungi, the in general (Cary et al., 2000; Bhatnagar et al., 2003; Yu
ubiquitous A. flavus is the known pathogen of cotton, corn, et al., 2004a; Yu and Keller, 2005). Due to the acute and
peanuts and other oil-seed crops, producing toxin both chronic toxicity of aflatoxins, nearly a hundred countries

ISSN 1875-0710 print, ISSN 1875-0796 online


D. Bhatnagar et al.

are known to have regulations limiting mycotoxin levels, scope of biological investigation from studying single genes,
with 61 having specific regulatory levels for total aflatoxins individual compounds (fungal secondary metabolites)
in foodstuffs and 39 having regulations for aflatoxins in or plant proteins to studying all genes, metabolites and
feedstuffs (FAO, 2004; Van Egmond et al., 2007). The U.S. proteins at once in a systematic fashion. A. flavus genomics
Food and Drug Administration has set limits of 20 μg/kg and proteomics of seed-based resistance provide the best
total aflatoxins for interstate commerce of food and feed investigative tools for simultaneous discovery and analysis
and 0.5 μg/kg of aflatoxin M1 for sale of milk, and in the of the biochemical function and genetic regulation of the
European Union these limits are even lower. critical genes governing fungal development, plant fungal
interaction and aflatoxin biosynthesis.
Chronic, as well as sporadic, aflatoxin contamination
in a variety of field crops and agricultural commodities 2. Genomics
worldwide has had a serious impact on the economics and
food safety of these products (Jelinek et al., 1989; Henry et Since 1995, the genomes, or genetic make-up, of dozens of
al., 2002). Vardon et al. (2003) estimated the annual cost bacteria and a few model eukaryotes have been completely
from aflatoxin contamination in the USA at roughly $500 sequenced, but fungal genomics was not a priority in these
million through two categories of loss, market rejection early efforts. Despite the slow start, however, genomic
and animal health impacts. Wu (2004) has suggested sequencing has now been completed for dozens of fungi,
that the total economic impact of aflatoxins should take including species that are of fundamental biological interest,
into account many other factors such as export market species that are important to industry and agriculture,
loss, sampling and testing costs, costs to food processors, and species that cause opportunistic human infections
grocery markets and consumers, and human health effects. (Bennett and Arnold, 2001; Desjardins and Bhatnagar, 2003;
Therefore, it is imperative that researchers across the globe Fakhoury and Payne, 2003; Yu et al., 2004a; Payne et al.,
develop strategies for the effective control of aflatoxin 2006; Machida et al., 2005; Nierman et al., 2005, Galagan
contamination of crops. New biotechnologies such as (1) et al., 2005). Genomic studies on several Aspergillus and
the use of disarmed, non-toxigenic biocompetitive strains Fusarium fungal species are well underway. Among these
of A. flavus in biocontrol of aflatoxin contamination, are structural, functional and comparative genomics of
and (2) identification of plant constituents that disrupt the toxin-producing species Fusarium graminearum
aflatoxin biosynthesis or fungal growth and their use in new (trichothecene producer), F. verticillioides (fumonisin
biochemical marker-based breeding strategies to enhance producer), and A. flavus (aflatoxin producer).
resistance in crops to aflatoxin, could potentially save the
agricultural industry in the U.S. alone hundreds of millions The term ‘genome’ has existed for over 75 years and
of dollars. However, functional genomics is needed to speed refers to an organism’s complete set of chromosomes
up our understanding of the field biology of the fungus in and the genes contained therein. The term ‘genomics’,
relationship to its interaction with the host plant. But this now universally accepted, was coined in 1986 by Thomas
insight into the behaviour of these biological systems first Roderick (Kuska, 1998) to describe the scientific discipline
requires the sequence information (structural genomics) of mapping, sequencing and analysing genomes. As more
of the aflatoxigenic fungus of interest. Metabolic profiling and more genomes are sequenced, the emphasis has shifted
along with structural and functional genomics could provide from just mapping and sequencing to understanding the
significant information on the fungal responses to various functions of genome with a comparison to other sequenced
alterations in its ecology. Understanding the complex genomes. Genomics combined with bioinformatics enables
interrelationships of plant and fungal gene products during the identification of all of the genes in an organism and
the host plant-A. flavus interaction is the key to developing the study of their functions. Therefore, genome analysis is
strategies to interrupt the aflatoxin contamination process now being divided into ‘structural genomics’, ‘comparative
through enhancing host-plant resistance. Plant factors have genomics’ and ‘functional genomics’. In addition, novel
been discovered through the use of proteomics and natural terminology, new acronyms, and innovative techniques
product chemistry that may influence fungal processes have been added to the scientific vocabulary (Bennett
involved in invasion and aflatoxin contamination. These and Arnold, 2001; Desjardins and Bhatnagar, 2003).
factors can also be divided into three categories: (1) seed Structural genomics has been defined as the ‘initial phase
proteins/inhibitors of fungal cell wall degrading enzymes, of genomic analysis: with a clear end point that results in
(2) seed/kernel natural products which may influence the construction of high resolution genetic, physical and
fungal growth and/or aflatoxin synthesis, and (3) plant transcript maps of an organism’ (Hieter and Boguski, 1997).
stress responsive proteins that have been shown to have Comparative or evolutionary genomics is the comparison
an indirect effect on aflatoxin contamination. of DNA sequences of related organisms through advanced
computer technologies, or bioinformatics. Functional
Subsequently, the fundamental strategy in a genomics, genomics is the identification of the functions of each
metabolomics and proteomics approach is to expand the coding sequence through analysis of gene expression by

 World Mycotoxin Journal 1 (1)


 The ‘omics’ tools: genomics, proteomics, metabolomics and their potential for solving the aflatoxin contamination problem

using libraries of Expressed Sequence Tags (ESTs) and EST sequences were identified (Yu et al., 2004b, 2007b).
microarray technologies, by targeted gene knock-out These EST sequences are available to the public at the
experiments, and transgenic expression. NCBI GenBank Database (http://www.ncbi.nlm.nih.gov/).
The A. flavus Gene Index was constructed at TIGR and
EST technology allows rapid identification of the majority, is currently maintained and curated by The Dana Farber
if not all, of the genes expressed in the fungal genome and Cancer Institute (http://compbio.dfci.harvard.edu/tgi). The
contributes to a better understanding of their functions, identified genes have been categorised according to their
regulation, and coordination of gene expression in potential involvement, directly or indirectly, in aflatoxin
response to internal and external factors, the relationship production based on function such as in global regulation,
between primary and secondary metabolism, plant-fungal signal transduction, pathogenicity, virulence, and fungal
interaction and fungal pathogenicity, as well as evolutionary development (Yu et al., 2004b, 2007b).
biology. For a detailed discussion of this technique refer to
Bohnert et al. (2001) or Ohlrogge and Benning (2000). Whole genome sequencing of Aspergillus flavus

