JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

Embed Size (px)

Citation preview

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    1/13

    Review

    Advancements in molecular marker development and theirapplications in the management of biotic stresses in peanuts

    Gyan P. Mishra*, T. Radhakrishnan, Abhay Kumar, P.P. Thirumalaisamy, Narendra Kumar,Tejas C. Bosamia, Bhagwat Nawade, Jentilal R. Dobaria

    Directorate of Groundnut Research, Post Box No. 05, Junagadh 362 001, Gujarat, India

    a r t i c l e i n f o

     Article history:

    Received 31 March 2015

    Received in revised form

    17 July 2015

    Accepted 20 July 2015

    Available online xxx

    Keywords:

    Genomics

    Diseases and pests

    Marker assisted selection

    Linkage-mapping

    Transgenics

    a b s t r a c t

    Peanut is grown extensively in different parts of world, where various biotic and abiotic factors limit its

    productivity and quality. The major fungal biotic constraints to peanut production include rust ( Puccinia

    arachidis  Speg.), stem-rot (Sclerotium rolfsii), collar-rot ( Aspergillus niger  Van Teighem), aa-root ( Asper-

     gillus  avus), and late leaf spot (Phaeoisariopsis personata  Ber. and M A Curtis), while viral disease con-

    straints are peanut bud necrosis disease (PBND) caused by peanut bud necrosis virus (PBNV) and peanut

    stem necrosis disease (PSND) caused by tobacco streak virus (TSV). Since, only a few sources of resistance

    are available in cultivated peanut for some diseases, which has resulted in the limited success of con-

    ventional breeding programmes on disease resistance. Moreover, even marker assisted breeding in

    peanut is in the nascent stage and identication of some major quantitative trait loci (QTLs) for a few

    fungal disease resitance genes has only recently been reported. Substantial efforts are underway to

    develop PCR-based markers for the construction of high-density genetic linkage maps. This will enable

    the breeders to effectively pyramid various biotic stress resistance genes into different agronomically

    superior breeding populations, in a much shorter time. It is expected that the availability of various cost-

    effective genomic resources (SNPs, whole genome sequencing, KASPar, GBS etc.) and more effective

    mapping populations (NAM, MAGIC etc.) in the coming years will accelerate the mapping of complex

    traits in peanut. This review provides an overview of the current developments and future prospects of 

    molecular marker development and their applications for improving biotic-stress resistance in peanut

    crop.

    © 2015 Elsevier Ltd. All rights reserved.

    1. Introduction

    Peanuts ( Arachis hypogaea L.), also known as groundnuts, are

    grown in more than 120 countries with different agro-climatic

    zones between latitudes 40   S and 40   N on approximately

    21e24 M ha of land annually (Sarkar et al., 2014). It is cultivated

    predominantly by small farms under low input conditions andranks third and fourth as a source of protein and edible oil,

    respectively (Bhauso et al., 2014). Several biotic stresses are known

    to limit peanut productivity, and their severity and extent of dis-

    tribution vary with the cropping system, growing season, and re-

    gion. Among biotic stresses, several diseases including rust

    (Puccinia arachidis   Speg.), early leaf spot (ELS,   Cercospora arach-

    idicola), late leaf spot (LLS,  Phaeoisariopsis personata  Ber. and M A

    Curtis), and aatoxin contamination by   Aspergillus   avus   and Aspergillus parasiticus are global constraints against peanut pro-

    duction (Subrahmanyam et al., 1984; Waliyar, 1991). Rust, stem-rot

    (Sclerotium rolfsii), collar-rot ( Aspergillus niger   Van Teighem), and

    leaf spots are also quite serious and together may cause the loss of 

    50e60% of pod yield in India (Dwivedi et al., 2003; Subrahmanyam

    et al., 1985). In the peanut growing regions, high yielding, well-adapted cultivars contain multiple resistances to biotic stresses

    that can provide enhanced and sustainable peanut production

    (Dwivedi et al., 2003).

    The world's largest peanut germplasm collection with more

    than 15,000 accessions is housed at the International Crops

    Research Institute for the Semi-Arid Tropics (ICRISAT) in India

    (Gowda et al., 2013). These accessions have many differences in

    their vegetative, reproductive, physiological, and biochemical traits.

    The global  Arachis  gene pool possesses the source of resistance to

    many biotic stresses, including rust, ELS, LLS, Groundnut Rosette

    Disease [GRD, caused by a complex of three agents: groundnut*  Corresponding author.

    E-mail address: [email protected] (G.P. Mishra).

    Contents lists available at  ScienceDirect

    Crop Protection

    j o u r n a l h o m e p a g e :   w w w . e l s e v i e r . c om / l o c a t e / c r o p r o

    http://dx.doi.org/10.1016/j.cropro.2015.07.019

    0261-2194/©

     2015 Elsevier Ltd. All rights reserved.

    Crop Protection 77 (2015) 74e86

    mailto:[email protected]://www.sciencedirect.com/science/journal/02612194http://www.elsevier.com/locate/croprohttp://dx.doi.org/10.1016/j.cropro.2015.07.019http://dx.doi.org/10.1016/j.cropro.2015.07.019http://dx.doi.org/10.1016/j.cropro.2015.07.019http://dx.doi.org/10.1016/j.cropro.2015.07.019http://dx.doi.org/10.1016/j.cropro.2015.07.019http://dx.doi.org/10.1016/j.cropro.2015.07.019http://www.elsevier.com/locate/croprohttp://www.sciencedirect.com/science/journal/02612194http://crossmark.crossref.org/dialog/?doi=10.1016/j.cropro.2015.07.019&domain=pdfmailto:[email protected]

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    2/13

    rosette virus (GRV), its satellite RNA (sat RNA), and a groundnut

    rosette assistor virus (GRAV)], Peanut Bud Necrosis Virus (PBND), A.

     avus   induced aatoxin contamination, bacterial wilt (Ralstonia

    solanacearum), leafminer ( Aproaerema modicella),   Spodoptera,  jas-

    sids (Empoasca kerri Pruthi), thrips (Frankliniella schultzei  Trybom)

    and termites (Odontotermes sp.) (Rao et al., 2002; Basu and Singh,

    2004; Amin et al., 1985; Rao et al., 2014).

    Since the 1960s, interspecic hybridization has received much

    attention in peanuts because several wild  Arachis   species show a

    very high level of resistance to many biotic stresses, such as rust,

    ELS, LLS, and stem rot (Holbrook and Stalker, 2003; Singh et al.,

    1984). However, success in transferring the resistance to culti-

    vated peanuts has been limited mainly because of cross compati-

    bility barriers, linkage drag, and long periods required for

    developing stable tetraploid interspecic derivatives (Wynne et al.,

    1991; Singh et al.,1997). Moreover, the partial and polygenic nature

    of biotic stresses makes the identication of resistant and suscep-

    tible lines very tedious using conventional screening techniques

    (Leal-Bertioli et al., 2009). Because of the frequent occurrence of 

    multiple diseases, peanut yields are often signicantly lower than

    their potential (Holbrook and Stalker, 2003). In the future, cultivars

    with multiple disease and pest resistances will be needed, which

    appears to be a very dif cult endeavour for this crop species (Basuand Singh, 2004).

    Marker-assisted selection (MAS) offers great promise for

    improving the ef ciency of conventional plant breeding ( Janila

    et al., 2013), including the potential to pyramid resistance genes

    in peanuts (Mishra et al., 2009; Varshney et al., 2014; Pandey et al.,

    2012). For any molecular breeding program, assessment of genetic

    diversity and development of genetic linkage maps are two very

    important steps (Dwivedi et al., 2003). Abundant polymorphisms in

    wild  Arachis  species have been observed, but progress in the mo-

    lecular breeding of cultivated peanuts is greatly constrained due to

    the low level of detectable molecular genetic variation (Mondal

    et al., 2005; Herselman, 2003; Raina et al., 2001; He and Prakash,

    2001). Therefore, the use of more robust assays such as single

    nucleotide polymorphisms (SNPs), competitive allele-specic PCR (KASPar) and genotyping by sequencing (GBS) approaches are

    needed. However, cost-effective SNP genotyping platforms are not

    readily available for tetraploid peanuts, but a large number of 

    robust markers such as SSRs and SNPs (including KASPar) would be

    valuable. SSRs are still considered the marker of choice in peanuts

    (Pandey et al., 2012), and a wide range of genotypes have been used

    for mapping (Table 1) of many important biotic and abiotic traits

    using SSR markers (Table 2).

    Despite being an important oilseed crop, very limited work in

    the area of molecular genetics and breeding of peanuts has been

    performed (Dwivedi et al., 2002; Raina et al., 2001). However, over

    the last decade, signicant developments have been made in the

    use of various molecular approaches for biotic stress management

    in peanuts, and new efforts such as functional genomics are likelyto play key roles in the future (Wang et al., 2011; Varshney et al.,

    2014; Gajjar et al., 2014). Recently,   Kanyika et al. (2015)   has

    identied 376 polymorphic SSR markers in 16 African groundnut

    cultivars with a wide range of disease resistance. These identied

    markers can be used to improve the ef ciency of introgression of 

    resistance to multiple important biotic constraints into farmer-

    preferred varieties of Sub-Saharan Africa. In this review, we made

    an attempt to capture the recent updates in molecular marker

    development and their applications in the management of various

    biotic stresses in peanut.

    2. Markers associated with rust and LLS resistance gene(s)

    Rust and leaf spots areeconomically very important foliarfungal

    diseases of peanuts that often occur together and not only reduce

    the yield but also adversely affect the fodder and seed quality

    (Subrahmanyam et al., 1985; Waliyar, 1991). Despite the economic

    importance of rust and LLS, very limited work has been carried out

    on hostefungus interaction, fungal genetic diversity, and physio-

    logical specialization (Mondal and Badigannavar, 2015). Several

    studies have emphasized the application of different types of mo-

    lecular markers, construction of peanut linkage maps, or tagging of 

    important agronomic traits, such as disease resistance (Wang et al.,

    2011; Gajjar et al., 2014). Recently, many DNA markers have been

    found to be putatively linked to rust and LLS resistance genes(Mondalet al., 2012a; Khedikar et al., 2010; Shoba et al., 2012; Sujay

    et al., 2012) (Table 2), a few of which have been validated and used

    in the breeding programme (Sujay et al., 2012; Gajjar et al., 2014;

    Varshney et al., 2014). Location of markers on the various linkage

    groups in Table 2, is derived after doing intensive meta-analysis of 

    all the published literature, including the most comprehensive and

    consensus linkage maps available in peanut.

