CITOQUININAS HPLC

  • Upload
    nadiarh

  • View
    219

  • Download
    0

Embed Size (px)

Citation preview

  • 8/13/2019 CITOQUININAS HPLC

    1/13

    Analytical methods for cytokininsPetr Tarkowski, Liya Ge, Jean Wan Hong Yong, Swee Ngin Tan

    The development of sensitive analytical methods for the determination of

    cytokinin levels in plant tissues is essential for elucidating the roles of

    cytokinins in life science. In the past decade, more advanced analytical

    systems have been applied for the rapid analyses of endogenous cytokinins.

    This review primarily focuses on emerging analytical methods designed to

    meet the requirements for cytokinin analyses in complex matrices with

    special emphasis on high-performance liquid chromatography, gas chroma-

    tography and capillary electrophoresis coupled with different detectors. We

    highlight the advantages and the limitations of the techniques. As sample

    preparation is a key factor in determining the success of cytokinin analyses,

    we devote a section to discussing sample-extraction and clean-up techniques

    prior to analysis.

    2008 Published by Elsevier Ltd.

    Keywords: Analysis Capillary electrophoresis; Cytokinin; Complex matrix; Gas chroma-

    tography; Liquid chromatography; Mass spectrometry; Immunoassay; Sample preparation

    1. Introduction

    Cytokinins are a group of plant hormones

    [1,2]. Certain cytokinins (e.g., kinetin and

    zeatin) show significant anti-ageing, anti-

    carcinogenic, and anti-thrombotic effects

    [3,4]. Rapid analyses of cytokinins are

    therefore of great importance to plant

    physiologists and scientists from other

    disciplines, especially in view of their

    potential role in suppressing mammalian

    tumor growth.

    Chemically, most naturally occurring

    cytokinins are adenine derivatives and can

    be classified by the configuration of their

    N6-side chain as either isoprenoid or aro-

    matic (Table 1). Since plant-tissue extracts

    are rather complex multi-component

    mixtures, and cytokinins typically occur intrace quantities (usually at levels below

    30 pmol/g of fresh weight [1,2]), suffi-

    ciently sensitive and selective analytical

    tools are required for determination of

    their endogenous levels. Regardless of the

    analytical method used, highly purified

    plant samples are preferred for the analy-

    ses[5].

    Traditionally, quantification of cyto-

    kinins has been performed using bioassays

    and immunoassays. Although bioassays

    are essential for the isolation of novel

    compounds, they are time-consuming and

    not very accurate. Immunoassay tech-

    niques can be used as a sensitive, viable

    alternative for determination of cytokinin

    levels[6]. The two most common forms of

    cytokinin immunoassay are radioimmu-

    noassay (RIA) [7] and enzyme-linked

    immunosorbent assay (ELISA) [8]. How-

    ever, scintillation proximity assay (SPA) is

    also gaining ground [9,10]. Recently,

    ELISA has overtaken RIA. In addition, by

    taking advantage of the highly specific

    structural requirements of antibodies (Abs)

    for binding, the detection of cytokinins in

    plant tissues by immunolocalization offers

    a powerful tool to study the distribution of

    these signaling molecules[11].

    Despite widespread applications, immu-

    noassays sometimes suffer from problems

    (e.g., cross-reactivity, imperfect validation,

    and variable results for plant sample anal-

    yses). Other common analytical methods

    are physico-chemical techniques {gas chromatography (GC) [12], high-performance

    liquid chromatography (HPLC) [13,14]

    and capillary electrophoresis (CE)[15]}.

    For biological samples, HPLC is still the

    major tool in analyses and purification of

    cytokinins.

    As a complementary method, CE is also

    currently available for cytokinin analyses.

    In some cases, CE can have distinct

    advantages over HPLC in terms of sim-

    plicity, rapid method development, and

    reduced cost of the operation, since packed

    columns, reciprocating pumps and mobile-

    phase gradient are not used[16,17].

    Recent advances in instrumentation

    [e.g., ultra-performance liquid chroma-

    tography (UPLC)] and the range of

    detectors available have enabled analytical

    scientists to measure and to identify

    cytokinins at trace concentrations[18,19].

    This review summarizes the current

    key methods for cytokinin analyses as

    follows:

    Petr Tarkowski

    Department of Biochemistry,

    Faculty of Science;and,

    Laboratory of Growth

    Regulators, Institute of

    Experimental Botany ASCR

    Palacky University, Slechtitelu

    11, Olomouc, CZ-783 71

    Czech Republic

    Liya Ge,

    Jean Wan Hong Yong,

    Swee Ngin Tan*

    Natural Sciences and Science

    Education Academic Group,

    Nanyang Technological

    University, 1 Nanyang Walk,

    Singapore 637616

    *Corresponding author.

    Fax: +65 68969432;

    E-mail:

    [email protected]

    Trends in Analytical Chemistry, Vol. 28, No. 3, 2009 Trends

    0165-9936/$ - see front matter 2008 Published by Elsevier Ltd. doi:10.1016/j.trac.2008.11.010 3230165-9936/$ - see front matter 2008 Published by Elsevier Ltd. doi:10.1016/j.trac.2008.11.010 323

    mailto:%[email protected]:%[email protected]
  • 8/13/2019 CITOQUININAS HPLC

    2/13

    Table 1. Structures, names and abbreviations of cytokinins

    N

    NN

    N

    HN R1

    R2

    1

    2

    3

    49

    8

    7

    56

    R1 R2 R3 Compound Abbreviation

    H2C CH3

    CH2OR3H H trans-zeatin ZR H trans-zeatin riboside ZRG H trans-zeatin 9-glucoside Z9G H trans-zeatin 7-glucoside* Z7GH G trans-zeatin O-glucoside ZOGR G trans-zeatin riboside O-glucoside ZROG