Microarray or chip analysis is a DNA hybridisation-based The A. flavus whole genome sequencing project was
approach (Donson et al., 2002). The technique enables funded by a United States Department of Agriculture /
the monitoring of the expression of thousands of genes in National Research Initiative (USDA/NRI) grant awarded
parallel, making it ideal for gene profiling in a genomics to Prof. Gary A. Payne and Prof. Ralph Dean, North
context (reviewed in Desjardins and Bhatnagar, 2003). DNA Carolina State University, Raleigh, North Carolina. The
microarrays consist of an ‘immobile phase’, either PCR Food and Feed Safety Research Unit of the Southern
amplified genomic sequences, cDNAs, or oligonucleotides Regional Research Center of the Agricultural Research
embedded onto a solid support such as glass or plastic Service (USDA/ARS/SRRC) provided funding for fine
slides, silicon wafers or nylon membranes (Baldwin et al., finishing and gene calling. The sequencing was completed
1999; Drăghici, 2003). at TIGR under the supervision of Dr. William C. Nierman
by a shotgun approach and Sanger sequencing protocol.
With the application of genomics and, in particular, of Primary assembly indicated that the A. flavus genome
EST and microarray technologies, we have witnessed a consists of 8 chromosomes and the genome size is about
revolution in our understanding of biological processes. 36.8 Mb with over 12,000 functional genes in the A. flavus
genome similar to that found in related Aspergillus species
Aspergillus flavus genomics (Galagan et al., 2005; Machida et al., 2005; Nierman et al.,
2005; Payne et al., 2006; Yu et al., 2007a). The annotation of
A. flavus genomics is aimed at understanding the genetic the A. flavus genome sequence data has been achieved by
control and regulation of toxin production by this using the A. flavus EST database, A. oryzae EST database,
important aflatoxigenic fungus as well as the evolutionary and the A. oryzae whole genome sequence. The availability
process in Aspergillus section Flavi (Bhatnagar et al., of the A. oryzae whole genome sequence (Machida et al.,
2002). More importantly, we need to understand what 2005) was vital in developing the chromosomal structure of
is the mechanism of toxin production in response to the A. flavus genome as well. The sequence data have been
environmental influences on the fungus, i.e. simultaneously deposited in the NCBI GenBank database (http://www.
to environmental conditions like nutrition status of crops, ncbi.nlm.nih.gov) and are also available at the A. flavus
temperature, water stress, pH, and volatile compounds website (http://www.aspergillusflavus.org).
from plants. We also need to understand the ecological/
evolutionary significance of A. flavus propagation, fungal Microarrays as tools for functional genomics studies
virulence, and aflatoxin formation as manifested by changes
in gene expression profiles and global signal transduction Several types of microarrays for A. flavus have been
within the fungus. These parameters are now being rapidly constructed within the last few years. The first cDNA
analysed using the genomic information that has been microarray consisting of 753 gene features including
recently obtained. known aflatoxin pathway genes and regulatory gene aflR
was constructed by the laboratory of Prof. Gary A. Payne,
Aspergillus flavus expressed sequence tags North Carolina State University. The unique ESTs identified
from a cDNA library constructed using A. flavus RNA
An A. flavus EST project has been completed using the wild under aflatoxin-producing condition were spotted on a
type strain NRRL 3357 (ATCC# 20026). Over 26,110 cDNA Telechem SuperAldehyde glass slide using an Affymetrix
clones from a normalised cDNA expression library were 417 Arrayer (O’Brian et al., 2003). A 5,031 gene-elements
sequenced at The Institute for Genomic Research (TIGR; A. flavus EST based amplicon microarray was constructed
currently J. Craig Venter Institute, JCVI). A total of 19,618 by the Food and Feed Safety Research Unit of the USDA/
A. flavus ESTs were generated, from which 7218 unique ARS/SRRC at TIGR. Using genomic DNA as template,

World Mycotoxin Journal 1 (1)


D. Bhatnagar et al.

the specific gene sequences were amplified using primers Genomics of crop plants infected by Aspergillus flavus
synthesised based on A. flavus unique EST sequences
assembled. A comprehensive whole genome A. flavus With the advent of modern advances in plant sciences
oligo microarray has also been constructed at TIGR by and genomic sequencing, it is encouraging to forecast
the Food and Feed Safety Research Unit. All of the 11,820 that many plant genome sequences will be made available
genes unique to A. flavus, the unique genes present in A. shortly. The importance of gene expression profiling by
oryzae but absent in A. flavus, and 10 genes cloned from genomic and proteomic analyses will play a major role
corn that show resistance to A. flavus infection, have been in the increasing power of molecular breeding that will
printed on this whole genome microarray. An Affymetrix help the breeders and biotechnologists to harness the
microarray funded with a grant from USDA/NRI awarded maximum potential of crop genomes to provide higher
to a consortium led by Prof. Gary A. Payne contains all of yield, novel value-added traits along with increased food
the A. flavus genes, A. oryzae unique genes, plus additional and feed safety. So far, the sequences of two small plant
genes of interest from corn, Fusarium species, mouse genomes, Arabidopsis and rice have been completed in
and human genomes. Additionally, a peanut/A. flavus 2000 and 2005, respectively (The Arabidopsis Genome
combined microarray, funded by the Crop Protection and Initiative, 2000; International Rice Genome Sequencing
Management Laboratory of the South Atlantic Area of the Project, 2005). Even with the currently available technology,
Agricultural Research Service (USDA/ARS/SAA), Tifton, whole genome sequencing of several important crops is
Georgia, is under construction at JCVI. This crop/fungus often a laborious and expensive process because of the large
combined array will contain oligonucleotides representing size of genomes (Table 1). As a result, several laboratories
over 10,000 peanut ESTs, and all of the annotated A. flavus have resorted to sequencing of large numbers of ESTs that
and A. oryzae unique genes (reviewed in Yu et al., 2007a). can deliver substantial amounts of genetic information
on protein-coding genes for comparative and functional
Expression profiling of genes involved in aflatoxin genomic studies. The latest available information indicates
formation using those microarrays, performed at USDA that among all characterised genomes to date, the maize
laboratories, the laboratories of North Carolina State genome is exceptionally dynamic and with over 2.3 million
University, TIGR and JCVI, identified hundreds of genes cDNA sequences from a variety of tissues and physiological
that are significantly up or down regulated under various regimens, the maize transcriptome is now the third best
nutritional and environmental conditions that either studied after mouse and man (Wessler, 2006; Rabinowicz
support or prevent aflatoxin formation (Cary et al., 2007; and Bennetzen, 2006; Messing and Dooner, 2006; Table 1).
Chang et al., 2007; Kim et al., 2006; O’Brian et al., 2003, Other crops such as cotton, peanut and tree crops are
2007; Price et al., 2005, 2006; Wilkinson et al., 2007a,b; lagging behind and it is unlikely that their genome will be
Yu et al., 2007b). Further studies using these microarray sequenced completely in the near future due to the large
resources for a genome-wide gene profiling and functional size of their genomes.
analysis in relation to aflatoxin formation are expected to
reveal the regulatory elements (global or specific) required
for aflatoxin production. Table 1. Comparative genome size of selected cultivated crops.
For comparison, genome sizes of Arabidopsis and human are
Comparative genomics also given.