    Validation of other linked markers will accelerate the process of 

    introgression of disease resistance into preferred peanut genotypes

    (Sujay et al., 2012; Gajjar et al., 2014). Near isogenic lines (NILs)

    developed for rust resistance were thoroughly screened with both

    foreground and background molecular markers (Yeri et al., 2014).

    For the identication of LLS resistance, Luo et al. (2005b) identied

    genes in the resistant genotype that were more highly expressedthan in the susceptible genotype (in response to  Cercosporidium

     personatum infection) by microarray analysis and validated them by

    real-time PCR. In a recombinant inbred line (RIL) population (VG

    9514    TAG 24), two transposable element (TE) markers, TE 360

    and TE 498, were found to be associated with the rust resistance

    gene. These two markers need further validation before they could

    be effectively applied for MAS of rust resistance in different back-

    grounds (Mondal et al., 2013).

    3. Soil-borne fungal diseases and associated markers

    (Collar rot, Stem rot, Aspergillus spp., Bacterial wilt and Scle-

    rotinia blight)

    Among soil-borne diseases, collar rot ( A. niger ) and stem-rot (S.rolfsii) are very important (Farr et al., 1989; Kolte, 1984). The search

    for peanut cultivars resistant to S. rolfsii originates all the way back

     Table 1

    List of a few genotypes, used for mapping of various resistance gene(s) (Dwivedi et al., 2003; Shoba et al., 2012;

    Sujay et al., 2012).

    Traits Genotypes

    Early leaf spot ICG 405, ICG 1705, ICG 6284, TMV 2

    Late leaf spot GPBD 4, ICGV 99001, ICGV 99004, COG 0437, TAG 24, TMV 2

    Rust GPBD 4, ICGV 99003, ICGV 99005, TG 26, TMV 2

    Rosette disease ICG 6323, ICG 6466, ICG 11044, JL 24

    Bacterial wilt ICG 7893, ICG 15222, and Chico

    Aatoxin production U 4-7-5, 55-437, J 11

    G.P. Mishra et al. / Crop Protection 77 (2015) 74e86    75

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    3/13

    to 1918 (McClintock, 1918), but a high degree of resistance is yet to

    be found. Recently, Thirumalaisamy et al. (2014) has reported some

    sources for stem rot resistance in peanut, but the genetics of 

    resistance and markers linked with the resistance gene(s) are still

    lacking. Thus, there is an urgent need to   nd molecular markers

    linked with the stem rot resistance QTLs, for its use in marker-

    assisted breeding programme.Very few published reports pertaining to the molecular char-

    acterization of   S. rolfsii   are available. Variations in the internal

    transcribed spacer (ITS) regions have revealed 12 sub-specic

    groupings, some of which correlated with their mycelial compati-

    bility groups (MCGs) (Harlton et al., 1995).  Punja and Sun (1997)

    detected 68 MCGs while comparing 128 isolates of   S. rolfsii  from

    36 host species collected from 23 geographic regions.  Cilliers et al.

    (2000) found Amplied Fragment Length Polymorphism (AFLP) as

    a suitable technique for the assessment of genetic variability be-

    tween isolates and MCGs of  S. rolfsii. Genetic variability among the

    virulent isolates of S. rolfsii was studied using molecular techniques,

    such as RAPD, ITS-PCR and RFLP (Prasad et al., 2010). Several mo-

    lecular markers associated with pod rot resistance and suscepti-

    bility in mutants and Giza5 peanut genotype were reported by

    Azzam et al. (2007) using RAPD markers, although these markers

    still need to be validated.

     A.  avus and  A. parasiticus  infect peanut seeds and produce af-

    latoxins, which are harmful to both domestic animals and humans

    (Waliyar et al., 2015; Singh et al., 2015a,b; Liu et al., 2014). The

    progress in peanut breeding for resistance to aatoxin is slow due

    to the lack of a cost-effective method for resistance identication in

    breeding materials or segregating progeny (Lei et al., 2006). Barros

    et al. (2007) and Singh et al. (2015a,b) analysed Aspergillus  isolates

    collected from peanut   elds in Argentina and India using AFLP,

    where different levels of polymorphism in different groups of iso-

    lates were recorded. Guo et al. (2008) has identified up-regulation

    of 8 resistance-related genes for   Aspergillus   infection.   Lei et al.

    (2006)   reported an AFLP (E45/M53-440) converted SCAR marker

    (AFs-412) to be closely linked to resistance of   A.  avus  infection.

    Screening of isolates using gene-specic PCR was found to be quite

    effective for the identication of gene defects in   A.  avus, which

    could be used as bio-control agents in peanut growing areas (Dodia

    et al., 2014). Moreover, to date, the genetic basis of   S. rolfsi   and

     A.  avus stress resistance traits in peanuts is not fully understood.

    Therefore, extensive efforts are required to 

    rst identify the resis-tance sources and then to   nd the markers associated with the

    resistance genes/QTLs for their future use in marker assisted

    breeding (Mishra et al., 2014).

    Bacterial wilt (R. solanacearum) is an important constraint to

    peanut production in several Asian and African countries, and

    planting of bacterial-wilt resistant cultivars is the most feasible

    method fordisease control. Genetic diversityanalysis using SSR and

    AFLP markers among selected peanut genotypes identied certain

    genotypes and primer combinations for developing mapping

    populations and breeding for high yield, resistant cultivars ( Jiang

    et al., 2007a). In an attempt to understand the molecular mecha-

    nism of bacterial wilt resistance, 25 differentially expressed

    candidate genes were identied by studying the differences in gene

    expression between inoculated and control seeds. Conrmation of 

    the functions of these genes needs to be validated through trans-

    genic studies (Ding et al., 2012). For Sclerotinia blight (Sclerotinia

    minor ) resistance,  Kelly et al. (2010)  characterized 96 US peanut

    mini-core collections using a resistance-associated SSR marker, and

    39 accessions as new potential sources of resistance were

    identified.

    4. Molecular studies on resistance to viral diseases

    (Groundnut rosette disease, PBND and PSND)

    Groundnut rosette disease is the most destructive viral disease

    of peanuts causing serious yield losses. Single recessive gene con-

    trol for resistance to the aphid vector ( Aphis craccivora) was

    observed in the breeding line ICG 12991, which was mapped3.9 cM

     Table 2

    SSR markers and its linkage group known to be associated to the rust and/or LLS resistance gene(s).

    S. No. Primers Linkage group Cross/genotypes Resistance Reference

    1 seq3A01238a a07 ICGV 99003 TMV 2 Rust   Varma et al., 2005

    2 seq5D05274   b07and a07 TMV 2 COG 0437 (F2)

    and ICGV 99005 TMV 2

    Rust and LLS   Shoba et al., 2012; Varma et al., 2005;

    Shirasawa et al., 2013

    3 seq16F01271   b03 ICGV 99005 TMV 2 Rust   Varma et al., 2005

    4 seq17F06152   b04 ICGV 99005 TMV 2 and 22 genotypes Rust and LLS   Varma et al., 2005; Mace et al., 2006;

    Shirasawa et al., 20135 seq13A07265   b01 ICGV 99005 TMV 2 and 22 genotypes Rust and LLS   Varma et al., 2005; Mace et al., 2006;

    Shirasawa et al., 2013

    6 seq2F05280   b02 22 genotypes Rust and LLS   Mace et al., 2006; Shirasawa et al., 2013

    7 seq8E12200   a01 22 genotypes Rust resistance   Mace et al., 2006

    8 seq16C06263   b03 22 genotypes Rust   Mace et al., 2006

    9 seq13A10250   b04 22 genotypes Rust   Mace et al., 2006

    10 seq2B10290   b03 22 genotypes LLS   Mace et al., 2006

    11 IPAHM103160   a03 and b03 TAG 24 xGPBD 4 Rust   Khedikar et al., 2010

    12 PM384100   e   TMV 2 COG 0437 (F2) LLS   Shoba et al., 2012

    13 PM137150   b06 TMV 2 COG 0437 (F2) LLS   Shoba et al., 2012; Shirasawa et al., 2013

    14 PM03168   a03 and b03 TMV 2 COG 0437 (F2) LLS   Shoba et al., 2012; Shirasawa et al., 2013

    15 PMc588183   e   TMV 2 COG 0437 (F2) LLS   Shoba et al., 2012

    16 PM375102   a04 TMV 2 COG 0437 (F2) LLS   Shoba et al., 2012

    17 seq8D09190   b10 and a09 TAG 24 GPBD 4 LLS   Sujay et al., 2012; Shirasawa et al., 2013

    18 GM1536410   b03 TG 26 xGPBD 4 Rust   Sujay et al., 2012

    19 GM2301137   b03 TG 26 xGPBD 4 Rust   Sujay et al., 2012

    20 GM2079418

      b03 TG 26 xGPBD 4 Rust   Sujay et al., 2012

    21 PM50110   b05 20 genotypes Rust   Mondal and Badigannavar, 2010

    22 PM35124   a06 and b04 20 genotypes Rust and LLS   Mondal and Badigannavar, 2010;

    Shirasawa et al., 2013

    23 seq4A05 and

    gi56931710

    a03 RILs from VG 9514 TAG 24 Rust   Mondal et al., 2012a

    a Subscript numerical values are for linked band size.

    G.P. Mishra et al. / Crop Protection 77 (2015) 74e86 76

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    4/13

    from an AFLP marker on LG 01 (Herselman et al., 2004). Peanut bud

    necrosis disease (PBND) caused by peanut bud necrosis virus

    (PBNV) and vectored by Thrips (Vijayalakshmi et al., 1995) i s a

    major viral disease of peanuts. In 2000, a new threat to peanuts

    emerged in the form of a viral disease named peanut stem necrosis

    disease (PSND) caused by the peanut stem necrosis virus (PSNV),

    which results in lethal necrosis (Rao et al., 2000). Tobacco streak

    virus (TSV) was found to be associated with the disease (Reddy

    et al., 2002). Despite several years of effort, a conrmed source of 

    genetic resistance/tolerance to TSV has not been identied in the

    gene pool of cultivated peanuts. However, genetically engineered

    resistance has been actively investigated in recent years as an

    alternative to cope with this type of situation (Mehta et al., 2013).

    Kamdar et al. (2014) studied 115 resistant and one susceptible line

    to PBND using SSR, but no clear differentiation was observed.