    RP H trans-zeatin riboside-5 0-monophosphate ZMP

    CH3

    CH2OR3H2C

    H H cis-zeatin cZR H cis-zeatin riboside cZRRP H cis-zeatin riboside-5 0-monophosphate cZMP

    H2C CH3

    CH2OR3

    H H dihydrozeatin DZR H dihydrozeatin riboside DZRG H dihydrozeatin9-glucoside DZ9GH G dihydrozeatinO-glucoside DZOGR G dihydrozeatin riboside O-glucoside DZROGRP H dihydrozeatin riboside-50-monophosphate DZMP

    H2C CH3

    CH3

    H isopentenyladenine iPR isopentenyladenine riboside iPRG Isopentenyladenine 9-glucoside iP9GRP isopentenyladenine riboside-50-monophosphate iPMP

    H2C H benzylaminopurine BAR benzylaminopurine riboside BARG benzylaminopurine 9-glucoside BA9GRP benzylaminopurine-5 0-monophosphate BAMP

    H2C

    HO

    H ortho-topolin oTR ortho-topolin riboside oTRG ortho-topolin9-glucoside oT9G

    H2C

    OH

    H meta-topolin mTR meta-topolin riboside mTRG meta-topolin 9-glucoside mT9G

    H2C OH H para-topolin pT

    R para-topolin riboside pTRG para-topolin 9-glucoside pT9G

    H2C

    O

    H kinetin KR kinetin riboside KRRP kinetin riboside-50-monophosphate KMP

    H, Hydrogen; R, b-D-ribofuranosyl; RP, b-D-ribofuranosyl-5 0-monophosphate; G, b-D-glucopyranosyl.*In Z7G, b-D-glucopyranosyl group is substituted at N7.

    Trends Trends in Analytical Chemistry, Vol. 28, No. 3, 2009

    324 http://www.elsevier.com/locate/trac

  • 8/13/2019 CITOQUININAS HPLC

    3/13

    sample preparation (Section 2);

    techniques for analysis, including GC, HPLC,

    UPLC, CE and immunoassay (Section 3); and,

    concluding remarks and the future prospects

    (Section 4).

    2. Sample preparation

    Sample preparation is a key procedure in any modern

    chemical analyses, especially for analytes present at

    trace or ultra-trace levels in complex matrices. The

    procedures used for cytokinin extraction and sample

    preparation are therefore critically important steps in the

    entire analytical process. Fig. 1 shows the comprehen-

    sive procedures used for extraction, pre-concentration

    and purification of cytokinins.

    2.1. Extraction techniques

    The qualitative and the quantitative aspects of cyto-

    kinins extracted from plant tissues vary with the types of

    extraction solvents and procedures employed [20]. In

    order to prevent any enzymatic degradation of cyto-

    kinins (e.g., conversion of cytokinin nucleotide to its

    riboside), plant material should be immediately frozen or

    extracted with proper solvent(s) after harvesting. Isola-

    tion and identification of picomolar quantities of cyto-

    kinins are often hampered by the other biological

    materials present in excess in the sample.

    Typically, plant tissue is frozen in liquid nitrogen and

    left in Bieleskis solvent, comprising methanol/chloro-

    form/water/formic acid (12/5/2/1, v/v/v/v)[21]. Hoye-rova et al. [22] compared the extraction efficiency of

    three different extraction solvents: (a) 80% (v/v) metha-

    nol; (b) Bieleskis solvent; and, (c) modified Bieleskis

    solvent (methanol/water/formic acid; 15/4/1, v/v/v). It

    was found that the modified Bieleskis solvent sufficiently

    suppressed the dephosphorylation of cytokinin mono-

    nucleotides and gave the highest responses for deuterated

    cytokinins (used as test compounds) in plant extracts.

    2.2. Liquid-liquid partition

    During liquid-liquid partitioning, the plant material is

    homogenized in liquid nitrogen, and cytokinins are ex-tracted with cold acetone. After centrifugation, the

    supernatant is evaporated in vacuo to dryness. The

    residue is dissolved in lukewarm (38C) acidified water

    (pH 3.5). A triple extraction of an aqueous solution of

    cytokinins with butanol saturated with acidified water

    removes chlorophyll and other impurities, along with

    small traces of cytokinins[23].

    2.3. Solid-phase extraction

    Solid-Phase Extraction (SPE) has been widely applied for

    purification, extraction and isolation of many com-

    pounds [16,2426]. One distinct advantage of SPE is

    that high extraction recovery can usually be obtained

    with a suitable sorbent and operating procedure, even

    under situations when other traditional extraction

    techniques have failed.

    Pre-concentration of cytokinins has commonly been

    achieved using SPE with C18cartridges. A more effectivebut more complex approach is to purify plant extracts by

    passing them through linked columns of polyvinylpoly-

    pyrrolidone powder, DEAEcellulose or DEAE-Sephacel,

    and C18-SPE [5]. Fast, efficient separation of cytokinins

    has been achieved using mixed-mode SPE with both

    reversed-phase (RP) and ion-exchange characteristics

    [24].

    After C18 SPE cartridges were used as a pre-concen-

    tration tool, further sample purification could be carried

    out using mixed-mode cation exchanger (MCX) SPE

    cartridges with good recoveries [16]. Purification of

    cytokinins using MCX SPE, as compared to DEAE

    Sephadex and C18SPE method, was suitable for removalof Ultraviolet (UV) absorbing contaminants with higher

    recoveries of cytokinins [22].

    However, due to the relatively high polarity of cyto-

    kinin nucleotides leading to poor retention by C18cartridges during SPE, the use of an anion-exchange

    sorbent in addition is an efficient alternative step to

    separate cytokinin nucleotides from cytokinin bases

    and sugar conjugates [25]. An efficient dual-step SPE

    method has therefore been developed for pre-concen-

    tration and purification of cytokinin nucleotides using

    Oasis HLB and MAX cartridges [26].