Results from the whole genome sequencing project show Crop Approx. Ploidy EST1
that the genome of A. flavus (36.3 Mb) is larger than that genome level
of A. nidulans (30.1 Mb) or A. fumigatus (29.4 Mb) and size1 (Mbp)
thus capable of a more complex pattern of secondary
metabolites. The expanded genome of A. flavus over other Arabidopsis thaliana 128 2n=10 1,276,692
Aspergillus species suggests that A. flavus is adapted to Oryza japonica (rice) 420 2n=24 1,214,083
growing in complex environments. An analysis of the Gossypium hirsutum (cotton) 2,700 2n=4x=52 177,202
function of these extra genes may reveal those genes that Arachis hypogea (peanut) 2,800 2n=4x=40 40,627
make this fungus a successful saprophyte as well and a Zea mays (maize) 2,500 2n=20 1,159,264
pathogen of plants and animals. Interestingly, comparative Glycine max (soybean) 1,115 2n=2x=40 392,337
genomic studies show that A. flavus is highly similar to A. Hordeum vulgare (barley) 5,000 2n=6x=42 437,713
oryzae (37.6 Mb) with respect to genome size and number Triticum aestivum (wheat) 16,500 2n=6x=42 1,050,932
of genes for secondary metabolism. These results support human 2,800 2n=46 8,134,112
the conclusions of others that A. oryzae is not a separate other mammals 1,400-3,700
species, but rather a domesticated ecotype of A. flavus
(Payne et al., 2006). 1http://www.ncbi.nlm.nih.gov/dbEST/dbEST_summary.html; Accessed:

14 October 2007.

 World Mycotoxin Journal 1 (1)


 The ‘omics’ tools: genomics, proteomics, metabolomics and their potential for solving the aflatoxin contamination problem

Improved methods for gene and genomic sequencing, and be processed in the cell by alternative splicing, protein
analysis of gene expression, promise to allow rapid strides modification (glycosylation, phosphorylation) and protein
in the identification and understanding of gene function degradation. A prime example is that there are more than
and phenotype. Availability of genetic linkage maps and 500,000 proteins in human cells derived from about 25,000
quantitative trait loci (QTL) maps will also be useful in identified genes.
mapping traits and marker-assisted selection (MAS),
anchoring physical maps from large insert libraries and Current research in proteomics requires first that proteins
positional cloning of important genes, comparisons of be resolved, sometimes on a massive scale. Protein
synteny within and across species and ordered genome separation can be performed using two-dimensional gel
sequencing. Gene expression assays using macroarray electrophoresis, which usually separates proteins first by
(nylon-based) or microarray (glass slide-based) screening isoelectric point and then by molecular weight (Wilkins
methods are powerful in associating genes with proper et al., 1996; Pennington et al., 1997). This approach
phenotypes and to associate genes with functions or takes advantage of the increased reproducibility, map
specific physiological conditions due to biotic or abiotic resolution, reliability and accuracy that two-dimensional
stress factors. polyacrylamide gel electrophoresis (2-D PAGE) offers
over 1-dimensional gel electrophoresis (Gorg et al., 1988)
Identification of specific genes and critical genetic to detect post- and cotranslational modifications that
pathways is essential for conventional and transgenic cannot be predicted from a DNA sequence. Individual
improvement programs. Analysis of gene function in spots are cut out of the gel and cleaved into peptides with
native plant environment is possible using efficient genetic proteolytic enzymes. These peptides can then be identified
transformation technology and it has been practised by mass spectrometry, specifically matrix-assisted laser
routinely for cotton, peanut and corn (Rajasekaran 2004; desorption-ionisation time-of-flight (MALDI-TOF) mass
Ozias-Akins et al., 1993; Ishida et al., 2007). In conventional spectrometry.
breeding, MAS may not be preferable for traits controlled
by a large number of QTLs, because the individual gene’s Proteomics of Aspergillus flavus
contribution may be too small to measure since many genes
with small effects are involved (Morgante and Salamini, Very few reports of studies on the proteomics of A. flavus
2003). For a review of the power and limitations of MAS are available in the literature. The first extensive proteomic
for resistance to mycotoxin accumulation see review by study conducted to identify the secreted proteins from a
Robertson et al. (2005). filamentous fungus was reported by Medina et al. (2004).
In this study, a proteome analysis was used to differentiate
The new genomics resources that are becoming available and identify the extracellular rutin-induced and non-
for A. flavus as well as the crops infected by the fungus induced proteins secreted by A. flavus. In a continuation
will greatly aid our understanding of the ecology and study Medina et al. (2005) reported a total of 51 unique
metabolism of this fungus as well. A. flavus secreted proteins from three growth conditions.
Ten proteins were unique to rutin-, five to glucose- and
3. Proteomics one to potato dextrose-grown A. flavus. Sixteen secreted
proteins were common to all three media. Fourteen
Proteomics is the study of a complete set of proteins in a identifications were hypothetical proteins or proteins of
cell and their structures and functions in the physiological unknown functions.
pathways of cells. The term ‘proteomics’ is analogous to
genomics, the study of the complete set of genes. The word Identification of host resistance-associated proteins
‘proteome’ is derived from a combination of ‘protein’ and through proteomics
‘genome’. The proteome of an organism is the set of proteins
produced by it during its life, based on its genome, i.e. its Developing resistance to fungal infection in wounded and
set of genes (for details see Twyman, 2004; Westermeier intact maize kernels would accelerate solving the aflatoxin
and Naven, 2002; Liebler, 2002). problem (Payne, 1992). Studies of subpericarp (wounded
kernel) resistance in maize have identified corresponding
Proteomics is often seen as the next step in the study of resistance mechanisms. In a number of studies resistance
biological systems, after genomics (Wilkins et al., 1997). associated proteins (RAPs) were identified (Brown et al.,
It is much more complicated than genomics, mostly 2004) using proteomics. By comparing lines of maize
because an organism’s genome is rather constant, but a resistant and susceptible to aflatoxin accumulation, proteins
proteome changes from cell to cell based on its biochemical associated with resistance, as well as the genes encoding
interactions with the genome and environmental factors them, can be identified thereby facilitating marker-assisted
that the cell encounters. The complexity of proteins relative breeding and/or genetic engineering efforts. Endosperm
to nucleic acids is also due to the fact that proteins can and embryo proteins from several resistant and susceptible