    Similarly, SSR-based diversity analysis among 15 peanut genotypes

    (resistant and susceptible to PBND) grouped together based on

    their origin rather than PBND resistance (Srinivasaraghavan et al.,

    2012). In the future, the availability of more cost effective

    genomic resources, such as SNPs and whole genome sequencing,

    will hasten complex trait mapping efforts (especially for viral dis-

    eases) in this crop (Pandey et al., 2012).

    5. Molecular studies on nematode resistance

    In many peanut production areas across the world, the peanut

    root-knot nematode causes signicant economic losses (Holbrook

    and Stalker, 2003). Many sources of resistance were identied in

    the germplasm, and lot of molecular work has been performed to

    nd the linked gene(s)/QTLs (Mishra et al., 2009). For the root-knot

    nematode (Meloidogyne arenaria), two dominant genes   viz .   Mae

    (restricted egg number) and  Mag  (restricted galling) conditioning

    resistance were identied, and a RAPD marker (Z3/265) was found

    to be linked to these genes (Garcia et al., 1996).

    Different types of DNA markers associated with root-knot

    nematode resistance were also identied (Burow et al., 1996;

    Garcia et al., 1996; Choi et al., 1999; Church et al., 2000; Wanget al., 2008a) and are currently being used for the development of 

    resistant peanut cultivars. A DNA marker with 6% recombination

    frequency to the resistance gene has been developed to screen the

    segregating populations for nematode resistance (Chu et al., 2007).

    The RFLP marker R2430E was found to link to a locus for resistance

    of the peanut root-knot nematode M. arenaria (Neal) Chitwood race

    1 (Pípolo et al., 2014). Genes controlling the resistance to nema-

    todes were introgressed through interspecic hybridization and

    resulted in the release of the   rst nematode-resistant peanut

    cultivar COAN (Simpson and Starr, 2001). Other works on the

    development of nematode-resistant peanut cultivars are presented

    in other sections of this review.

    6. Single nucleotide polymorphism (SNP) technologies for biotic stresses

    Recent developments in SNP technologies for peanuts have

    indicated that, in the near future, additional options may be

    available for the rapid identication of large numbers of poly-

    morphic markers in peanuts (Kanazin et al., 2002; Khera et al.,

    2013). A single base pair extension (SBE) was found to be an ef -

    cient method for high-throughput SNP mapping in peanuts, and

    ve candidate genes for resistance were identied on the genetic

    map (Alveset al., 2008). Recently, Khera et al.(2013) useda set of96

    informative SNPs to develop competitive allele-specic PCR (KAS-

    Par) assays in peanuts, designated as GKAMs (Groundnut KASPar

    Assay Markers), through which 90 GKAMs were validated for

    different biotic stresses. A comprehensive list of the various

    markers associated with different diseases and pests is given in

    Table 3.

    7. Advancements in linkage mapping 

    Peanut is a self-pollinated allotetraploid ( 2n  ¼ 4x  ¼ 40, AABB)

    crop having ten basic chromosomes, harboring about 2813 Mb of 

    DNA content (Arumuganatham, 1991). Peanut genome is nearly 20

    times larger than that of   Arabidopsis thaliana   and is somewhat

    similar to  Gossypium hirsutum  and  Zea mays. Variation in genome

    size among accessions of  A. hypogaea (2n  ¼  4x  ¼ 40) and  Arachis

    duranensis   (2n   ¼   2x   ¼   20) (Singh et al., 1996) and between

     A. hypogaea and   A. monticola  (Temsch and Greilhuber, 2000) has

    also been reported. The   rst RFLP-based genetic linkage map of 

    peanuts was constructed using an F2   population (Halward et al.,

    1993). A list of a few classical and the most recent genetic linkage

    maps, including consensus maps, are given in Table 4.

    Compared to that of many other legume crops, the development

    of molecular markers in cultivated peanuts has progressed at a

    relatively slow pace. Although many markers have been developed

    in both wild and cultivated peanuts, the genetic linkage map is not

    yet saturated. There is a need to saturate the linkage mapto provide

    suf cient markers for marker-assisted breeding (Dwivedi et al.,2003). Constant increases in peanut marker identities and linkage

    groups together with the availability of genome sequence data in

    the recent past have opened up the possibility of integrating new

    markers to some LGs and identifying closely linked markers

    (Mondal et al., 2012a). Efforts are currently underway for applying

    genomics to peanut biotic stress resistance breeding through

    mapping and MAS for different biotic stresses (Pandey et al., 2012).

    8. QTL analysis for biotic stress resistance

    In peanuts, many markers were recently found to be associated

    with QTLs for various biotic stresses, namely, rust and LLS (Sujay

    et al., 2012; Mondal and Badigannavar, 2010),   Cylindrocladium

    black-rot and ELS (Stalker and Mozingo, 2001), nematodes (Chuet al., 2007; Nagy et al., 2010), tomato spotted wilt virus (TSWV)

    (Qin et al., 2012), aphid vector of GRD (Herselman et al., 2004). The

    majority of the identied QTLs are not major and account for less

    than 10% of the phenotypic variation explained (PVE). Major QTLs

    identied for rust and LLS in the germplasm source (Khedikar et al.,

    2010; Sujay et al., 2012) and for nematode resistance (Nagy et al.,

    2010; Simpson, 2001) were of a wild species origin.

    In peanuts, candidate genome regions controlling disease

    resistance were identied by placing the 34 sequence-conrmed

    candidate disease resistance genes and 05 QTLs against LLS on

    the genetic map of the A-genome of  Arachis. Upon grouping, these

    genes and QTLs were found to be present on the upper and lower

    regions of LG 04 and 02, respectively, indicating the prominence of 

    these regions for imparting disease resistance (Leal-Bertioli et al.,2009). QTL analysis in a   ‘Tifrunner     GT-C20’-derived mapping

    population has identied 54 QTLs in the F2  map, including 02 for

    thrips, 15 for TSWV, and 37 for LS. However, in the F5 map, 23 QTLs

    could be identied, including 01 for thrips, 09 for TSWV, and 13 for

    LS. This is the rst QTL study reporting novel QTLs for thrips, TSWV,

    and LS, which needs renement in the future (Wang et al., 2013,

    2014). Using RIL population (VG 9514   TAG 24), 13 main and 31

    epistatic QTLs for total developmental period (TDP) were detected

    for bruchid resistance. Two years screening identied two common

    main QTLs, qTDP-b08 for TDP (57e82% PVE) and qAE2010/11-a02

    for adult emergence (13e21% PVE) (Mondal et al., 2014).

    Shoba et al. (2013) reported Ah 4-26 and PM 384 as the closest

    markers for QTLs of 100 kernel weight and LLS severity, respec-

    tively. Because PMc 588 and Ah 4-26 are the 

    anking markers for

    G.P. Mishra et al. / Crop Protection 77 (2015) 74e86    77

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    5/13

    PM 384, they can therefore be used for marker-assisted breeding of 

    LLS resistance. A genetic map derived from a SunOleic97R NC94022 cross (Qin et al., 2012) was developed, and multiple

    phenotyping data have identied a total of 155 QTLs, of which, 01

    and 03 major QTLs for TSWV and LLS resistance, respectively, were

    identied (Guo et al., 2013).

    A QTL region with 82.62% phenotypic variation for rust resis-

    tance (Sujay et al., 2012) was validated and introgressed in some

    susceptible varieties using a marker-assisted backcrossing (MABC)

    approach by employing four markers, namely IPAHM103, GM2079,

    GM1536 and GM2301 (Varshney et al., 2014). It is expected that

    multi-location testing of the promising introgressed lines will

    result in the identication of entries for its possible release as va-

    rieties with enhanced disease resistance. This result emphasized

    the utility of using markers for QTL selection through MAS for

    improving the rust resistance of any elite varieties (Varshney et al.,2014).

    To date, neither background nor genome-wide selection has

    been performed in peanuts, and this analysis must await the

    development of high-throughput, economical assays for a large

    number of markers (Holbrook et al., 2011). Details of some QTLs

    that have been identied to be associated with biotic stress-related

    traits in peanuts are given in Table 5. Identication of major QTLs

    for various biotic stress resistances in peanuts will enable ef cient

    MABC for the development of resistant cultivars. Recently, 39

    marker trait associations (MTAs) for   A.  avus, ELS, LLS, and GRD

    were identied using genome wide association studies (GWAS). Of 

    these MTAs, 01 was for   Aspergillus   (24.69% PV), 31 for GRD

    (10.25e39.29% PV), 06 for ELS (9.18e10.99% PV), and 01 for LLS

    (18.10% PV). It is expected that upon validation these MTAs may be

    deployed for marker-assisted improvement of peanut genotypes

    (Pandey et al., 2014).

    9. Biotic stresses and gene expression studies

    (Transcriptome analysis, microRNAs, epigenetic regulation,

    microarrays)

    In cultivated peanuts, which do not have whole genome

    sequence information available yet, transcriptome data are an

    alternative source of information, and ESTs can be used to identify

    candidate genes (Pandey et al., 2012). During recent years, a wealth

    of genomic data has been generated in peanut by high throughput

    transcriptome sequencing (Chopra et al., 2014, 2015). However, the

    available transcriptome sequences are not complete; many have

    low N50 values, ranging from500 to 750 bp (Guimar~aes et al., 2012;

    Chopra et al., 2014). On the basis of gene annotations, Bosamia et al.(2015)   has identied 2784 SSR containing sequences, of which,

    2027 (72.81%) were annotated and assigned in 4124 gene ontology

    terms and 31.91% sequences were found to be associated with

    response to stimulus encompassing biotic as well as abiotic

    stresses.

    Transcriptome analysis of cDNA collections from   Arachis sten-

    osperma   infected with   C. personatum  revealed a number of tran-

    scription factor families and defence-related genes. Additionally,

    the expression of   ve  A. stenosperma   resistance gene analogues

    (RGAs) and four retrotransposon (FIDEL-related) sequences were

    also analysed by qRT-PCR, which was used to design EST-SSRs of 

    which 214 were shown to be polymorphic (Guimar~aes et al., 2012).

    Bertioli et al. (2003) generated 78 RGAs based on the nucleotide-

    binding site (NBS) regions from   A. hypogaea   and other wild

     Table 3

    List of different markers associated with various biotic stresses.