    2.4. Immunoaffinity purification

    Immunoaffinity purification methods, based on anti-

    body-antigen (Ab-Ag) interactions, can provide selective

    sample enrichment, and thus greatly enhance limits of

    detection (LODs) of trace analyses [27], so, when an

    immunoaffinity approach is used as the purification step

    before final analysis, highly purified cytokinin prepara-

    tions containing only traces of other UV-absorbing

    material can be obtained.

    Several papers have reported on the immunoextrac-

    tion of cytokinins using monoclonal and polyclonal

    antibodies (mAbs and pAbs) [27,28]. The set-up for asuitable affinity system requires an appropriate Ab, and

    consideration of matrix factors that are not specific to

    phytohormone analysis [28]. Generic cytokinin mAbs

    are frequently used in this respect [13,27]. Immunoaf-

    finity columns purify analytes according to structural

    similarities, so they hold promise for the discovery of new

    cytokinins [28]. Immunoextraction has higher selectiv-

    ity than conventional SPE, but low throughput. An

    efficient, off-line, batch-immunoextraction method was

    developed for the purification of new cytokinins and

    their ribosides[27].

    Trends in Analytical Chemistry, Vol. 28, No. 3, 2009 Trends

    http://www.elsevier.com/locate/trac 325

  • 8/13/2019 CITOQUININAS HPLC

    4/13

    Octadecyl C18Cartridge

    Wash by acidified

    water (pH 3.0)

    Elution by ethanol;

    water: acetic acid(80:20:1)

    Flow through

    (Some CKnucleotides may be

    lost here)

    Elution by 0.35 M

    NH4OH in 60%

    CH3OH

    CK bases,

    ribosides,

    glucosides

    Oasis MCX Cartridge

    Evaporate and

    redissolve 1 M HCOOH

    Elution by 0.35 M

    NH4OHCK nucleotides

    (not full complement)

    Wash by CH3OH

    IAA, ABA

    Oasis H

    Elut

    Eva

    redissolv

    Oasis M

    Wash by 1 M

    NH4OH

    Extraction (Bieleskis solution or

    modified Bieleskis solution)

    -20C

    PolyclarVT Cartridge

    DEAE-Sephacel Cartridge

    Plant materials

    Flow-through fraction

    Flow-through fraction

    CK bases, ribosides,glucosides

    Elution by 1 M

    NH4HCO3

    CK nucleotides

    Immuno-affinity Column Chromatography

    CK

    mononucleotides

    CK

    dinucleotides

    CK

    trinucleotides

    Dephosphorylation by treatment with phosphatase

    Hydrolysis glucosides

    by -glucosidaseMonoQ Column

    Remove CH3OH, pH adjust to 1.8

    4C

    Remove CH3OH, pH adjust to 3.025C

    W

    N

    Figure 1. Procedures used for extraction, pre-concentration and purification of cytokinins.

    326

    http://www.elsevier.

    com/locate/trac

  • 8/13/2019 CITOQUININAS HPLC

    5/13

    3. Techniques for analyses of cytokinins

    3.1. Gas Chromatography

    GC-based methods provide high resolution and low

    LODs, but they are labor-intensive and costly. GC has

    been used for the analyses of cytokinins since the early

    1970s [20,29]. As cytokinins are not volatile com-pounds, they have to be derivatized to increase their

    volatility and also to improve their thermal stability prior

    to GC analysis. Derivatization methods have some

    inherent technical problems (e.g., hydrolysis of the

    derivatives, multiple product formation, and limited

    volatility[30,31]). However, GC with mass spectrometry

    (GC-MS) was a reliable, specific means for identification

    and quantification of cytokinins, until the recent intro-

    duction of using a combination of HPLC-MS. Structures

    of almost all naturally-occurring cytokinins were eluci-

    dated by GC-MS before the 1990s [20,29].

    3.1.1. Derivatization methods. Derivatization procedures

    for GC-MS analyses have been well described: silylation

    (trimethylsilyl) with N-methyl-n(trimethylsilyl)trifluoro-

    acetamide in 50% pyridine containing 1% trimethyl-cholorsilane for hours at room temperature, or a shorter

    time at 80C. This approach gives good results with an

    LOD of about 10 lg of anhydrous cytokinins [29].

    Complications have been associated with derivatiza-

    tion of cytokinins for GC-MS analysis using trimethylsilyl

    [20,32], permethyl [33], t-butyldimethylsilyl [34],

    trifluoroacetyl [35] and acetyl [12] derivatives. Penta-

    fluorobenzyl derivatives of cytokinin free bases for

    negative-ion MS were reported by Hocart et al. [36]. A

    Figure 2. HPLC chromatograms of standard mixtures of (A) 20 and (B) 18 cytokinins (Adapted from [13,39], with permission).

    Trends in Analytical Chemistry, Vol. 28, No. 3, 2009 Trends

    http://www.elsevier.com/locate/trac 327

  • 8/13/2019 CITOQUININAS HPLC

    6/13

  • 8/13/2019 CITOQUININAS HPLC

    7/13

    novel method based on N-methyl-N-(tert-butyldimethyl-

    silyl)trifluoroacetamide as a derivatization reagent in a

    comprehensive chemical derivatization protocol for the

    GC-MS analysis of cytokinins and other phytohormones

    has also been reported [37].

    3.1.2. Chromatographic conditions and detection. The

    separation conditions have not changed much since the

    1970s, although currently fused-silica capillary columns

    are used instead of packed glass columns. The separation

    power of capillary GC and the selectivity of MS detection

    make GC-MS a powerful technique for cytokinin

    analyses[29]. The main advantage of using the GC-MS-

    based identification, compared with the other soft-ioni-

    zation techniques, is differentiation of the various sugar

    moieties. Besides MS detection, the use of two other

    detectors, namely flame-ionization detector (FID) and

    electron-capture detector (ECD), has been reported

    [12,33]. The main drawback of FID is limited sensitivitythat makes it inapplicable in the trace analysis of cyto-

    kinins. Even though ECD is quite sensitive, halogenated

    derivatives are required in order to obtain satisfactory

    LODs.