World Mycotoxin Journal 1 (1)


D. Bhatnagar et al.

genotypes were compared and over a dozen constitutively seed and provides an easy entry site for fungi and insects.
expressed protein spots, either unique or 5-fold up- Possession of unique or increased levels of hydrophilic
regulated in resistant lines (Mp420 and Mp313E), were storage or stress-related proteins by resistant lines could
identified and analysed with electrospray ionisation tandem increase protein synthesis and host defence under stress
mass spectrometry (ESI-MS/MS) after in-gel digestion with conditions. Thus, commercially-useful, aflatoxin-resistant
trypsin (Chen et al., 2000, 2002). maize lines may include not only antifungal proteins but
also high-level expression of stress-related proteins.
Both constitutive and induced proteins are required for Further studies including physiological and biochemical
kernel resistance to aflatoxin production (Chen et al., characterisation, genetic mapping, plant transformation
2001). A major difference between resistant and susceptible using RAP genes, gene silencing through RNA interference
maize genotypes is the relatively high level of antifungal (RNAi) and marker-assisted breeding should clarify the
proteins constitutively expressed by the resistant lines. roles of stress-related RAPs in kernel resistance.
These constitutive proteins may delay fungal invasion, and
subsequent aflatoxin formation, until infection-induced The direct involvement of glyoxalase I, a purported stress-
antifungal proteins are synthesised. The identified proteins related aflatoxin resistance protein, has been evaluated
can be grouped into three categories based on their peptide (Chen et al., 2004). The substrate for glyoxalase I,
sequence homology: (1) storage proteins, e.g. globulins and methylglyoxal, is a potent cytotoxic compound produced
late embryogenesis abundant proteins, (2) stress-responsive from glycolysis and photosynthesis intermediates,
proteins, e.g. aldose reductase, a glyoxalase I protein and a glyceraldehyde-3-phosphate and dihydroxyacetone
16.9 kDa heat shock protein, and (3) antifungal proteins, phosphate. Methylglyoxal induces aflatoxin production
including the pathogenesis-related protein PR-10 (Chen through up-regulation of aflatoxin biosynthetic pathway
et al., 2006). transcripts, including the qflR regulatory gene. Thus,
glyoxalase I may directly affect resistance by removing the
The proteomes of near-isogenic maize lines from the methylglyoxal aflatoxin-inducing substrate.
same backcross in a collaborative project between the
International Institute for Tropical Agriculture (IITA) and 4. Metabolomics
the USDA differing significantly in aflatoxin accumulation
were analysed and proteins identified in all three of the Metabolites are small molecules that are either
predicted categories (Brown et al., 2003). RAPs are more intermediates or final products of metabolism. Metabolites
easily identified in these lines as the confounding effects directly involved in the normal growth, development, and
of differences in the genetic backgrounds of the lines are reproduction are primary metabolites, whereas those not
absent. involved in these processes such as aflatoxins are called
secondary metabolites. Secondary metabolites may have
Examples of some of the resistance-associated proteins, important ecological significance. Therefore, metabolomics
identified via proteomics of maize kernels, with can be defined as the ‘systematic study of the unique
demonstrated antifungal activities against A. flavus are chemical fingerprints that specific cellular processes leave
given in Table 2. Increased temperature and drought, behind’, i.e. the study of their small-molecule metabolite
which often occur together, are major factors associated profiles (Daviss, 2005; Preti, 2005). The metabolome
with aflatoxin contamination of maize kernels (Payne, represents the collection of all metabolites in a biological
1992, 1998). If drought stress is imposed during grain organism (such as metabolic intermediates, hormones and
filling, then dry matter accumulation is reduced in kernels other signalling molecules, and secondary metabolites),
(Payne, 1992, 1998) which often results in cracks in the which are the end products of its gene expression (Fiehn,

Table 2. Examples of resistance-associated proteins (RAP) against Aspergillus flavus identified through proteomics.

Protein Resistant genotype1 Putative function

Trypsin inhibitor (14 kDa) MI82; CI2; T115 Antifungal; anti-amylase


Glyoxalase I Mp420; Mp313E Stress-related; antitoxigenic
Pathogenesis related protein (PR-10) Mp420; Mp313E; GT-MAS:gk Antifungal; ribonucleolytic
Peroxiredoxin antioxidant (PER1) Mp420; Mp313E Peroxidase; stress-related
Zeamatin GT-MAS:gk Antifungal

1Maize genotypes in which an association with resistance has been identified; other resistant genotypes are involved as well.

 World Mycotoxin Journal 1 (1)


 The ‘omics’ tools: genomics, proteomics, metabolomics and their potential for solving the aflatoxin contamination problem

2001). Thus, while mRNA gene expression data and the two strains grown under conducive or non-conductive
proteomic analyses do not tell the whole story of what temperatures, 28 °C and 37 °C, respectively, revealed large
might be happening in a cell, metabolic profiling can give differences in the small molecule compositions of the two
an instantaneous snapshot of the physiology of that cell. strains. Although the mutant strain appeared to have some
The metabolome is also dynamic, and constantly changing unique components when compared to the wild type,
because it is a reflection of the metabolic reactions, where chromatograms of the mutant samples lacked many of
outputs from one enzymatic chemical reaction are inputs the prominent peaks present in the wild type (Glassbrook
to other chemical reactions (hypercycles). and Payne, 2005). Thus the sec- mutant appears to be
altered in several pathways of secondary metabolism, not
The chromatographic separation techniques developed just aflatoxin biosynthesis. These findings suggest that the
in the late 1960’s, which made the initial detection of mutation in the sec- strain may be in a global regulatory
metabolites possible, can be called the beginning of of secondary metabolism. Further experiments are in
metabolomics. However, modern day metabolomics began progress, using mass spectrometry and pulse/pulse-chase
in 1970 with the work of Arthur Robinson, although it was techniques, to identify specific compounds and pathways
not called metabolomics then. The name metabolomics impacted by the mutation. Additionally, this metabolic
was coined twenty years later in the 1990s when the first profile will be compared with a transcriptional profile
paper using the word metabolome was published (Oliver (using an EST microarray) to determine the changes in
et al., 1998). gene expression resulting from the sec- mutation.