    Markers Disease/Causal organism Reference

    RAPD Nematode   Garcia et al., 1996; Burow et al., 1996

    RFLP Nematode   Choi et al., 1999; Church et al., 2000

    AFLP Bacterial wilt   Jiang et al., 2003; Ren et al., 2008

    AFLP Rosette disease   Herselman et al., 2004

    AFLP   A. avus   Lei et al., 2005

    SCAR    A. avus   Lei et al., 2006RAPD Pod-rot   Azzam et al., 2007

    AS-PCR a Nematode   Chu et al., 2007

    RAPD Rust   Mondal et al., 2007

    AFLP and SSR Bacterial wilt   Jiang et al., 2007a; Jiang et al., 2007b

    AFLP Rust   Hou et al., 2007

    AFLP LLS   Xia et al., 2007

    SSR Sclerotinia blight   Kelly et al., 2010

    RAPD and ISSR Rust and LLS   Mondal et al., 2008

    SSR Nematode   Wang et al., 2008a

    ISSR Rust and LLS   Mondal et al., 2009

    SSR    A. avus   Hong et al., 2009

    Microarray   A. avus   Guo et al., 2011

    EST-SSR Rust   Mondal et al., 2012b

    SNP Rust and Leaf spots   Khera et al., 2013

    TEa Rust   Mondal et al., 2013

    a TE: Transposable Element; AS-PCR: Allele Specic-PCR.

     Table 4

    List some classical and latest genetic linkage maps.

    Populations Marker(s)/Loci Map distance (cM) References

    F2   RFLP 1063.0   Halward et al., 1993

    Back Cross RFLP 2210.0   Burow et al., 2001

    Arachis duranensis 1724 marker loci 1081.3   Nagy et al., 2012

    02 RILs 324 marker loci 1352.1   Qin et al., 2012

    02 RILs 225 SSR 1152.9   Sujay et al., 2012

    10 RILs, 01 BC 897 marker loci 3863.6   Gautami et al., 2012

    03 RILs 3693 marker loci 2651.0   Shirasawa et al., 2013

    G.P. Mishra et al. / Crop Protection 77 (2015) 74e86 78

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    6/13

    relatives. A collection of peanut nucleotide binding site leucine-rich

    repeat (NBSeLRR) resistance gene candidates (RGCs) were identi-

    ed by mining the GenBank databases (Radwan et al., 2010).Further, Yuksel et al. (2005)  also isolated 234 RGAs based on the

    primer sequence information from NBS-LRR and LRR-‘Toll’-like

    motif (LRR-TM) classes. On the similar note,   Proite et al. (2007)

    identied 35 putative non-redundant RGAs and 26 pathogenesis

    related ESTs from an accession of   A. stenosperma, resistant to

    different foliar diseases.

    At present, a total of 281,754 ESTs are available for  Arachis spp.

    on the NCBI website (www.ncbi.nlm.nih.gov, accessed January 24,

    2015), of which the contribution of each species is as follows:  A.

    hypogaea   (205,442),   A. duranensis   (35,291),   Aipaensis ipaensis

    (32,787), A. stenosperma  (6264),  Alberta magna (750),  A. hypogaea

    subsp. fastigiata (745), A. appressipila (400) and A. diogoi (75). Many

    of these have been generated for the identication of biotic stress

    resistance genes (e.g., 21,777 ESTs were identi

    ed from developingseeds against Aspergillus infection (Guo et al., 2008), and 8000 ESTs

    from the root tissues of  A. stenosperma  resistant to   M. arenaria

    (Proite et al., 2007). Guo et al. (2009) identied 17,376 ESTs from the

    leaves of TSWV and leaf spot resistant and susceptible cultivars, of 

    which 5717 had unknown functions. In response to   A.    avus

    infection under drought stress, 29 protein spots showing differen-

    tial expression between resistant and susceptible cultivars were

    identied and further validated using RT-PCR analysis (Wang et al.,

    2010). In addition, some defense-related transcripts, such as puta-

    tive oxalate oxidase (EU024476) and NBS-LRR domains, were also

    identied.

    Large numbers of available ESTs in the public databases were

    used for EST-SSR marker development. From 16424 unigenes, using

    MISA (MIcroSAtellite identication tool) search, a total of 2456

    novel EST-SSR primer pairs were designed; of which 366 unigenes,

    having relevance to various stresses and other functions were PCR 

    validated using a set of 11 diverse peanut genotypes (both resistant

    and susceptible to various diseases). Of these, 340 primer pairs

    yielded clear and scorable PCR products and 39 primer pairs

    exhibited polymorphisms which are of potential use for the resis-

    tance breeding programme (Bosamia et al., 2015). Out of 411 sor-

    ghum EST-SSR (SbEST-SSR) markers tested in peanuts, 39% could be

    successfully amplied, of which 14% showed polymorphism among

    resistant and susceptible cultivars for rust and LLS diseases ( Savadi

    et al., 2012). Out of 259 EST-SSR markers, which were developed

    using   A. hypogaea   ESTs (NCBI database), SSR_GO340445 and

    SSR_HO115759 were found to be closely linked to the rust resis-

    tance gene (Mondal et al., 2012b). Chen et al. (2008a, 2008b, 2011)

    identified and cloned the resistance gene to TSWV and character-

    ized two peanut oxalate oxidase genes.

    In response to A.  avus infection, many genes were found to beup-regulated in peanuts, of which PR-10 and pathogenesis-induced

    protein (PIP) genes were cloned (Xie et al., 2009a,b). Resveratrol

    imparts resistance in plants against both UV radiation and fungal

    infection; the gene that synthesizes resveratrol synthase has been

    cloned from peanuts, and expression analysis indicates that it is

    expressed in the root (Zhou et al., 2008; Han et al., 2010). Lipid

    transfer proteins (LTP), which were reported to be involved in

    disease resistance in plants, have also been cloned from peanuts

    (Zhao et al., 2009).

    Transcriptome analysis of the migratory plant parasitic nema-

    tode   Ditylenchus africanus   (peanut pod nematode) from mixed

    stages revealed the involvement of putative proteins in develop-

    mental and reproductive processes, as well as the role of unigenes

    in oxidative stress and anhydrobiosis (Haegeman et al., 2009). Eightdifferentially expressed genes were identified in  A. stenosperma

    roots in response to  M. arenaria  infection using  in silico  ESTs and

    microarray analysis (Guimar~aes et al., 2010, 2011).

    From non-protein coding genes, microRNAs were transcribed by

    RNA polymerase II (Kim, 2005), and understanding of its functional

    and regulatory roles could open new windows for crop improve-

    ment including disease tolerance. Zhao et al. (2010) and Pan and Liu

    (2010)   have identied 89 peanut miRNAs belonging to 14 new

    miRNA and 22 conserved miRNA families, which can provide the

    basis for peanut miRNA research for resistance to different stresses.

    Relationships between functional molecules and plant phenotypes

    can be studied more specically using proteomics (Wang et al.,

    2011), and proteins that may play roles in aatoxin resistance in

    imbedded peanut seeds have been discovered through proteomic

    studies (Wang et al., 2008b).

    In eukaryotes, epigenetic regulation is known to contribute to

    gene silencing and plays critical roles in development and genome

    defence against viruses, transposons, and transgenes (Lister et al.,

    2008; Gehring et al., 2006; La et al., 2011). It has been observed

    that bacterially infected plant tissue shows a net reduction in DNA

    methylation, which may affect the disease resistance genes

    responsible for surveillance against pathogens (Alvarez et al., 2010).

    An investigation regarding epigenetic regulation mechanisms of 

    the peanut allergen gene   Arah3   in developing peanut embryos

    demonstrated an association between the loss of histone H3 from

    the proximal promoter and high expression of   Arah3  during em-

    bryo maturation (Fu et al., 2010). For better understanding and

    management of various bioticstressesin peanuts, there is an urgent

     Table 5

    List of QTLs identified for various biotic-stress related traits in peanut.

    Traits No of QTLs identied Phenotypic variance explained (PVE) % Reference

    LLS 28 10.07e67.8   Khedikar et al., 2010;

    Sujay et al., 2012

    Leaf rust 13 2.54e82.62   Khedikar et al., 2010; Sujay et al., 2012

     Aspergillus  avus   6 6.2e22.7   Liang et al., 2009

    Bruchid resistance 13 13e82   Mondal et al., 2014

    Aphid vector of rosette disease 8 1.18e

    76.16   Herselman et al., 2004In F 2 map 54 (Total)   e   Wang et al., 2013, 2014

    TSWV 15 4.40e34.92

    Leaf spot 37 6.61e27.35

    Thrips 02 12.14e19.43

    In F 5 map 23 (Total)   e

    Thrips 01 5.86

    TSWV 09 5.20e14.14

    Leaf spot 13 5.95e21.45

    LLS 05 4.2e43.8   Leal-Bertioli et al., 2009

    TSWV 01 16.7   Guo et al., 2013

    LLS 03 12.42e20.59

    G.P. Mishra et al. / Crop Protection 77 (2015) 74e86    79

    http://www.ncbi.nlm.nih.gov/http://www.ncbi.nlm.nih.gov/http://www.ncbi.nlm.nih.gov/

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    7/13

    need to determine the epigenetic regulatory mechanisms of 

    different biotic stress resistant genes and promoters.

    For global gene expression proling, microarray analysis is a

    powerful tool (Casson et al., 2005). Because a peanut gene chip is

    commercial unavailability, high-throughput gene expression anal-

    ysis is currently very limited. However, peanut microarrays have

    been specically designed to solve particular issues, such as the

    characterization of  A. parasiticus infection-induced changes in gene

    expression and gene expression proling in different peanut tissues

    (Luo et al., 2005a; Payton et al., 2009). Approximately 25,000 ESTs

    from cDNA libraries were constructed using the bacterial wilt

    resistant peanut leaf before and after bacterial infection (Huang

    et al., 2008).  Shan et al. (2007)  used soybean gene chips to anal-

    yse the differential gene expression of peanut varieties that are

    resistant and susceptible to   A.   avus   infection. Using microarray

    analysis, Guoet al. (2011) identied62 genes in Aspergillus resistant

    peanut cultivars that were up-expressed in response to  Aspergillus

    infection, and 22 putative resistance genes that were constitutively

    overexpressed. In addition to microarray-based gene expression

    analysis, the expression and regulation of individual genes has also

    been studied in peanuts (Wang et al., 2011). There is a need for the

    development of a comprehensive genome-scale platform for

    developing  Aspergillus-resistant cultivars through targeted marker-assisted breeding and genetic engineering.