    3.2. High-performance liquid chromatography

    HPLC is a particularly suitable chromatographic tech-

    nique for cytokinin analyses, as cytokinins exhibit gra-

    dations in polarity and are readily detected by UV

    absorbance [38]. HPLC enables rapid, high-resolution

    purification of cytokinins from plant extracts prior to

    analysis by MS, immunoassay, or bioassay. HPLC anal-

    ysis alone can also provide reliable identification of

    cytokinins in plant extracts. However, as absorbance at a

    single UV wavelength is inadequate for this purpose, the

    most widely used procedure for the quantifying cyto-

    kinins is isotope-dilution MS, especially with LC-Elec-

    trospray Ionization-MS (LC-ESI-MS)[13,14].

    3.2.1. Chromatographic conditions and separation. Cyto-

    kinin-free bases and their sugar conjugates are relatively

    hydrophobic compounds that behave like weak bases, so

    they are well separated on RP columns under acidic

    conditions [20]. However, the more ionic cytokinin

    nucleotides are not so well separated by RP-HPLC.Typically, the nucleotides are converted to ribosides with

    alkaline phosphatase [14], or derivatized [25] to lower

    their polarity, before RP-HPLC separation.

    Nowadays, separation and analyses of cytokinins by

    HPLC are carried out using RP-C18 or C8 columns

    [13,39,40]. The volatile eluent additives (e.g., acetic/

    formic acid and their ammonium salts) are usually

    added to solvents containing aqueous methanol/

    acetonitrile [39,40]. To achieve good separation, gradi-

    ent elution by increasing content of organic modifier is

    often used [3941]. For preparative purification ofPropionylatedoTOG,

    2MeSoTOG

    andBA9G

    Chenopodium

    rubrumcells

    SCXSPEandC

    18

    SPE.

    Alkalinephosphatase

    treatmentofnucleotides.

    CapillaryLCcolumn

    (150

    0.3mmp

    ackedwith

    4lm

    Symmetry

    ODS

    packingmaterial)

    50%

    aqueousACN,1%

    glycerol,1%

    formicacid

    05min,20

    lL/min;

    545min,4

    .5lL/min

    Isocratic

    frit-FAB

    D

    oublefocusing

    m

    agneticsector

    [47]

    pT,mT,pTR,oT,mTR,BA,

    MeoT,MemT,oTR,BAR,MeoTR

    andMemTR

    Arabidopsistha

    liana

    and

    Populus

    canadensis

    leaves

    Octadecyl-silicaSPE,

    DEAE-SephadexSPE,C18

    SPE,andIACAlkaline

    phosphatasetreatmentof

    nucleotides

    SymmetryC

    18column

    (3.5lm,

    150mm

    2.1mm)

    A:MeOH;B:0.1%

    formic

    acidadjustedtopH2.9

    withammonium

    0.25mL/min

    (25%

    effluent

    wasintroducedintoESI

    source)

    Gradient05min,

    3015%

    A;525min,

    1540%

    A;

    2530min,40%

    A

    ESI

    Si

    nglequadrupole

    [48]

    PropionylatedpT,mT,pTR,oT,

    mTR,BA,MeoT,MemT,oTR,

    BARandMeoTR

    CapillaryLCco

    lumn

    (150

    0.3mm

    packedwith4lm

    SymmetryODS

    packingmateria

    l)

    55%

    aqueousACN,1%

    glycerol,1%

    formicacid

    05min,20lL/m

    in;

    565min,4.0lL

    /min

    Isocratic

    frit-FAB

    Doublefocusing

    magneticsector

    Z,ZR,Z7G,Z9G,DZ,DZR,iP,

    iPR,iP9G,ZOG

    andZROG

    Macadamia

    integrifolia

    C18

    SPEAlkaline

    phosphatasetreatmentof

    nucleotides

    C18

    column(3l

    m,

    20mm

    2.1mm

    )Guard

    column(C18,5lm,

    4mm

    2.0mm)

    A:10mM

    ammonium

    acetate;B:350mLMeOH,

    100mLACN,50mL,

    10mM

    ammonium

    acetate

    0.1mL/min

    Gradient03min,

    510%

    B;327min,

    1043%

    B;

    2730min,4380%

    B;3033min,

    80100%

    B

    API

    Q

    -TOF

    [49]

    2MeSoTOG,6-[2-(

    b-D-glucopyranosyloxy)benzylam

    ino]-2-methylthiopurine;MeoT,ortho-methoxytopolin;M

    emT,meta-methoxytopolin.

    Trends in Analytical Chemistry, Vol. 28, No. 3, 2009 Trends

    http://www.elsevier.com/locate/trac 329

  • 8/13/2019 CITOQUININAS HPLC

    8/13

    Table 3. Optimal separation conditions for cytokinins using different CE approaches[15,53]

    Analytes Sample matrix Samplepreparation

    Mode Buffer Capillarydimensions

    Separationvoltage

    In

    iP, iPR, Z, ZR,DZ,DZR, BA andBAR.

    STD sugar beet SPE CZE 150 mMphosphoricacid, pH 1.8

    77 cm (effectivelength61 cm) 75 lm

    20 kV 10

    STD, tobacco MEKC 20 mM SDS,50 mM borate,pH 9.2

    10

    BA, BA9G, BAR,mTR, oTR, KR, ZR,DZR, iP and iPR.

    STD NS CD-modifiedCZE

    100 mMphosphate-Tris(pH 2.5) bufferwith 25 mM c-CD

    47 cm (effectivelength 40 cm)50 lm.

    20 kV N

    BA, K, and otherplant hormonesincluding ABA,IAA, NAA, GA and2,4-D.