Separation and detection of metabolites are two issues Research is underway to place the identified metabolites
that need to be addressed in obtaining metabolic profile into metabolic pathways and thus better understand the
(Dunn and Ellis, 2005). Detection of the analytes, following metabolic changes associated with the sec- mutant. This
separation by chromatographic or other methods can be type of pathway mapping provides a framework for sorting
achieved through the use of capillary electrophoresis (CE), large quantities of metabolic profiling data and formulating
high performance liquid chromatography (HPLC), gas testable hypotheses.
chromatography, especially when interfaced with mass
spectrometry (GC-MS) and nuclear magnetic resonance 5. Conclusion
(NMR).
One of the challenges of systems biology is the integration
Metabolomics can be an excellent tool for determining the of genomic, proteomic and metabolomic information
phenotype caused by a genetic manipulation, such as gene to give a more complete picture of living organisms.
deletion or insertion. Sometimes this can be a sufficient The results of genomic analysis are expected to provide
goal in itself, for instance, to detect any phenotypic changes important information for developing novel strategies to
in a genetically-manipulated fungus. More exciting is the control aflatoxin contamination by identifying targets for
prospect of predicting the function of unknown genes by inhibiting fungal growth or toxin production, as well as
comparison with the metabolic perturbations caused by developing ‘designer’ biological control agents. With the
deletion/insertion of known genes (Weckwerth, 2003). large volume of information being reported with respect
to proteomics of host and genomics and field ecology
Metabolomics of Aspergillus flavus of the fungus, novel strategies will also emerge based
on a clear understanding of the aflatoxin contamination
The sec- (secondary metabolite minus) mutant of A. process, especially at the molecular level. If the interaction
parasiticus SRRC 143 produces fewer conidia on complete between the plant and the fungus can be better understood
and minimum media and fails to produce aflatoxin or through the use of proteomics, genomics and natural
to convert labelled aflatoxin pathway precursors into product chemistry, particularly in how plant factors may
aflatoxin. Transcripts of the aflatoxin pathway genes are influence fungal processes contributing to virulence and
barely detectable and transcripts of the pathway regulatory aflatoxin contamination, this information could accelerate
gene aflR are reduced 5-10 fold compared to wild type the development of breeding through marker selection
(Kale et al., 2003). Although the sec- mutant has impaired and/or gene insertion technologies for enhancing host
sporulation, it grows and produces an amount of biomass plant resistance.
comparable to WT on a minimal medium, indicating that
its core metabolism is intact.

In an attempt to better characterise the changes in a sec-


mutant of A. flavus, metabolic profiling was conducted on
the wild type (WT) and mutant strains (Glassbrook et al.,
2006). An initial comparison of the metabolic profile of

World Mycotoxin Journal 1 (1)