    10. Transgenic approach for biotic stress management

    (Fungal, viral, bacterial and insect-pest resistance)

    The lack of available resistance genes within crossable germ-

    plasms of peanuts necessitates the use of genetic engineering

    strategies to impart genetic resistance against various biotic

    stresses (Vasavirama and Kirti, 2012). Transgenic peanut lines

    possessing fungal resistance genes offer an alternative to traditional

    resistance and fungicide applications in managing fungal diseases

    (Chenault et al., 2005).   Vasavirama and Kirti (2012)   generated

    transgenic peanuts using a double gene construct with   SniOLP 

    (Solanum nigrum   osmotin-like protein) and   Rs- AFP2   (Raphanussativus antifungal protein-2) genes under separate constitutive 35S

    promoters, which then showed enhanced disease resistance to LLS.

    Transgenic peanuts with rice chitinase-3 overexpression (with the

    CaMV 35S promoter) exhibited a resistance for leaf spot that was

    higher than that of the control, and a good correlation was observed

    between chitinase activity and fungal pathogen resistance (Iqbal

    et al., 2012).

    Similarly, peanuts transgenic for the chitinase gene (Rchit ) from

    rice showed 2- to 14-fold higher chitinase activity than the wild

    type, and a signicant negative correlation was observed between

    the chitinase activity and the frequency of infection to the three

    tested pathogens ( A.   avus,   LLS and rust) (Prasad et al., 2013).

    Transgenic peanut lines containing antifungal genes (rice chitinase

    and/or alfalfa glucanase), when evaluated for their reaction toSclerotinia blight, showed varying degrees of resistance (Chenault

    et al., 2005). Signicantly reduced lesion size was recorded in

    transgenic plants expressing a barley oxalate oxidase gene

    compared to the controls, which means that oxalate oxidase can

    confer enhanced resistance to Sclerotinia blight in peanuts

    (Livingstone et al., 2005). A non-heme chloroperoxidase gene (cpo-

    p) from Pseudomonas pyrrocinia, a growth inhibitor of mycotoxin-

    producing fungi, introduced into peanuts resulted in transgenic

    plants that showed inhibition of   A.   avus   hyphal growth and

    reduced aatoxin contamination of peanut seeds (Niu et al., 2009).

    The epidemiology of PBNV is yet to be fully understood for

    peanut, including the search for linked markers for resistance

    gene(s) and the development resistant transgenic lines for its

    management. To this end, attempts have been made to develop

    transgenic peanuts expressing PBNV genes using viral coat protein

    (CP) genes (Satyanarayana et al., 1995). Evaluation of transgenic

    peanuts with CP genes showed that resistance could be achieved

    against PSND (Mehta et al., 2013). Transgenic peanut lines con-

    taining the CP gene of the peanut stripe virus (PStV) were less

    susceptible to PStV. TSWV has a broad host range, and it spreads

    through ubiquitous thrips. The nucleocapsid (N) protein gene of the

    lettuce isolate of TSWV was inserted into the peanut genome,

    which showed divergent levels of gene expression (Yang et al.,

    1998). Transgenic peanuts expressing the N protein of TSWV and

    subjected to natural infection of the virus under   eld conditions

    resulted in a signicantly lower incidence of spotted wilt compared

    to that of non-transgenic lines (Yang et al., 2004). Increased insect

    tolerance of the transgenic plants was recorded when the cowpea

    trypsin inhibitor gene was transfected into peanuts (Xu et al., 2003;

    Zhuang et al., 2003). The transgenic lines developed against various

    diseases in peanuts are compiled and presented in  Table 6, but to

    date no transgenic peanut cultivars have been released

    commercially.

    During the last decade, strong public opposition to genetically

    modied (GM) food crops, especially in European countries, are

     juxtaposed with issues such as freedom to operate related to

    patented technologies (Holbrook et al., 2011). However, during lasttwo years, many patents have been approved in various countries

    for  eld trials of GM food crops. Two genetically modied types of 

    corn, namely   ‘TC1507’  and   ‘SmartStax’  developed by the US com-

    panies Pioneer and Monsanto, respectively, won EU approval for

    their cultivation in EU countries in February of 2014 and November

    of 2013, respectively (Mcmanus, 2014). In India, the Union Ministry

    of Environment and Forests (MoEF) has approved eld trials for GM

    food crops (rice, wheat, maize, castor and cotton) to determine

    their suitability for commercial production (Business Standard,

    2014). Altogether, the possibility is good that various transgenic

    peanut crops will be used in the next few years.

    11. Achievements and future prospects

    The dynamic challenges of peanut farming demand a quick

    response from breeders to develop newcultivars, a process that can

    be aided by the application of molecular markers (Chu et al., 2011).

    ‘NemaTAM ’  is the rst root-knot nematode-resistant peanut variety

    developed using the MABC approach in the USA (Simpson et al.,

    2003). The   ‘Tifguard High O/L’   cultivar, with nematode resistance

    and a high oleic:linoleic acid (high O:L) ratio in seeds, was devel-

    oped using Tifguard  (nematode-resistant cultivar) as the recurrent

    female parent and  Georgia-02C  and  Florida-07  (high O:L cultivars)

    as donor parents after three rounds of accelerated backcrossing

    using MAS (Chu et al., 2011).

    A QTL region explaining up to 82.62% of the phenotypic varia-

    tion for rust resistance was introgressed from cultivar  ‘GPBD 4’ into

    three rust susceptible varieties (ICGV 91114, JL 24 and TAG 24)through MABC using four markers (IPAHM103, GM2079, GM1536,

    GM2301). This has generated several promising introgression lines

    with enhanced rust resistance and higher yields. These linked

    markers may be used to improve rust resistance in peanut breeding

    programmes across the world (Varshney et al., 2014). Through

    validating LLS and rust resistance-linked markers in different

    peanut genotypes, six superior genotypes were identied. More-

    over, three lines that are superior for productivity traits and equal

    for disease resistance to GPBD 4 (the resistant parent) are being

    included in variety release trials for multi-location evaluation

    (Sukruth et al., 2015).  Gajjar et al. (2014)  studied 22 SSR markers

    reportedly linked to rust and LLS disease resistance on 95 diverse

    genotypes for marker validation, from which 16 SSRs could be

    validated.

    G.P. Mishra et al. / Crop Protection 77 (2015) 74e86 80

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    8/13

    Because of evolutionary selection pressure and their high

    genetic-diversity, wild peanut species are considered to be excel-

    lent sources of various disease resistance genes that remain under-

    utilized (Stalker and Simpson, 1995). A dense genetic linkage map

    should enable breeders to effectively pyramid genes for various

    biotic stress resistances, such as rust, LLS, and nematode resistance,

    into agronomically superior lines in a much shorter time period

    than would be possible by conventional techniques (Gajjar et al.,

    2014). RILs are being developed to map various genes/QTLs un-

    derlying these traits. Currently, more than 14,000 SSR markers are

    available from different sources; however, substantial efforts are

    still required to develop suf cient PCR-based markers for the

    construction of a high-density genetic linkage map and for routine

    applications in the molecular breeding of peanuts (Mishra et al.,2014; Bosamia et al., 2015).

    The International Peanut Genome Initiative with a time line of 

    2012e2016 is working on different aspects of biotic stress man-

    agement at the genome level, including screening peanut acces-

    sions for new sources of disease resistance, discovering new

    sources of disease resistance, identifying a core set of informative

    markers for their deployment in resistance breeding programmes,

    searching and validating markers that can be used in pre-breeding

    for disease and pest resistance, placing candidate genes for disease

    resistance on the  Arachis genetic map, and identifying core sets of 

    markers for QTLs associated with biotic stress resistance (http://

    www.peanutbioscience.com/peanutgenomeinitiative.html ).

    Nevertheless, the genomic progress made in peanuts over the last

    few years has been quite encouraging, and it is expected that aclearer genomic picture of peanuts will be available by the year

    2016 for researchers to utilize.

    Recently, the International Peanut Genome Initiative (IPGI) has

    sequenced the genome of two wild species,   A. duranensis   (AA

    genome) and   A. ipaensis  (BB genome), supposedly the ancestral

    parents of cultivated peanut, which is an allotetraploid species. The

    hope is that the available sequence information will provide re-

    searchers with approximately 96 percent of all peanut genomic

    information and provide the molecular map needed to more

    quickly breed drought-resistant, disease-resistant, lower-input and

    higher-yielding varieties (http://www.icrisat.org/newsroom/news-

    releases/icrisat-pr-2014-media13.htm ;   Mondal and Badigannavar,

    2015).

    However, compared to other crops such as rice, soybeans and

    chickpeas, the ongoing molecular breeding programme for peanuts

    is relatively slow primarily due to low genetic diversity among the

    cultivated gene pool, introgression barriers between wild and

    cultivated lines, non-availability of genome sequence information

    until last year and inadequate funding. Because a draft sequence of 

    the Arachis genome (AA and BB) is available, the expectation is that

    in the next few years the quality of peanuts will be improved

    through the use of modern biotechnological approaches ( Janila

    et al., 2013). Ongoing research efforts across the world have not

    only resulted in better understanding of the peanut genome but

    also facilitated ongoing marker-assisted peanut breeding programs

    (Holbrook et al., 2011). The use of biotechnological interventions in

    peanut breeding for the development of resistance cultivars against

    various biotic stresses is quite promising. Despite substantial ad-vancements in the biotic management strategies, the global peanut

    production is still threatened by a multitude of insect pests and

    diseases. The situation demands judicious blending of conventional

    and modern crop improvement technologies for more ef cient and

    rapid tackling of these problems (Krishna et al., 2015). An outline of 

    the tentative biotechnological interventions for peanuts is pre-

    sented in Fig. 1.

    12. Conclusions

    We have reviewed the recent biotechnological developments

    for the biotic stress management of peanut crops with special

    preference given to genomic approaches. The goal of these studies

    is to  nd more effective and economical high-throughput tools tomanage various biotic stresses in peanuts. Conventional breeding

    along with phenotyping tools have largely been used in peanut

    improvement programs, and low genetic variability has been

    considered one of the major bottlenecks. Therefore, mutations

    were used to introduce variability, and wide-hybridizations were

    attempted to determine the variability from wild   Arachis   species.

    For the creation of newsourcesof genetic diversity, approaches that

    are being explored include the development of transgenics, TILLING

    (targeting-induced local lesions in genomes) populations and

    synthetic allotetraploids (Holbrook et al., 2011). Additionally, a few

    other robust mapping populations such as NAM (nested association

    mapping) and MAGIC (multi-parent advanced generation inter-

    cross) are being developed by various institutions working on

    peanut crops. To date, the genetic basis of many important traits in

     Table 6

    Transgenics developed for various biotic stress resistances in peanut.