    STD, transgenictobacco flower

    LLE MEKC 50 mM boratecontaining50 mM SDS,pH 8.0

    48.5 cm (40 cmeffectivelength) 50 lm.

    15 kV 55

    Z, DZ, ZOG,DZOG, mTR,iP and BA

    STD, coconutwater Dual-stepSPE MEKC Combinationof 10 mMphosphate and10 mM boratebuffercontaining50 mM SDS,pH 10.4

    57 cm (effectivelength 47 cm)76 lm.

    15 kV 5p0

    oT, mT, pT, oTR,mTR and pTR

    STD, coconutwater

    Dual-stepSPE

    MEKC 20 mM boricacid and50 mM SDS,pH 8.0, with anextra 20% (v/v)MeOH added

    60 cm (effectivelength 50 cm)76 lm.

    15 kV 5p0

    K and KR STD, coconutwater

    Dual-stepSPE

    CZE 100 mMammoniumphosphatebuffer, pH 2.5

    40 cm (effectivelength 30 cm)76 lm

    15 kV 5p0

    330

    http://www.elsevier.

    com/locate/trac

  • 8/13/2019 CITOQUININAS HPLC

    9/13

    iP, DZ, Z, BA, K, oT,DZOG, ZOG, DZR,ZR, oTR and KR

    STD, coconutwater

    Dual-stepSPE

    CZE 25 mMammoniumformate/formicacid buffer(pH 3.4) and3% ACN (v/v)

    65 cm 50 lm 25 kV Osastin

    DZMP, iPMP,cZMP, ZMP, BAMPand KMP

    STD, coconutwater

    dual-stepSPE

    CZE 25 mMammoniumformate/formicacid buffer(pH 3.8) and2% MeOH (v/v)

    57 cm 50 lm Gradientseparationvoltage (25 kVfor 32 min, andthen lineargradient to30 kV in 5 min,finally 30 kV toend ofseparation)

    Osastin

    oT, mT, pT, oTR,mTR, pTR, oT9G,mT9G, pT9G, ZR,cZR, Z and cZ

    STD, bananapulp

    Dual-stepSPE

    Partialfilling-MEKC

    50 mMammoniumformate/ammoniumhydroxide atpH 9.0.

    Micellarsolution with70 mM ALSand 10%MeOH wasinjected for90 s at 50 mbarbefore thesample and120 s at50 mbar afterthe sample

    100 cm 50 lm 20 kV Osastin

    STD, Standard; NS, not stated; GA, Gibberellic acid; ABA, Abscisic acid; IAA, Indole-3-acetic acid; NAA, a-naphthalene acetic acid; 2,4-D, 2,4-di

    http://www

    .elsevier.com/locate/trac

    331

  • 8/13/2019 CITOQUININAS HPLC

    10/13

    cytokinins, a column size of 150 10 mm i.d. can be a

    good compromise between cost and sample-loading

    capacity [20]. HPLC columns with i.d. ranging from

    conventional (4.6 mm), narrow-bore (2.1 mm), micro

    (1 mm) to capillary columns (0.3 mm)[3941]are used

    for cytokinin analyses.Fig. 2shows representative HPLC

    chromatograms of cytokinins.As the best performance of early ESI interfaces was at

    flow rates of around 0.55 lL/min[42], the process was

    compatible with chromatographic miniaturization, so

    narrow-bore LC coupled to MS was used as a relatively

    new method for the analyses of cytokinins [13].

    Coupling capillary LC to ESI-MS significantly improves

    mass sensitivity[43]. However, it needs to be taken into

    account that down-scaling decreases injection volume,

    loading capacity and dynamic range. The problem of

    restricted injection volume, which is critical in analyzing

    biological samples, could be overcome by using both on-

    pre-column and on-chromatographic column trace

    enrichment. Sample loading is performed undernon-eluting conditions using a pre-column or directly

    (analytical column), which is followed by backflush

    (pre-column) or direct gradient elution (analytical

    column). Prinsen et al. [44] comprehensively compared

    important analytical parameters (sensitivity, linearity

    range, robustness, sample throughput) of conventional,

    micro and capillary LC combined with tandem MS (MS2)

    detection.

    3.2.2. Detection. As cytokinins exhibit strong UV

    absorbances between 220 nm and 300 nm, UV detection

    is suitable for their quantification [20]. Coincidentally,

    the UV-visible (UV-VIS) absorbance detector is the most

    widely used detector for HPLC, so HPLC-UV is widely

    used to separate and detect cytokinins. For example,

    HPLC fractions collected after chromatographic separa-

    tion are analyzed by bioassays, immunoassays or con-

    verted to volatile derivatives for GC analysis. However,

    using this non-specific UV-absorbance method for

    detection requires significantly higher amounts of sam-

    ple that needs extensive purification.

    The LC-MS approach offers a new tool to detect,

    quantify and characterize cytokinins in plant-tissue

    extracts at biologically meaningful levels. Furtherimprovement in LC systems as well as mass analyzers

    may overcome the low detectability of cytokinins.

    Different ionization techniques were used for MS analy-

    ses of cytokinins in combination with RP-HPLC,

    including thermospray (TS) [45], fast atom bombard-

    ment (FAB) [4648], atmospheric pressure ionization

    (API) [19,49], and ESI [13,14,18,19,43,44,48,50].

    Although use of frit-FAB MS has been reported [46

    48], ESI-MS is currently the most common LC-MS

    method in cytokinin analyses. Compared to frit-FAB LC-

    MS, ESI-MS has a fairly high sensitivity and is associated

    with lower background [13,14,18,19,43,44,48,50]. In

    1997, the first application of LC-ESI-MS2 with multiple

    reaction monitoring (MRM) for cytokinin determination

    was reported as a fast method for the quantification of

    16 different cytokinins with an LOD of 1 pmol [50].

    Subsequently, improved gradient elution together with a

    capillary column provided an LOD at low-fmol levels[44].