D. Bhatnagar et al.

References Chen, Z.-Y., Brown, R.L., Damann, K.E. and Cleveland, T.E., 2004.
Identification of a maize kernel stress-related protein and its effect
Baldwin, D., Crane, V. and Rice, D., 1999. A comparison of gel-based, on aflatoxin accumulation. Phytopathology 94: 938-945.
nylon filter and microarray techniques to detect differential RNA Chen, Z.-Y., Brown, R.L., Rajasekaran, K., Damann, K.E. and Cleveland,
expression in plants. Current Opinion in Plant Biology 2: 96-103. T.E., 2006. Identification of a maize kernel pathogenesis-related
Bennett, J.W. and Arnold, J., 2001. Genomics for fungi. In: Howard, R.J. protein and evidence for its involvement in resistance to Aspergillus
and Gow, N.A.R. (Eds.) The Mycota VIII, Biology of the fungal cell. flavus infection and aflatoxin production. Phytopathology 96:
Springer-Verlag, Heidelberg, Berlin, Germany, pp. 267-297. 87‑95.
Bhatnagar, D., Yu, J. and Cleveland, T.E., 2002. Applying the genomic Daviss, B., 2005. Growing pains for metabolomics. The Scientist 19:
wrench – new tool for an old problem. Mycopathologia 155: 9. 25-28.
Bhatnagar, D., Ehrlich, K.C. and Cleveland, T.E., 2003. Molecular Desjardins, A.E. and Bhatnagar, D., 2003. Fungal genomics: an
genetic analysis and regulation of aflatoxin biosynthesis. Applied overview. Applied Mycology and Biotechnology 3: 1-13.
Microbiology and Biotechnology 61: 83-93. Donson, J., Fang, Y.W., Espiritu-Santo, G., Xing, W.M., Salazar,
Bohnert, D.J., Ayoubi, P., Borchert, C., Bressan, R.A. and Burnap, R.L., A., Miyamoto, S., Armendarez, V. and Volkmuth, W., 2002.
2001. A genomics approach towards salt stress tolerance. Plant Comprehensive gene expression analysis by transcript profiling.
Physiology and Biochemistry 39: 295-311. Plant Molecular Biology 48: 75-97.
Brown, R.L., Chen, Z.-Y., Menkir, A. and Cleveland, T.E., 2003. Using Drăghici, S., 2003. Data analysis tools for DNA microarrays. Chapman
biotechnology to enhance host resistance to aflatoxin contamination & Hall/CRC Press, Boca Raton, FL, USA, 512 pp.
of maize – a mini-review. African Journal of Biotechnology 2: Dunn, W.B. and Ellis, D.I., 2005. Metabolomics: current analytical
557‑562. platforms and methodologies. Trends in Analytical Chemistry 24:
Brown, R.L., Chen, Z.-Y. and Cleveland, T.E., 2004. Molecular biology 285-294.
for control of mycotoxigenic fungi. In: Arora, D., Bridge, P. and Eaton, D.L. and Gallagher, E.P., 1994. Mechanisms of aflatoxin
Bhatnagar, D. (Eds.) Fungal biotechnology in agricultural, food, carcinogenesis. Annual Review of Pharmacology and Toxicology
and environmental applications. Marcel Dekker, New York, USA, 34:135-172.
pp. 69-77. Ehrlich, K.C., Montalbano, B.G. and Cotty, P.J., 2003. Sequence
Cary, J.W., Linz, J.E. and Bhatnagar, D., 2000. Aflatoxins: biological comparison of aflR from different Aspergillus species provides
significance and regulation of biosynthesis. In: Cary, J.W., Linz, J.E. evidence for variability in regulation of aflatoxin production. Fungal
and Bhatnagar, D. (Eds.) Microbial foodborne diseases: mechanisms Genetics and Biology 38: 63-74.
of pathogenesis and toxin synthesis. Technomic Publishing, Fakhoury, A.M. and Payne, G.A,. 2003. Genomics of filamentous fungi:
Lancaster, PA, USA, pp. 317-361. a general review. In: Arora, D.L., Bridge, P.D. and Bhatnagar, D.
Cary, J.W., O’Brian, G.R., Nielsen, D.M., Nierman, W.C., Harris- (Eds.) Handbook of fungal biotechnology. Marcel Dekker Inc., New
Coward, P., Yu, J., Bhatnagar, D., Cleveland, T.E., Payne, G.A. and York, USA, pp. 55-68.
Calvo, A.M., 2007. Elucidation of veA-dependent genes associated Fiehn, O., 2001. Combining genomics, metabolome analysis, and
with aflatoxin and sclerotial production in Aspergillus flavus by biochemical modelling to understand metabolic networks.
functional genomics. Applied Microbiology and Biotechnology Computational and Functional Genomics 2: 155-168.
76: 1107-1118. Food and Agriculture Organization of the United Nations (FAO), 2004.
Chang, P.-K., Wilkinson, J.W., Horn, B.W., Yu, J., Bhatnagar, D. and Worldwide regulations for mycotoxins in food and feed in 2003.
Cleveland, T.E., 2007. Genes differentially expressed by Aspergillus Food and Nutrition Paper 81. Rome, Italy, 180 pp.
flavus strains after loss of aflatoxin production by serial transfers. Galagan, J.E., Calvo, S.E., Cuomo, C., Ma, L.J., Wortman, J.R., Batzoglou,
Applied Microbiology and Biotechnology 76: 977-984. S., Lee, S.I., Basturkmen, M., Spevak, C.C., Clutterbuck, J., Kapitonov,
Chen, Z.-Y., Brown, R.L., Damann, K.E. and Cleveland, T.E., 2000. V., Jurka, J., Scazzocchio, C., Farman, M., Butler, J., Purcell, S., Harris,
Proteomics analysis of kernel embryo and endosperm proteins S., Braus, G.H., Draht, O., Busch, S., D’Enfert, C., Bouchier, C.,
of corn genotypes resistant or susceptible to Aspergillus flavus Goldman, G.H., Bell-Pedersen, D., Griffiths-Jones, S., Doonan, J.H.,
infection. In: Robens, J. (Ed.) Proceedings of the USDA-ARS Yu, J., Vienken, K., Pain, A., Freitag, M., Selker, E.U., Archer, D.B.,
Aflatoxin Elimination Workshop (Yosemite, CA, USA, October Penalva, M.A., Oakley, B.R., Momany, M., Tanaka, T., Kumagai, T.,
2000). USDA-ARS, Beltsville, MD, USA, p. 88. Asai, K., Machida, M., Nierman,, W.C., Denning, D.W., Caddick,
Chen, Z.-Y., Brown, R.L., Cleveland, T.E., Damann, K.E. and Russin, M., Hynes, M., Paoletti,M., Fischer, R., Miller, B., Dyer, P., Sachs,
J.S., 2001. Comparison of constitutive and inducible maize M.S., Osmani, S.A. and Birren B.W., 2005. Sequencing of Aspergillus
kernel proteins of genotypes resistant or susceptible to aflatoxin nidulans and comparative analysis with A. fumigatus and A. oryzae.
production. Journal of Food Protection 64: 1785-1792. Nature 438: 1105-1115.
Chen, Z.-Y., Brown, R.L., Damann, K.E. and Cleveland, T.E., 2002. Glassbrook, N.J. and Payne, G.A., 2005. Metabolic profiling of
Identification of unique or elevated levels of kernel proteins in Aspergillus flavus during aflatoxin biosynthesis. In: Robens, J. (Ed.)
aflatoxin-resistant maize genotypes through proteome analysis. Proceedings of the USDA-ARS Aflatoxin Elimination Workshop
Phytopathology 92: 1084-1094. (Raleigh, NC, USA, October 2005). USDA-ARS, Beltsville, MD,
USA, p. 49.

10 World Mycotoxin Journal 1 (1)


 The ‘omics’ tools: genomics, proteomics, metabolomics and their potential for solving the aflatoxin contamination problem