    Genes Resistance Reference

    Coat protein PBND   Satyanarayana et al., 1995

    cry1AC Elasmopalpus lignoscellus   Singsit et al., 1997

    Nucleocapsid (N) Tomato spotted wilt virus   Yang et al., 1998; Magbanua et al., 2000; Yang et al., 2004

    C owpea trypsin i nhi bitor gene Insec t toleran ce   Xu et al., 2003; Zhuang et al., 2003

    Coat protein Peanut stripe potyvirus   Higgins et al., 2004

    Chitinase and glucanase Sclerotinia blight   Chenault et al., 2005Barley oxalate oxidase Sclerotinia blight   Livingstone et al., 2005; Partridge-Telenko et al., 2011

    cry1EC Spodoptera litura   Tiwari et al., 2008; Tiwari et al., 2011

    cry1X Helicoverpa armigera and  Spodoptera litura   Entoori et al., 2008

    cry1EC  and  Chi11 Spodoptera litura and  Phaeoisariopsis personata   Beena et al., 2008

    Coat Protein gene PStV PStV     Hapsoro et al., 2008

    Mustard defensin LLS   Anuradha et al., 2008

    Chloroperoxidase (cpo-p)   A. avus   Niu et al., 2009

    Rice chitinase-3 Leaf spot   Iqbal et al., 2012

    Rchit  and  CHI    Fusarium wilt and Cercospora arachidicola   Rohini and Sankara, 2001; Iqbal et al., 2011

    SniOLP  and  Rs- AFP2   LLS   Vasavirama and Kirti, 2012

    Rice chitinase gene (Rchit )   A. avus, LLS and rust   Prasad et al., 2013

    Coat protein PSND   Mehta et al., 2013

    cry1AcF Spodoptera litura   Keshavareddy et al., 2013

    cry8Ea1 Holotrichia parallela   Geng et al., 2013

    Tfgd2-RsAFP2 fusion gene ELS and LLS   Bala et al., 2015

    G.P. Mishra et al. / Crop Protection 77 (2015) 74e86    81

    http://www.peanutbioscience.com/peanutgenomeinitiative.htmlhttp://www.peanutbioscience.com/peanutgenomeinitiative.htmlhttp://www.icrisat.org/newsroom/news-releases/icrisat-pr-2014-media13.htmhttp://www.icrisat.org/newsroom/news-releases/icrisat-pr-2014-media13.htmhttp://www.icrisat.org/newsroom/news-releases/icrisat-pr-2014-media13.htmhttp://www.icrisat.org/newsroom/news-releases/icrisat-pr-2014-media13.htmhttp://www.peanutbioscience.com/peanutgenomeinitiative.htmlhttp://www.peanutbioscience.com/peanutgenomeinitiative.html

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    9/13

  • 8/18/2019 JCRP_2015_Advancements in Molecular Marker Developments in Peanuts

    10/13

    159, 823e837.Business Standard, 2014. GM Crops: Field Trials Get Green Signal. February 28, 2014.

    http://www.business-standard.com/article/economy-policy/gm-crops-eld-trials-get-green-signal-114022701174_1.html.

    Casson, S., Spencer, M., Walker, K., Lindsey, K., 2005. Laser capture micro-dissectionfor the analysis of gene expression during embryogenesis of Arabidopsis. Plant

     J. 42, 111e123.Chen, X., Brenneman, T., Culbreath, A., Holbrook, C., Guo, B., 2008b. Characterization

    of two peanut oxalate oxidase genes and development of peanut cultivarsresistant to stem rot (Sclerotium rolfsii). Phytopathology 98, S36.

    Chen, X., Culbreath, A., Brenneman, T., Holbrook, C., Guo, B., 2008a. Identificationand cloning of TSWV resistance gene(s) in cultivated peanuts and developmentof markers for breeding selection. Phytopathology 98, S36.

    Chen, X., Wang, M.L., Holbrook, C., Culbreath, A., Liang, X., Brenneman, T., Guo, B.,2011. Identification and characterization of a multigene family encodinggermin-like proteins in cultivated peanut ( Arachis hypogaea L.). Plant Mol. Biol.Rep. 29, 389e403.

    Chenault, K.D., Melouk, H.A., Payton, M.E., 2005. Field reaction to Sclerotinia blightamong transgenic peanut lines containing antifungal genes. Crop Sci. 45,511e515.

    Choi, K., Burow, M.D., Church, G., Burow, G., Paterson, A.H., Simpson, C.E., Starr, J.L.,1999. Genetics and mechanism of resistance to  Meloidogyne arenaria in peanutgermplasm. J. Nematol. 31, 283e290.

    Chopra, R., Burow, G., Farmer, A., Mudge, J., Simpson, C.E., Burow, M.D., 2014.Comparisons of   de novo  transcriptome assemblers in diploid and polyploidspecies using peanut ( Arachis spp.) RNA-seq data. PLoS One 9 (12), e115055.http://dx.doi.org/10.1371/journal.pone.0115055 .

    Chopra, R., Burow, G., Farmer, E., Mudge, J., Simpson, C.E., Wilkins, T.A., Baring, M.R.,Puppala, N., Chamberlin, K.D., Burow, M.D., 2015. Next-generation tran-scriptome sequencing, SNP discovery and validation in four market classes of peanut,   Arachis hypogaea   L. Mol. Genet. Genomics.   http://dx.doi.org/10.1007/s00438-014-0976-4 .

    Chu, Y., Holbrook, C.C., Timper, P., Ozias-Akins, P., 2007. Development of a PCR-based molecular marker to select for nematode resistance in peanut. Crop Sci.47, 841e847.

    Chu, Y., Wu, C.L., Holbrook, C.C., Tillman, B.L., Person, G., Ozias-Akins, P., 2011.Marker-assisted selection to pyramid nematode resistance and the high oleictrait in peanut. Plant Genome 4, 110e117.

    Church, G.T., Simpson, C.E., Burow, M.D., Paterson, A.H., Starr, J.L., 2000. Use of RFLPmarkers for identication of individuals homozygous for resistance to  Meloi-dogyne arenaria  in peanut. Nematology 2, 575e580.

    Cilliers, A.J., Herselman, L., Pretorius, Z.A., 2000. Genetic variability within andamong mycelial compatibility groups of   Sclerotium rolfsii   in South Africa.Phytopathology 90, 1026e1031.

    Ding, Y.F., Wang, C.T., Tang, Y.Y., Wang, X.Z., Wu, Q., Hu, D.Q., Yu, H.T., Zhang, J.C.,Cui, F.G., Song, G.S., Gao, H.Y., Yu, S.L., 2012. Isolation and analysis of differen-tially expressed genes from peanut in response to challenge with  Ralstonia

    solanacearum. Electron. J. Biotechnol.   http://dx.doi.org/10.2225/vol15-issue5-fulltext-1.Dodia, S.M., Mishra, G.P., Radhakrishnan, T., Dobaria, J.R., 2014. Identication of 

     Aspergillus avus  isolates for developing biocontrol agent based on the gene-defects in the aatoxin biosynthesis gene-cluster and  anking-regions. J. PureApp. Microbiol. 8, 4623e4635.

    Dwivedi, S.L., Crouch, J.H., Nigam, S.N., Ferguson, M.E., Paterson, A.H., 2003. Mo-lecular breeding of groundnut for enhanced productivity and food security inthe semi-arid tropics: opportunities and challenges. Adv. Agron. 80, 153e221.

    Dwivedi, S.L., Pande, S., Rao, J.N., Nigam, S.N., 2002. Components of resistance to lateleaf spot and rust among interspecic derivatives and their signicance in afoliar disease resistance breeding in groundnut ( Arachis hypogaea L.). Euphytica125, 81e88.

    Entoori, K., Sreevathsa, R., Arthikala, M.K., Kumar, P.A., Kumar, A.R.V.,Madhusudhan, B., Makarla, U., 2008. A chimeric  cry1X  gene imparts resistanceto   Spodoptera litura   and   Helicoverpa armigera   in the transgenic groundnut.EurAsia J. Biosci. 2, 53e65.

    Farr, D.F., Bills, G.F., Chamuris, G.P., Rossman, A.Y., 1989. Fungi on Plants and PlantProducts in the United States. American Phytopathological Society, St. Paul,

    Minnesota.Fu, G., Zhong, Y., Li, C., Li, Y., Lin, X., Liao, B., Tsang, E.W., Wu, K., Huang, S., 2010.

    Epigenetic regulation of peanut allergen gene   Arah3   in developing embryos.Planta 231, 1049e1060.

    Gajjar, K.N., Mishra, G.P., Radhakrishnan, T., Dodia, S.M., Rathnakumar, A.L.,Kumar, N., Kumar, S., Dobaria, J.R., Kumar, A., 2014. Validation of SSR markerslinked to the rust and late leaf spot diseases resistance in diverse peanut ge-notypes. Aust. J. Crop Sci. 8, 927e936.

    Garcia, G.M., Stalker, H.T., Shroeder, E., Kochert, G., 1996. Identication of RAPD,SCAR, and RFLP markers tightly linked to nematode resistance genes intro-gressed from Arachis cardenasii  into Arachis hypogaea. Genome 39, 836e845.

    Gautami, B., Fonceka, D., Pandey, M.K., Moretzsohn, M.C., Sujay, V., Qin, H., Hong, Y.,Faye, I., Chen, X., Prakash, A.B., Shah, T.M., Gowda, M.V.C., Nigam, S.N., Liang, X.,Hoisington, D.A., Guo, B., Bertioli, D.J., Rami, J.F., Varshney, R.K., 2012. An in-ternational reference consensus genetic map with 897 marker loci based on 11mapping populations for tetraploid groundnut ( Arachis hypogaea L.). PLoS One 7(7), e41213.  http://dx.doi.org/10.1371/journal.pone.0041213.

    Gehring, M., Huh, J.H., Hsieh, T.F., Penterman, J., Choi, Y., Harada, J.J., Goldberg, R.B.,Fischer, R.L., 2006. DEMETER DNA glycosylase establishes MEDEA polycomb

    gene self-imprinting by allele-specic demethylation. Cell 124, 495e506.Geng, L., Chi, J., Shu, C., Gresshoff, P.M., Song, F., Huang, D., Zhang, J., 2013.

    A chimeric  cry8Ea1  gene  anked by MARs ef ciently controls  Holotrichia par-allela. Plant Cell Rep. 32, 1211e1218.

    Gowda, C.L.L., Upadhyaya, H.D., Sharma, S., Varshney, R.K., Dwivedi, S.L., 2013.Exploiting genomic resources for ef cient conservation and use of chickpea,groundnut, and pigeonpea collections for crop improvement. Plant Genome 6(3). http://dx.doi.org/10.3835/plantgenome2013.05.0016.