    It is obvious that the main advantage of the LC-MS

    approach over that of GC-MS is elimination of the

    derivatization step. However, in order to increase

    sensitivity, pre-column derivatization for LC-MS cyto-

    kinin analyses was used to give stronger quasi-

    molecular ion currents and to obtain more spectral

    information [14,43,4648]. Table 2 presents a sum-

    mary of some representative LC-MS methods for cyto-

    kinin analyses.

    3.3. Ultra-performance liquid chromatographyUPLC instrumentation can provide liquid flow at pres-

    sures of up to 1000 bar, and features columns packed

    with 1.7-lm particulate packing materials, so UPLC

    extends beyond the chromatographic limits of conven-

    tional HPLC instrumentation. UPLC can achieve higher

    resolutions, lower sensitivities and more rapid separa-

    tions [18,19].

    UPLC-MS provides significant advantages concerning

    selectivity, sensitivity and speed, and is undoubtedly a

    suitable system for the study of cytokinins [18,19,51].

    Schwartzenberg et al. successfully applied an efficient

    UPLC-MS2 method to establish the profile of 40 cyto-

    kinins found within bryophyte Physcomitrella patens[18]. Dolezal et al. also applied UPLC-MS2 to isolate new

    cytotoxic members of aromatic cytokinins present

    endogenously in extracts of Arabidopsis thaliana and

    Agrobacterium tumefaciens [19]. Recently, Novak et al.

    developed an efficient UPLC-MS2 method for cytokinin

    profiling in plant tissues, which is almost four-fold faster

    than the standard HPLC analysis[51].

    3.4. Capillary electrophoresis

    CE is particularly suitable for the analyses of cytokinins,

    due to its speed, high resolving power, and minimal

    requirements for sample and buffer [1517]. Althoughsome successful examples have been reported, the LOD of

    CE is somewhat higher that the LODs of HPLC and GC,

    which is a consequence of lower amounts of sample in-

    jected during analysis and also in having a shorter

    optical path length, so most CE applications in cytokinin

    analyses require sensitivity to be enhanced by using

    more specific detection systems (e.g., MS) and on-line or

    off-line sample pre-concentration to increase sample-

    solute concentration [26,52]. Ge et al. [15] compre-

    hensively reviewed information pertaining to this aspect

    of cytokinin analyses.

    Trends Trends in Analytical Chemistry, Vol. 28, No. 3, 2009

    332 http://www.elsevier.com/locate/trac

  • 8/13/2019 CITOQUININAS HPLC

    11/13

  • 8/13/2019 CITOQUININAS HPLC

    12/13

    3.5. Immunoassays

    Prior to the introduction of hyphenated techniques

    (e.g., LC-MS or GC-MS), immunoassay techniques were

    the methods of choice for trace analysis of phytohor-

    mones due to their low LODs [610]. Several quanti-

    tative immunological methods were developed to detect

    cytokinins. Despite having some problems associatedwith immunological methods, a few reports have

    shown that Abs are useful tools to detect trace cyto-

    kinins in plant tissues, and even at the ultra-structural

    level of cells. Immunolocalization of endogenous cyto-

    kinins provides complementary insights into their

    involvement at the cellular level [11].

    Immunoassay techniques (e.g., RIA, ELISA and SPA)

    can be used as sensitive, viable options for the deter-

    mination of cytokinins [610]. RIAs are very sensitive

    methods, which can detect nmol or pmol of molecules

    and have provided useful information on the

    biochemical processes dealing with ligand-receptor

    systems [7,54]. Due to the strong cross-reactions, pAbsfor RIA cannot be used directly on individual

    cytokinins in crude plant extracts, so substances

    interfering with RIA should be removed from the ex-

    tracts [54].

    Compared with RIA, ELISA is less expensive and

    easier to set up; moreover, the problems associated

    with the disposal of radioactive waste can be avoided.

    For the detection of cytokinins, avidin-biotin amplified

    ELISA, immunoaffinity purification and immunocyto-

    chemical techniques have been developed [8,55].

    ELISA detects cytokinins in the fmol range.

    Hapten-homologous and hapten-heterologous competi-tive ELISAs were developed for detecting endogenous

    cytokinin levels in crude plant extracts without

    intense purification so they needed less plant extract

    [55]. ELISAs are still widely used for cytokinin anal-

    yses.

    SPA is a novel radioisotopic technique, applicable to

    assays involving ligand-Ab binding, which eliminates

    the need to separate free and bound ligand, and to use

    scintillation fluid as required in conventional RIA [9,10].

    First described by Wang et al. [9], SPA is a sensitive

    assay for the quantification of cytokinins as free bases at

    concentration of less than 0.02 ng. Yong et al. used the

    SPA method to measure the distribution of cytokinins in

    cotton leaves and xylem sap[10]. Unfortunately, SPA is

    not yet widely used by the research community for the

    routine analysis of cytokinins.

    A disadvantage of the cytokinin immunoassays,

    compared with LC-MS, is that they estimate the com-

    bined content of similar groups (free bases, ribosides, 9-

    glucosides and nucleotides) of cytokinins, due to their

    lower specificity, rather than specific cytokinins [610].

    However, the effective range and the sensitivity of

    immunoassays are similar to those reported for LC-MS.

    4. Conclusions and perspectives

    We have addressed recent trends in separation and

    determination of cytokinins. Since free cytokinins pres-

    ent in plants are at extremely low levels, we also

    summarized the comprehensive sample-preparation

    steps prior to cytokinin analyses.Each method that can be used for cytokinin analyses

    has its own sensitivity and selectivity. Most recently

    published papers on cytokinins were based on

    applications of GC, LC, and CE. Recent literature also

    indicated that MS2 combined with GC [12], LC[13,14]

    or CE [15] would provide more convincing and satis-

    factory results for cytokinin analyses in most cases.