Glassbrook, N.J., Kale, S.P., Bhatnagar, D., Bennett, J.W., Wilkinson, Morgante, M. and Salamini, F., 2003. From plant genomics to breeding
J.R., Yu, J., Cary, J.W., Cleveland, T.E. and Payne, G.A., 2006. The practice. Current Opinion in Biotechnology 14: 214-219.
sec- mutant of Aspergillus parasiticus has pronounced alterations Nierman, W.C., Pain, A., Anderson, M.J., Wortman, J.R., Kim, H.S.,
in its biochemical profile. 2006 Annual Meeting of the American Arroyo, J., Berriman, M., Abe, K., Archer, D.B., Bermejo, C., Bennett,
Society for Microbiology (Orlando, FL, USA, May 2006). Abstract J., Bowyer, P., Chen, D., Collins, M., Coulsen, R., Davies, R., Dyer, P.S.,
No. O-040. Farman, M., Fedorova, N., Fedorova, N., Feldblyum, T.V., Fischer, R.,
Gorg, A., Postel, W. and Gunther, S., 1988. The current state of two- Fosker, N., Fraser, A., Garcia, J.L., Garcia, M.J., Goble, A., Goldman,
dimensional electrophoresis with immobilized pH gradients. G.H., Gomi, K., Griffith-Jones, S., Gwilliam, R., Haas, B., Haas, H.,
Electrophoresis 9: 531-546. Harris, D., Horiuchi, H., Huang, J., Humphray, S., Jimenez, J., Keller,
Henry, S.H., Bosch, F.X. and Bowers J.C., 2002. Aflatoxin, hepatitis and N., Khouri, H., Kitamoto, K., Kobayashi, T., Konzack, S., Kulkarni, R.,
worldwide liver cancer risks. Experimental Medicine and Biology Kumagai, T., Lafon, A., Latge, J.P., Li ,W., Lord, A., Lu, C., Majoros,
504: 229-233 W.H., May, G.S., Miller, B.L., Mohamoud, Y., Molina, M., Monod,
Hieter, P. and Boguski M., 1997. Functional genomics: it’s all how you M., Mouyna, I., Mulligan, S., Murphy, L., O’Neil, S., Paulsen, I.,
read it. Science 278: 601-602. Penalva, M.A., Pertea, M., Price, C., Pritchard, B.L., Quail, M.A.,
International Rice Genome Sequencing Project, 2005.The map-based Rabbinowitsch, E., Rawlins, N., Rajandream, M.A., Reichard, U.,
sequence of the rice genome. Nature 436: 793–800. Renauld, H., Robson, G.D., Rodriguez de Cordoba, S., Rodriguez-
Ishida, Y., Hiei, Y. and Komari, T., 2007. Agrobacterium-mediated Pena, J.M., Ronning, C.M., Rutter, S., Salzberg, S.L., Sanchez, M.,
transformation of maize. Nature Protocols 2: 1614-1621. Sanchez-Ferrero, J.C., Saunders, D., Seeger, K., Squares, R., Squares,
Jelinek, C.F., Pohland, A.E. and Wood, G.E., 1989. Worldwide S., Takeuchi, M., Tekaia, F., Turner, G., Vazquez de Aldana, C.R.,
occurrence of mycotoxins in food and feeds – an update. Journal Weidman, J., White, O., Woodward, J., Yu, J.H., Fraser, C., Galagan,
of the Association of Official Analytical Chemists 72: 223-230. J.E., Asai, K., Machida, M., Hall, N., Barrell, B. and Denning, D.W.,
Kale, S.P., Cary, J.W., Baker, C., Walker, D., Bhatnagar, D. and Bennett, 2005. Genomic sequence of the pathogenic and allergenic filamentous
J.W., 2003. Genetic analysis of morphological variants of Aspergillus fungus Aspergillus fumigatus. Nature 438: 1151-1156.
parasiticus deficient in secondary metabolite production. O’Brian, G.R., Fakhoury, A.M. and Payne G.A., 2003. Identification
Mycological Research 107: 831-840. of genes differentially expressed during aflatoxin biosynthesis in
Kim, J.H., Campbell, B.C., Molyneux, R., Mahoney, N., Chan, K.L., Aspergillus flavus and Aspergillus parasiticus. Fungal Genetics and
Yu, J., Wilkinson, J.R., Cary, J., Bhatnagar, D. and Cleveland, T.E., Biology 39: 118-127.
2006. Gene targets for fungal and mycotoxin control. Mycotoxin O’Brian, G.R., Georgianna, D.R., Wilkinson, J.R., Yu, J., Abbas, H.K.,
Research 22: 3-8. Nierman, W.C., Bhatnagar, D., Cleveland, T.E. and Payne, G.A.,
Kuska, B., 1998. Beer, Bethesda, and biology: how ‘genomics’ came 2007. The effect of elevated temperature on gene transcription and
into being. Journal of the National Cancer Institute 90:93. aflatoxin biosynthesis. Mycologia 99: 232-239.
Liebler, D.C., 2002. Introduction to proteomics: tools for the new Ohlrogge, J. and Benning, C., 2000. Unraveling plant metabolism by
biology. Humana Press, Totowa, NJ, USA, 201 pp. EST analysis. Current Opinion in Plant Biology 3:224-228.
Machida, M., Asai, K., Sano, M., Tanaka, T., Kumagai, T., Terai, G., Oliver, S.G., Winson, M.K., Kell, D.B. and Baganz, F., 1998. Systematic
Kusumoto, K., Arima, T., Akita, O., Kashiwagi, Y., Abe, K., Gomi, K., functional analysis of the yeast genome. Trends in Biotechnology
Horiuchi, H., Kitamoto, K., Kobayashi, T., Takeuchi, M., Denning, 16: 373-378.
D.W., Galagan, J.E., Nierman, W.C., Yu, J., Archer, D.B., Bennett, Otim, M.O., Mukiibi-Muka, G., Christensen, H. and Bisgaard, M.,
J.W., Bhatnagar, D., Cleveland, T.E., Fedorova, N.D., Gotoh, O., 2005. Aflatoxicosis, infectious bursal disease and immune response
Horikawa, H., Hosoyama, A., Ichinomiya, M., Igarashi, R., Iwashita, to Newcastle disease vaccination in rural chickens. Avian Pathology
K., Juvvadi, P.R., Kato, M., Kato, Y., Kin, T., Kokubun, A., Maeda, 34: 319-323.
H., Maeyama, N., Maruyama, J., Nagasaki, H., Nakajima, T., Oda, Ozias-Akins, P., Schnall, J.A., Anderson, W.F., Singsit, C., Clemente,
K., Okada, K., Paulsen, I., Sakamoto, K., Sawano, T., Takahashi, M., T.E., Adang, M.J. and Weissinger, A.K., 1993. Regeneration of
Takase, K., Terabayashi, Y., Wortman, J.R., Yamada, O., Yamagata, Y., transgenic peanut plants from stably transformed embryogenic
Anazawa, H., Hata, Y., Koide, Y., Komori, T., Koyama, Y., Minetoki, callus. Plant Science 93: 185-194
T., Suharnan, S., Tanaka, A., Isono, K., Kuhara, S., Ogasawara, Payne, G.A., 1992. Aflatoxin in maize. CRC Critical Reviews in Plant
N. and Kikuchi H., 2005. Genome sequencing and analysis of Sciences 10: 423-440.
Aspergillus oryzae. Nature 438: 1157-1161. Payne, G.A., 1998. Process of contamination by aflatoxin-producing
Medina, M.L., Kiernan, U.A. and Francisco, W.A., 2004. Proteomic fungi and their impact on crops. In: Sinha, K.K. and Bhatnagar, D.
analysis of rutin-induced secreted proteins from Aspergillus flavus. (Eds.) Mycotoxins in agriculture and food safety. Marcel Dekker,
Fungal Genetics and Biology 41: 327-335. New York, USA, pp. 279-306.
Medina, M.L., Haynes, P.A., Breci, L. and Francisco, W.A., 2005. Payne, G.A. and Brown, M.P., 1998. Genetics and physiology of aflatoxin
Analysis of secreted proteins from Aspergillus flavus. Proteomics biosynthesis. Annual Review of Phytopathology 36: 329-362.
5: 3153-3161. Payne, G.A., Nierman, W.C., Wortman, J.R., Pritchard, B.L., Brown,
Messing, J. and Dooner, H.K., 2006. Organization and variability of the D., Dean, R.A., Bhatnagar, D., Cleveland, T.E., Machida, M. and
maize genome. Current Opinion in Plant Biology 9: 157-163. Yu, J., 2006. Whole genome comparison of Aspergillus flavus and
A. oryzae. Medical Mycology 44: S9-S11.