    Guimar~aes, P.M., Brasileiro, A.C.M., Leal-Bertioli, S.C.M., Pappas, G., Togawa, R.,Bonfim, O., 2011. Comparative 454 pyrosequencing of transcripts from two wild

     Arachis genotypes under biotic and abiotic stress. In: Proc: Plant and AnimalGenome XIX Conference, 15e19th Jan; 2011, San Diego, USA.

    Guimar~aes, P.M., Brasileiro, A.C.M., Morgante, C.V., Martins, A.C.Q., Pappas, G.,Silva Jr., O.B., Togawa, R., Leal-Bertioli, S.C.M., Araujo, A.C.G., Moretzsohn, M.C.,Bertioli, D.J., 2012. Global transcriptome analysis of two wild relatives of peanutunder drought and fungi infection. BMC Genomics 13, 387.  http://dx.doi.org/10.1186/1471-2164-13-387 .

    Guimar~aes, P.M., Brasileiro, A.C.M., Proite, K., Araújo, A .C.G., Leal-Bertioli, S.C.M., Pic-Taylor, A., daSilva, F.R., Morgante, C.V., Ribeiro, S.D.G., Bertioli, D.J., 2010. A studyof gene expression in the nematode resistant wild peanut relative,   Arachisstenosperma, in response to challenge with   Meloidogyne arenaria. Trop. PlantBiol. 3, 183e192.

    Guo, B., Chen, X., Hong, Y., Liang, X., Dang, P., Brenneman, T., Holbrook, C.,Culbreath, A., 2009. Analysis of gene expression proles in leaf tissues of cultivated peanuts and development of EST-SSR markers and gene discovery.Int. J. Plant Genomics.   http://dx.doi.org/10.1155/2009/715605.

    Guo, B., Fedorova, N.D., Chen, X., Wan, C.H., Wang, W., Nierman, W.C., Bhatnagar, D.,Yu, J., 2011. Gene expression proling and identication of resistance genes to

     Aspergillus   avus  infection in peanut through EST and microarray strategies.Toxins 3, 737

    e753.

    Guo, B., Pandey, M., Culbreath, A., Brenneman, T., Holbrook, C., Wang, M.L.,Barkley, N., 2013. Saturation of Genetic Maps for Identication of QTLs Con-trolling Disease Resistance, Oil Quality, and Morphological Descriptors in Pea-nut ( Arachis hypogaea   L.). Georgia Peanut Commission Research. Report,February 13, 2013. www.americanpeanuts.com.

    Guo, B.Z., Chen, X., Dang, P., Scully, B.T., Liang, X., Holbrook, C.C., Yu, J.,Culbreath, A.K., 2008. Peanut gene expression profiling in developing seeds atdifferent reproduction stages during  Aspergillus parasiticus  infection. BMC Dev.Biol. 8, 12.  http://dx.doi.org/10.1186/1471-213X-8-12.

    Haegeman, A., Mitreva, Jacob, J., Vanholme, B., Gheysen, G., 2009. Expressedsequence tags of the peanut pod nematode   Ditylenchus africanus: the   rsttranscriptome analysis of an Anguinid nematode. Mol. Biochem. Parasit. 167,32e40.

    Halward, T., Stalker, H.T., Kochert, G., 1993. Development of an RFLP linkage map indiploid peanut species. Theor. Appl. Genet. 87, 379e384.

    Han, J.J., Liu, W., Bi, Y.P., 2010. Molecular cloning of peanut resveratrol syntheticenzyme 1 (PNRS1) and its expression in prokaryote. Acta Agron. Sin. 36,

    341e

    346.Hapsoro, D., Aswidinnoor, H., Suseno, R., Jumanto, Sudarsono, D., 2008. Inheritanceof resistance to PStV in transgenic peanuts containing cp PStV gene. J. HPT Trop.8, 31e38.

    Harlton, C.E., L evesque, C.A., Punja, Z.K., 1995. Genetic diversity in   Sclerotium(Athelia) rolfsii and related species. Phytopathology 85, 1269e1281.

    He, G., Prakash, C.S., 2001. Evaluation of genetic relationships among botanicalvarieties of cultivated peanut ( Arachis hypogaea   L.) using markers. Genet.Resour. Crop Evol. 48, 347e352.

    Herselman, L., 2003. Genetic variation among Southern African cultivated peanut( Arachis hypogaea L.) genotypes as revealed by AFLP analysis. Euphytica 133,319e327.

    Herselman, L., Thwaites, R., Kimmins, F.M., Courtois, B., van der Merwe, P.J.,Seal, S.E., 2004. Identication and mapping of AFLP markers linked to peanut( Arachis hypogaea  L.) resistance to the aphid vector of groundnut rosette dis-ease. Theor. Appl. Genet. 109, 1426e1433.

    Higgins, C.M., Hall, R.M., Mitter, N., Cruickshank, A., Dietzgen, R.G., 2004. Peanutstripe potyvirus resistance in peanut ( Arachis hypogaea L.) plants carrying viralcoat protein gene sequences. Transgenic Res. 13, 59e67.

    Holbrook, C.C., Ozias-Akins, P., Chu, Y., Guo, B., 2011. Impact of molecular geneticresearch on peanut cultivar development. Agronomy 1, 3e17.

    Holbrook, C.C., Stalker, H.T., 2003. Peanut breeding and genetic resources. PlantBreed. Rev. 22, 297e356.

    Hong, Y.B., Li, S.X., Liu, H.Y., Zhou, G.Y., Chen, X.P., Wen, S.J., Liang, X.Q., 2009.Correlation analysis of SSR markers and host resistance to  Aspergillus   avusinfection in peanut ( Arachis hypogaea L.). Mol. Plant Breed. 7, 360e364.

    Hou, H.M., Liao, B.S., Lei, Y., Ren, X.P., Wang, S.Y., Li, D., Jiang, H.F., Huang, J.Q.,Chen, B.Y., 2007. Identication of AFLP markers for resistance to peanut rust.Chin. J. Oil Crop Sci. 29, 89e92.

    Huang, J.Q., Yan, L.Y., Lei, Y., Jiang, H.F., Liao, B.S., 2008. Peanut cDNA library con-struction and EST sequence analysis. Chin. J. Oil Crop Sci. 10, 121e125.

    Iqbal, M.M., Nazir, F., Ali, S., Asif, M.A., Zafar, Y., Iqbal, J., Ali, G.M., 2012. Overexpression of rice chitinase gene in transgenic peanut ( Arachis hypogaea   L.)improves resistance against leaf spot. Mol. Biotechnol. 50, 129e136.

    Iqbal, M.M., Zafar, Y., Nazir, F., Ali, S., Iqbal, J., Asif, M.A., Rashid, O., Ali, G.M., 2011.Over expression of bacterial chitinase gene in Pakistani peanut ( Arachis hypo-

     gaea L.) cultivar GOLDEN. Afr. J. Biotechnol. 10, 5838e5844. Janila, P., Nigam, S.N., Pandey, M.K., Nagesh, P., Varshney, R.K., 2013. Groundnut