    From these results, we conclude that MS has rapidly

    become a highly sensitive, selective tool for cytokinin

    analyses. Its use with GC, LC or CE ensures more

    reliable detection, identity confirmation and quantifi-

    cation of cytokinins, as well as screening for potentially

    novel cytokinins.As an established classical approach, immunoassay

    (especially ELISA) is still commonly used as a sensitive,

    viable method for the determination of endogenous

    cytokinins [611]. Furthermore, immuno-chemical

    staining using appropriate Abs appears to be the only

    means of getting information about endogenous cyto-

    kinin distribution at cellular and sub-cellular levels

    [11].

    We anticipate that LC-MS2 will continue to play an

    important role in cytokinin analyses in the near future.

    Next to excellent sensitivity, this technique can provide

    structural information based on the fragmentationpatterns. Unlike the well-established GC-MS method, LC-

    MS2 can analyze cytokinins without derivatization.

    The recently developed UPLC technique, using a

    combination of higher pressure and small diameter

    particles as column packing, could also be a useful, high-

    throughput approach for the routine analyses of cyto-

    kinins. However, to date, UPLC has been used only to a

    limited extent as a separation technique to analyze

    cytokinins[18,19,51].

    As the basic separation principles of CE differ from

    those of HPLC and the other chromatographic tech-

    niques, it could be an attractive complementary tech-

    nique in analyses of cytokinins. MS detection has been

    used in conjunction with CE to determine different

    cytokinins in biological matrices[15]. In order to screen

    numerous samples, on-line sample pretreatment (pre-

    concentration and removal of interfering substances),

    and CE separation in microchip formats require further

    development.

    We anticipate that the development of analytical

    methods will enable us to unravel some of the mysteries

    concerning cytokinins in plants as well as their beneficial

    effects in medicine[14].

    Trends Trends in Analytical Chemistry, Vol. 28, No. 3, 2009

    334 http://www.elsevier.com/locate/trac

  • 8/13/2019 CITOQUININAS HPLC

    13/13

    References[1] D.W.S. Mok, M.C. Mok, Annu. Rev. Plant Physiol. Plant Mol. Biol.

    52 (2001) 89.

    [2] H. Sakakibara, Annu. Rev. Plant Biol. 57 (2006) 431.

    [3] K. Vermeulen, M. Strnad, V. Krystof, L. Havlcek, A. Van der Aa,

    M. Lenjou, G. Nijs, I. Rodrigus, B. Stockman, H. Van Onckelen,

    D.R. Van Bockstaele, Z.N. Berneman, Leukemia 16 (2002) 299.

    [4] S.I.S. Rattan, L. Sodagam, Rejuvenation Res. 8 (2005) 46.

    [5] G.A. Tucker, J.A. Roberts (Editors), Plant Hormone Protocols,

    Humana Press Inc., Totowa, NJ, USA, 2000.

    [6] R.O. Morris, D.G. Blevins, J.T. Dietrich, R.C. Durley, S.B. Gelvin,

    J. Gray, N.G. Hommes, M. Kaminek, L.J. Mathews, R. Meilan,

    T.M. Reinbott, L. Sayavedrasoto, Aust. J. Plant Physiol. 20 (1993)

    621.

    [7] A. Grayling, D.E. Hanke, Phytochemistry 31 (1992) 1863.

    [8] R. Maldiney, B. Leroux, I. Sabbagh, B. Sotta, L. Sossountzov,

    E.A. Miginiac, J. Immunol. Methods 90 (1986) 151.

    [9] J. Wang, D.S. Letham, E. Taverner, J. Badenoch-Jones, C.H. Hocart,

    Physiol. Plant. 95 (1995) 91.

    [10] J.W.H. Yong, S.C. Wong, D.S. Letham, C.H. Hocart, G.D. Farquhar,

    Plant Physiol. 124 (2000) 767.

    [11] E. Casanova, A.E. Valdes, B. Fernandez, L. Moysset, M.I. Trillas,

    J. Plant Physiol. 161 (2004) 95.

    [12] P.O. Bjorkman, E. Tillberg, Phytochem. Anal. 7 (1996) 57.

    [13] O. Novak, P. Tarkowski, D. Tarkowska, K. Dolezal, R. Lenobel,

    M. Strnad, Anal. Chim. Acta 480 (2003) 207.

    [14] A. Nordstrom, P. Tarkowski, D. Tarkowska, K. Dolezal, C. Astot,

    G. Sandberg, T. Moritz, Anal. Chem. 76 (2004) 2869.

    [15] L. Ge, J.W.H. Yong, S.N. Tan, E.S. Ong, Electrophoresis 27 (2006)

    4779.

    [16] L. Ge, J.W.H. Yong, S.N. Tan, X.H. Yang, E.S. Ong, J. Chromatogr.,

    A 1048 (2004) 119.

    [17] L. Ge, J.W.H. Yong, S.N. Tan, X.H. Yang, E.S. Ong, Electrophoresis

    26 (2005) 1768.

    [18] K. Dolezal, I. Popa, E. Hauserova, L. Spchal, K. Chakrabarty,

    O. Novak, V. Krystof, J. Voller, J. Holub, M. Strnad, Bioorg.

    Med. Chem. 15 (2007) 3737.

    [19] K. von Schwartzenberg, M.F. Nunez, H. Blaschke, P.I. Dobrev,O. Novak, V. Motyka, M. Strnad, Plant Physiol. 145 (2007)

    786.

    [20] R. Horgan, I.M. Scott, in: L. Rivier, A. Crozier (Editors), Principles

    and Practice of Plant Hormone Analysis, Academic Press, London,

    UK, 1987, Chapter 5, p. 303.

    [21] R.L. Bieleski, Anal. Biochem. 9 (1964) 431.

    [22] K. Hoyerova, A. Gaudinova, J. Malbeck, P.I. Dobrev, T. Kocabek,

    B. Solcova, A. Travnckova, M. Kam nek, Phytochemistry 67

    (2006) 1151.