World Mycotoxin Journal 1 (1) 11


D. Bhatnagar et al.

Pennington, S.R., Wilkins, M.R., Hochstrasser, D.F. and Dunn, M.J., Weckwerth, W., 2003. Metabolomics in systems biology. Annual
1997. Proteome analysis: from protein characterization to biological Review of Plant Biology 54: 669-689.
function. Trends in Cell Biology 7: 168-173. Wessler, S.R., 2006. Genome studies and molecular genetics: Part
Preti, G., 2005. Metabolomics comes of age? The Scientist 19: 8. 2 Maize genomics. The maize community welcomes the maize
Price, M.S., Conners, S.B., Tachdjian, S., Kelly, R.M. and Payne, G.A., genome sequencing project. Current Opinion in Plant Biology 9:
2005. Aflatoxin conducive and non-conducive growth conditions 147-148.
reveal new gene associations with aflatoxin production. Fungal Westermeier, R. and Naven, T., 2002. Proteomics in practice: a
Genetics and Biology 42: 506-518. laboratory manual of proteome analysis. Wiley-VCH, Weinheim,
Price, M.S., Yu, J., Nierman, W.C., Kim, H.S., Pritchard, B., Jacobus, Germany, 310 pp.
C.A., Bhatnagar, D., Cleveland, T.E. and Payne, G.A., 2006. The Wilkins, M.R., Pasquali, C., Appel, R.D., Ou, K., Golaz, O., Sanchez, J.C.,
aflatoxin pathway regulator aflR induces gene transcription inside Yan, J.X., Gooley, A.A., Hughes, G., Humphrey-Smith, I., Williams,
and outside of the aflatoxin biosynthetic cluster. FEMS Microbiology K.L. and Hochstrasser, D.F., 1996. From proteins to proteomes: Large-
Letters 255: 275-279. scale protein identification by two-dimensional electrophoresis and
Rabinowicz, P.D. and Bennetzen, J.L., 2006. The maize genome as amino acid analysis. Biotechnology 14: 61-65.
a model for efficient sequence analysis of large plant genomes. Wilkins, M.R., Williams, K.L., Appel, R.D. and Hochstrasser, D.F.,
Current Opinion in Plant Biology 9: 149-156 1997. Proteome research: new frontiers in functional genomics.
Rajasekaran, K., 2004. Agrobacterium-mediated genetic transformation Springer Verlag, Berlin, Germany, 243 pp.
of cotton. In: Curtis, I.S. (Ed.) Transgenic crops of the world Wilkinson, J.R., Yu, J., Abbas, H.K., Scheffler, B.E., Kim, H.S., Nierman,
– essential protocols. Kluwer Academic Press, Dordrecht, the W.C., Bhatnagar, D. and Cleveland, T.E., 2007a. Aflatoxin formation
Netherlands, pp. 243-254. and gene expression in response to carbon source media shift in
Richard, J.L. and Payne, G.A., 2003. Mycotoxins: risks in plant, Aspergillus parasiticus. Food Additives and Contaminants 24:
animal, and human systems. Task Force Report No. 139. Council 1051-1060.
for Agricultural Science and Technology (CAST), Ames, IA, USA, Wilkinson, J.R., Yu, J., Bland, J.M., Nierman, W.C., Bhatnagar, D.
199 pp. and Cleveland, T.E., 2007b. Amino acid supplementation reveals
Robertson, L.A., Payne, G.A. and Holland, J.B., 2005. Marker-assisted differential regulation of aflatoxin biosynthesis in Aspergillus
breeding for host resistance to mycotoxin contamination. In: Abbas, flavus NRRL 3357 and Aspergillus parasiticus SRRC 143. Applied
H.K. (Ed.) Aflatoxin and food safety. CRC Press, Boca Raton, FL, Microbiology and Biotechnology 74: 1308-1319.
USA, pp. 423-435. Wu, F., 2004. Mycotoxin risk assessment for the purpose of setting
The Arabidopsis Genome Initiative, 2000. Analysis of the genome international regulatory standards. Environmental Science and
sequence of the flowering plant Arabidopsis thaliana. Nature 408: Technology 38: 4049-4055.
796-815. Yu, J.H. and Keller, N., 2005. Regulation of secondary metabolism in
Turner, P.C., Moore, S.E., Hall, A.J., Prentice, A.M. and Wild, C.P., filamentous fungi. Annual Review of Phytopathology 43: 437-458.
2003. Modification of immune function through exposure to dietary Yu, J., Chang, P.K., Ehrlich, K.C., Cary, J.W., Bhatnagar, D., Cleveland,
aflatoxin in Gambian children. Environmental Health Perspectives T.E., Payne, G.A., Linz, J.E., Woloshuk, C.P. and Bennett J.W., 2004a.
111: 217-220. Clustered pathway genes in aflatoxin biosynthesis. Applied and
Twyman, R.M., 2004. Principles of proteomics. BIOS Scientific Environmental Microbiology 70: 1253-1262.
Publishers, New York, USA, 350 pp. Yu, J., Whitelaw, C.A., Nierman, W.C., Bhatnagar, D., and Cleveland,
Valdivia, A.G., Martinez, A., Damian, F.J., Quezada, T., Ortiz, T.E., 2004b. Aspergillus flavus expressed sequence tags for
R., Maryinez, C., Llamas, J., Rodriguez, M.L., Yamamoto, L., identification of genes with putative roles in aflatoxin contamination
Jaramillo, F., Loarca-Pina, M.G. and Reyes, J.L., 2001. Efficacy of of crops. FEMS Microbiology Letters 237: 333-340.
N-acetylcysteine to reduce the effects of aflatoxin B1 intoxication Yu, J., Payne, G.A., Nierman, W.C., Machida, M., Bennett, J.W.,
in broiler chickens. Poultry Science 80: 727-734. Campbell, B.C., Robens, J.F., Bhatnagar, D., Dean, R.A. and
Van Egmond, H.P., Schothorst, R.C. and Jonker, M.A., 2007. Cleveland, T.E., 2007a. Aspergillus flavus genomics in controlling
Regulations relating to mycotoxins in food: perspectives in a global aflatoxin contamination of food and feed. Food Additives and
and European context. Analytical and Bioanalytical Chemistry 389: Contaminants (in press).
147-157. Yu, J., Ronning, C.M., Wilkinson, J.R., Campbell, B.C., Payne, G.A.,
Vardon, P., McLaughlin, C. and Nardinelli, C., 2003. Potential economic Bhatnagar, D., Cleveland, T.E., and Nierman, W.C., 2007b. Gene
costs of mycotoxins in the United States. In: Richard, J.L. and profiling for studying the mechanism of aflatoxin biosynthesis
Payne, G.A. (Eds.) Mycotoxins: risks in plant, animal, and human in Aspergillus flavus and A. parasiticus. Food Additives and
Systems. Task Force Report No. 139. Council for Agricultural Contaminants 24: 1035-1042.
science and Technology (CAST), Ames, IA, USA, pp. 136-142.

12 World Mycotoxin Journal 1 (1)

You might also like