    G.P. Mishra et al. / Crop Protection 77 (2015) 74e86    83

    http://refhub.elsevier.com/S0261-2194(15)30074-0/sref15http://refhub.elsevier.com/S0261-2194(15)30074-0/sref15http://refhub.elsevier.com/S0261-2194(15)30074-0/sref15http://www.business-standard.com/article/economy-policy/gm-crops-field-trials-get-green-signal-114022701174_1.htmlhttp://www.business-standard.com/article/economy-policy/gm-crops-field-trials-get-green-signal-114022701174_1.htmlhttp://www.business-standard.com/article/economy-policy/gm-crops-field-trials-get-green-signal-114022701174_1.htmlhttp://www.business-standard.com/article/economy-policy/gm-crops-field-trials-get-green-signal-114022701174_1.htmlhttp://refhub.elsevier.com/S0261-2194(15)30074-0/sref17http://refhub.elsevier.com/S0261-2194(15)30074-0/sref17http://refhub.elsevier.com/S0261-2194(15)30074-0/sref17http://refhub.elsevier.com/S0261-2194(15)30074-0/sref17http://refhub.elsevier.com/S0261-2194(15)30074-0/sref18http://refhub.elsevier.com/S0261-2194(15)30074-0/sref18http://refhub.elsevier.com/S0261-2194(15)30074-0/sref18http://refhub.elsevier.com/S0261-2194(15)30074-0/sref18http://refhub.elsevier.com/S0261-2194(15)30074-0/sref18http://refhub.elsevier.com/S0261-2194(15)30074-0/sref19http://refhub.elsevier.com/S0261-2194(15)30074-0/sref19http://refhub.elsevier.com/S0261-2194(15)30074-0/sref19http://refhub.elsevier.com/S0261-2194(15)30074-0/sref19http://refhub.elsevier.com/S0261-2194(15)30074-0/sref19http://refhub.elsevier.com/S0261-2194(15)30074-0/sref20http://refhub.elsevier.com/S0261-2194(15)30074-0/sref20http://refhub.elsevier.com/S0261-2194(15)30074-0/sref20http://refhub.elsevier.com/S0261-2194(15)30074-0/sref20http://refhub.elsevier.com/S0261-2194(15)30074-0/sref20http://refhub.elsevier.com/S0261-2194(15)30074-0/sref20http://refhub.elsevier.com/S0261-2194(15)30074-0/sref20http://refhub.elsevier.com/S0261-2194(15)30074-0/sref20http://refhub.elsevier.com/S0261-2194(15)30074-0/sref21http://refhub.elsevier.com/S0261-2194(15)30074-0/sref21http://refhub.elsevier.com/S0261-2194(15)30074-0/sref21http://refhub.elsevier.com/S0261-2194(15)30074-0/sref21http://refhub.elsevier.com/S0261-2194(15)30074-0/sref22http://refhub.elsevier.com/S0261-2194(15)30074-0/sref22http://refhub.elsevier.com/S0261-2194(15)30074-0/sref22http://refhub.elsevier.com/S0261-2194(15)30074-0/sref22http://refhub.elsevier.com/S0261-2194(15)30074-0/sref22http://refhub.elsevier.com/S0261-2194(15)30074-0/sref22http://dx.doi.org/10.1371/journal.pone.0115055http://dx.doi.org/10.1007/s00438-014-0976-4http://dx.doi.org/10.1007/s00438-014-0976-4http://dx.doi.org/10.1007/s00438-014-0976-4http://refhub.elsevier.com/S0261-2194(15)30074-0/sref25http://refhub.elsevier.com/S0261-2194(15)30074-0/sref25http://refhub.elsevier.com/S0261-2194(15)30074-0/sref25http://refhub.elsevier.com/S0261-2194(15)30074-0/sref25http://refhub.elsevier.com/S0261-2194(15)30074-0/sref25http://refhub.elsevier.com/S0261-2194(15)30074-0/sref26http://refhub.elsevier.com/S0261-2194(15)30074-0/sref26http://refhub.elsevier.com/S0261-2194(15)30074-0/sref26http://refhub.elsevier.com/S0261-2194(15)30074-0/sref26http://refhub.elsevier.com/S0261-2194(15)30074-0/sref26http://refhub.elsevier.com/S0261-2194(15)30074-0/sref27http://refhub.elsevier.com/S0261-2194(15)30074-0/sref27http://refhub.elsevier.com/S0261-2194(15)30074-0/sref27http://refhub.elsevier.com/S0261-2194(15)30074-0/sref27http://refhub.elsevier.com/S0261-2194(15)30074-0/sref27http://refhub.elsevier.com/S0261-2194(15)30074-0/sref27http://refhub.elsevier.com/S0261-2194(15)30074-0/sref27http://refhub.elsevier.com/S0261-2194(15)30074-0/sref27http://refhub.elsevier.com/S0261-2194(15)30074-0/sref27http://refhub.elsevier.com/S0261-2194(15)30074-0/sref28http://refhub.elsevier.com/S0261-2194(15)30074-0/sref28http://refhub.elsevier.com/S0261-2194(15)30074-0/sref28http://refhub.elsevier.com/S0261-2194(15)30074-0/sref28http://refhub.elsevier.com/S0261-2194(15)30074-0/sref28http://refhub.elsevier.com/S0261-2194(15)30074-0/sref28http://refhub.elsevier.com/S0261-2194(15)30074-0/sref28http://dx.doi.org/10.2225/vol15-issue5-fulltext-1http://dx.doi.org/10.2225/vol15-issue5-fulltext-1http://dx.doi.org/10.2225/vol15-issue5-fulltext-1http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref30http://refhub.elsevier.com/S0261-2194(15)30074-0/sref31http://refhub.elsevier.com/S0261-2194(15)30074-0/sref31http://refhub.elsevier.com/S0261-2194(15)30074-0/sref31http://refhub.elsevier.com/S0261-2194(15)30074-0/sref31http://refhub.elsevier.com/S0261-2194(15)30074-0/sref31http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref32http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref33http://refhub.elsevier.com/S0261-2194(15)30074-0/sref34http://refhub.elsevier.com/S0261-2194(15)30074-0/sref34http://refhub.elsevier.com/S0261-2194(15)30074-0/sref34http://refhub.elsevier.com/S0261-2194(15)30074-0/sref35http://refhub.elsevier.com/S0261-2194(15)30074-0/sref35http://refhub.elsevier.com/S0261-2194(15)30074-0/sref35http://refhub.elsevier.com/S0261-2194(15)30074-0/sref35http://refhub.elsevier.com/S0261-2194(15)30074-0/sref35http://refhub.elsevier.com/S0261-2194(15)30074-0/sref35http://refhub.elsevier.com/S0261-2194(15)30074-0/sref36http://refhub.elsevier.com/S0261-2194(15)30074-0/sref36http://refhub.elsevier.com/S0261-2194(15)30074-0/sref36http://refhub.elsevier.com/S0261-2194(15)30074-0/sref36http://refhub.elsevier.com/S0261-2194(15)30074-0/sref36http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://refhub.elsevier.com/S0261-2194(15)30074-0/sref37http://dx.doi.org/10.1371/journal.pone.0041213http://dx.doi.org/10.1371/journal.pone.0041213http://refhub.elsevier.com/S0261-2194(15)30074-0/sref39http://refhub.elsevier.com/S0261-2194(15)30074-0/sref39http://refhub.elsevier.com/S0261-2194(15)30074-0/sref39http://refhub.elsevier.com/S0261-2194(15)30074-0/sref39http://refhub.elsevier.com/S0261-2194(15)30074-0/sref39http://refhub.elsevier.com/S0261-2194(15)30074-0/sref39http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://refhub.elsevier.com/S0261-2194(15)30074-0/sref40http://dx.doi.org/10.3835/plantgenome2013.05.0016http://refhub.elsevier.com/S0261-2194(15)30074-0/sref42http://refhub.elsevier.com/S0261-2194(15)30074-0/sref42http://refhub.elsevier.com/S0261-2194(15)30074-0/sref42http://refhub.elsevier.com/S0261-2194(15)30074-0/sref42http://refhub.elsevier.com/S0261-2194(15)30074-0/sref42http://refhub.elsevier.com/S0261-2194(15)30074-0/sref42http://refhub.elsevier.com/S0261-2194(15)30074-0/sref42http://refhub.elsevier.com/S0261-2194(15)30074-0/sref42http://dx.doi.org/10.1186/1471-2164-13-387http://dx.doi.org/10.1186/1471-2164-13-387http://dx.doi.org/10.1186/1471-2164-13-387http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://refhub.elsevier.com/S0261-2194(15)30074-0/sref44http://dx.doi.org/10.1155/2009/715605http://dx.doi.org/10.1155/2009/715605http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://refhub.elsevier.com/S0261-2194(15)30074-0/sref46http://www.americanpeanuts.com/http://www.americanpeanuts.com/http://dx.doi.org/10.1186/1471-213X-8-12http://refhub.elsevier.com/S0261-2194(15)30074-0/sref49http://refhub.elsevier.com/S0261-2194(15)30074-0/sref49http://refhub.elsevier.com/S0261-2194(15)30074-0/sref49http://refhub.elsevier.com/S0261-2194(15)30074-0/sref49http://refhub.elsevier.com/S0261-2194(15)30074-0/sref49http://refhub.elsevier.com/S0261-2194(15)30074-0/sref49http://refhub.elsevier.com/S0261-2194(15)30074-0/sref49http://refhub.elsevier.com/S0261-2194(15)30074-0/sref49http://refhub.elsevier.com/S0261-2194(15)30074-0/sref49http://refhub.elsevier.com/S0261-2194(15)30074-0/sref50http://refhub.elsevier.com/S0261-2194(15)30074-0/sref50http://refhub.elsevier.com/S0261-2194(15)30074-0/sref50http://refhub.elsevier.com/S0261-2194(15)30074-0/sref51http://refhub.elsevier.com/S0261-2194(15)30074-0/sref51http://refhub.elsevier.com/S0261-2194(15)30074-0/sref51http://refhub.elsevier.com/S0261-2194(15)30074-0/sref51http://refhub.elsevier.com/S0261-2194(15)30074-0/sref52http://refhub.elsevier.com/S0261-2194(15)30074-0/sref52http://refhub.elsevier.com/S0261-2194(15)30074-0/sref52http://refhub.elsevier.com/S0261-2194(15)30074-0/sref52http://refhub.elsevier.com/S0261-2194(15)30074-0/sref53http://refhub.elsevier.com/S0261-2194(15)30074-0/sref53http://refhub.elsevier.com/S0261-2194(15)30074-0/sref53http://refhub.elsevier.com/S0261-2194(15)30074-0/sref53http://refhub.elsevier.com/S0261-2194(15)30074-0/sref53http://refhub.elsevier.com/S0261-2194(15)30074-0/sref53http://refhub.elsevier.com/S0261-2194(15)30074-0/sref53http://refhub.elsevier.com/S0261-2194(15)30074-0/sref54http://refhub.elsevier.com/S0261-2194(15)30074-0/sref54http://refhub.elsevier.com/S0261-2194(15)30074-0/sref54http://refhub.elsevier.com/S0261-2194(15)30074-0/sref54http://refhub.elsevier.com/S0261-2194(15)30074-0/sref54http://refhub.elsevier.com/S0261-2194(15)30074-0/sref54http://refhub.elsevier.com/S0261-2194(15)30074-0/sref55http://refhub.elsevier.com/S0261-2194(15)30074-0/sref55http://refhub.elsevier.com/S0261-2194(15)30074-0/sref55http://refhub.elsevier.com/S0261-2194(15)30074-0/sref55http://refhub.elsevier.com/S0261-2194(15)30074-0/sref55http://refhub.elsevier.com/S0261-2194(15)30074-0/sref55http://refhub.elsevier.com/S0261-2194(15)30074-0/sref55http://refhub.elsevier.com/S0261-2194(15)30074-0/sref56http://refhub.elsevier.com/S0261-2194(15)30074-0/sref56http://refhub.elsevier.com/S0261-2194(15)30074-0/sref56http://refhub.elsevier.com/S0261-2194(15)30074-0/sref56http://refhub.elsevier.com/S0261-2194(15)30074-0/sref56http://refhub.elsevier.com/S0261-2194(15)30074-0/sref56http://refhub.elsevier.com/S0261-2194(15)30074-0/sref56http://refhub.elsevier.com/S0261-2194(15)30074-0/sref56http://refhub.elsevier.com/S0261-2194(15)30074-0/sref56http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.elsevier.com/S0261-2194(15)30074-0/sref58http://refhub.elsevier.com/S0261-2194(15)30074-0/sref58http://refhub.elsevier.com/S0261-2194(15)30074-0/sref58http://refhub.elsevier.com/S0261-2194(15)30074-0/sref58http://refhub.elsevier.com/S0261-2194(15)30074-0/sref59http://refhub.elsevier.com/S0261-2194(15)30074-0/sref59http://refhub.elsevier.com/S0261-2194(15)30074-0/sref59http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref62http://refhub.elsevier.com/S0261-2194(15)30074-0/sref62http://refhub.elsevier.com/S0261-2194(15)30074-0/sref62http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref64http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref63http://refhub.elsevier.com/S0261-2194(15)30074-0/sref62http://refhub.elsevier.com/S0261-2194(15)30074-0/sref62http://refhub.elsevier.com/S0261-2194(15)30074-0/sref62http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref61http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref60http://refhub.elsevier.com/S0261-2194(15)30074-0/sref59http://refhub.elsevier.com/S0261-2194(15)30074-0/sref59http://refhub.elsevier.com/S0261-2194(15)30074-0/sref59http://refhub.elsevier.com/S0261-2194(15)30074-0/sref58http://refhub.elsevier.com/S0261-2194(15)30074-0/sref58http://refhub.elsevier.com/S0261-2194(15)30074-0/sref58http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.elsevier.com/S0261-2194(15)30074-0/sref57http://refhub.els