    [23] R. Horgan, Analytical procedures for cytokinins, in: J.R. Hillman

    (Editor), Isolation of Plant Growth Substances, Society for

    Experimental Biology, Seminar Series 4, Cambridge University

    Press, Cambridge, UK, 1978, p. 97.

    [24] P.I. Dobrev, M. Kamnek, J. Chromatogr., A 950 (2002) 21.

    [25] K. Takei, T. Yamaya, H. Sakakibara, J. Plant Res. 116 (2003)265.

    [26] L. Ge, J.W.H. Yong, S.N. Tan, X.H. Yang, E.S. Ong, J. Chromatogr.,

    A 1133 (2006) 322.

    [27] E. Hauserova, J. Swaczynova, K. Dolezal, R. Lenobel, I. Popa,

    M. Hajduch, D. Vydra, K. Fuksova, M. Strnad, J. Chromatogr.,

    A 1100 (2005) 116.

    [28] B. Nicander, U. Stahl, P.O. Bjorkman, E. Tillberg, Planta 189

    (1993) 312.

    [29] G. Teller, in: D.W.S. Mok, M.C. Mok (Editors), Cytokinins:

    Chemistry, Activity and Function, CRC Press, Boca Raton, FL,

    USA, 1994.

    [30] E.J. Trione, G.M. Banowetz, B.B. Krygier, J.M. Kathrein,

    L.A. Sayavedra-Soto, Anal. Biochem. 162 (1987) 301.

    [31] L.M.S. Palni, S.A.B. Tay,J.K. MacLeod, in: H.F.Linkens,J.F. Jackson

    (Editors), Modern Methods of Plant Analysis, New Series, Vol. 3,

    Springer-Verlag, Berlin, Germany, 1987.

    [32] B.H.Most, J.C.Williams, K.J.Parker,J. Chromatogr. 38 (1968) 136.

    [33] J.K. MacLeod, R.E. Summons, D.S. Letham, J. Org. Chem. 41

    (1976) 3959.

    [34] C.H. Hocart, O.C. Wong, D.S. Letham, S.A.B. Tay, J.K. MacLeod,

    Anal. Biochem. 153 (1986) 85.

    [35] M. Ludewig, K. Dorffling, W.A. Konig, J. Chromatogr., A 243

    (1982) 93.

    [36] C.H. Hocart, J. Wang, D.S. Letham, J. Chromatogr., A 811 (1998)

    246.

    [37] C. Birkemeyer, A. Kolasa, J. Kopka, J. Chromatogr., A 993 (2003)

    89.

    [38] C. Chen, in: H.F. Linskens, J.F. Jackson (Editors), High Perfor-

    mance Liquid Chromatography in Plant Sciences, Springer, Berlin,

    Germany, 1987, p. 23.

    [39] L. Ge, J.W.H. Yong, N.K. Goh, L.S. Chia, S.N. Tan, X.H. Yang,

    E.S. Ong, J. Chromatogr., B 829 (2005) 26.

    [40] J.A. Van Rhijn, H.H. Heskamp, E. Davelaar, W. Jordi, M.S. Leloux,

    U.A.T. Brinkman, J. Chromatogr., A 929 (2001) 31.

    [41] B. Fernandez, M. Centeno,I. Feito, R. Sanchez-Tames, A. Rodriguez,

    Phytochem. Anal. 6 (1995) 49.

    [42] J. Abian, A.J. Oosterkamp, E. Gelp, J. Mass Spectrom. 34 (1999)

    244.

    [43] W.A. Stirk, O. Novak, K. Vaclavkova, P. Tarkowski, M. Strnad,

    J. van Staden, Planta 227 (2008) 1279.

    [44] E. Prinsen, W. Van Dongen, E.L. Esmans, H.A. Van Onckelen,

    J. Chromatogr., A 826 (1998) 25.

    [45] E. Prinsen, P. Redig, M. Strnad, I. Gals, W. van Dongen, H. van

    Onckelen, in: K.M.A. Gartland, M. Davey (Editors), Methods in

    Molecular Biology, Agrobacterium Protocols, Humanae Press,

    New Jersey, USA, 1995, p. 245.

    [46] C. Astot, K. Dolezal, T. Moritz, G. Sandberg, J. Mass Spectrom. 33

    (1998) 892.

    [47] K.Dolezal,C.Astot, J. Hanus, J. Holub, W.Peters,E. Beck,M. Strnad,

    G. Sandberg, Plant Growth Regul. 36 (2002) 181.

    [48] D. Tarkowska, K. Dolezal, P. Tarkowski, C. Astot, J. Holub,

    K. Fuksova, T. Schmulling, G. Sandberg, M. Strnad, Physiol.

    Plant 117 (2003) 579.

    [49] A.T. Fletcher, J.C. Mader, J. Plant Growth Regul. 26 (2007) 351.

    [50] E. Prinsen, W. van Dongen, E. Esmans, H. van Onckelen, J. Mass

    Spectrom. 32 (1997) 12.

    [51] O. Novak, E. Hauserova, P. Amakorova, K. Dolezal, M. Strnad,

    Phytochemistry 69 (2008) 2214.

    [52] L. Ge, J.W.H. Yong, S.N. Tan, E.S. Ong, Electrophoresis 27 (2006)

    2171.

    [53] L. Ge, J.W.H. Yong, S.N. Tan, L. Hua, E.S. Ong, Electrophoresis 29

    (2008) 2024.

    [54] N.C. Cook, D.U. Bellstedt, G. Jacobs, Scientia Hort. 87 (2001) 53.

    [55] A. Szekacs, G. Hegedus, I. Tobias, M. Pogany, B. Barna, Anal.

    Chim. Acta 421 (2000) 135.

    Trends in Analytical Chemistry, Vol. 28, No. 3, 2009 Trends

    http://www elsevier com/locate/trac 335