液相色谱与质谱接口技术 - 范文中心

液相色谱与质谱接口技术

02/22

Image courtesy of Bruker Daltonik GmbH

Interfaces for LC–MS

Filip Lemière, Dept of Chemistry, University of Antwerp, Belgium.

Introduction

When searching for a suitable techniquefor the analysis of mixtures, often

containing unknowns and/or analytes inlow concentration, the combination ofliquid chromatography (LC, separation)with mass spectrometry (MS, sensitivityand structural information) appears to bean obvious choice. However, LC–MS is an“odd couple” (1). First, there is the natureof the analytes. LC is preferred over gaschromatography (GC) because of thehigher polarity and lower volatility of thesamples. One of the prerequisites for massspectrometric analysis is the formation ofvolatilized ions. Second, and a harderproblem to solve, is the necessary

elimination of the mobile phase. A flow-rate of 1 mL/min water is converted in1244 mL/min vapour at atmosphericpressure, which is far too much to behandled by the standard MS vacuum

systems. Third, salts and other additives ofthe mobile phase are often involatile (e.g.,phosphates, NaCl etc.).

Several coupling methods have beendeveloped to solve these problems.

Methods such as particle beam (PB) andmoving belt/wire (2) rely on removal of thesolvent prior to entering the MS. Couplingwith continuous flow fast atom

bombardment (cf-FAB) (3, 4) or directliquid introduction (DLI) (5) reduces theflow entering the MS using some kind ofsplitting device. A serious drawback of thelast approach is the reduction in sensitivitycaused by the split factor. The entire flow-rate, about 1 mL/min, used with a classic4.6 mm i.d. column is only tolerated bytechniques such as thermospray (TSP) andatmospheric pressure chemical ionization(APCI). Electrospray (ES) has a workingrange from nL/min to 0.2 mL/min, whichcan be extended to mL/min flows. Themost attractive difference with other

interfacing techniques is that the ES signalis dependent on the concentration ratherthan the amount injected. Thisphenomenon makes ES virtually

independent of flow-rate allowing the useof splitters to reduce the amount of eluentintroduced into the MS source and

opening the possibility of miniaturizing theLC technique to capillary (300 µm i.d., 4 µL/min flow-rate) and nano (75 µm i.d.,200–300 nL/min flow-rate) dimensions.The same features also allow coupling ofcapillary electrophoresis, which has a flow-rate in the nL/min range, with MS. A stranger in this series with most of itsapplications in ‘inorganic’ analysis is

inductively coupled plasma (ICP)–MS. Forthis reason a special section will bedevoted to this technique (See “The

Inductively Coupled Plasma Ion Source forLC–MS”). Most LC–MS interfaces andapplications are focused on the analysis ofintact molecules, often organic materials.However, biomolecule samples areanalysed using this technique (6).

Apart from being an inlet system for theMS, an LC–MS interface is also the couplingof a detector (MS) to a chromatograph.The choice of LC–MS interface strongly

influences the characteristics of the MS asa detector for LC. Therefore, we shouldkeep in mind what characteristics are idealfor an LC detector (Table 1).

The First LC–MS Interfaces

The first experiments to couple LC to MSdate back to the late 1960s. Thoughfascinating at the time of their

development, the earliest LC–MS interfacesare now almost obsolete. The introductionof techniques that allow delivery ofthermolabile biomolecules into the MSshow an exponential increase in the

number of publications employing LC–MS.However, in order to give a somewhat

wider overview older LC–MS interfaces willbriefly be described.

Direct Liquid Introduction

The first attempts to introduce a liquid intoan MS using the classic electron impactionization (EI)/chemical ionization (CI)source were based on the simple principlethat by minimizing the amount of liquid,the vacuum system would remove the

solvent leaving the analyte in the gas phasefor ionization. By using larger pumpsystems and differential pumping,

through two differentially pumped vacuumlocks. A heater in the ion source evaporatesthe sample from the belt allowing MSanalysis (Figure 2). Most moving-beltanalyses deal with volatile analytes usingCI/EI; however, less volatile molecules suchas nucleosides and nucleotides are analysedusing this system (9).

Thermospray

The TSP interface was developed by

M. Vestal and co–workers (10–13). A majoradvantage of TSP over other LC–MSinterfaces is its ability to handle the highflow-rates delivered by LC (up to 2 mL/min).As the name thermospray implies, heatingthe liquid flow leaving an LC systemcreates a spray of superheated mistcontaining small liquid droplets. Severaltechniques are developed to heat and

vaporize the effluent (12, 13); however, themost successful method involves directingthe liquid flow through an electrically

heated capillary (11), which can be directlyintroduced into the MS ion source. Thedroplets are further vaporized as theycollide against the walls of the heated ionsource. This ion source is equipped with amechanical pump line opposite to thespray in order to evacuate the excesssolvent vapour (Figure 3). The rapidheating and protective effects of thesolvent allow the analysis of non-volatilesamples without pyrolysis. The analyte ionsare sampled into the MS through asampling cone, if necessary aided by anapplied electric field (repeller oraccelerating electrode).

Ionization of the analytes in TSP occurs by means of several processeswherein two classes of ionization type can be distinguished: one without anexternal ionization source, so–called ‘real thermospray’ and one with anexternal ionization.

The real thermospray uses a volatile

Figure 1: Scheme of the DLI interface. 1 ϭconnection to LC column, 2 ϭdiaphragm 5 µmopening to MS, 3 ϭneedle valve, 4 ϭcooling region, 5 ϭto UV detector or waste.

Figure 2: Schematic showing the principal components of a moving-belt interface.

Figure 3: Thermospray interface. (a) configuration for ‘real-TSP-ionization’ (filament off) orexternal ionization (filament on). (b) configuration with discharge electrode for external ionization and repeller electrode. (Adapted from reference 14.)

Figure 4: Schematic showing the principal components of a particle beam or MAGIC interface.

Figure 5: Overview of a differentially pumped API source coupled to a mass spectrometer.

using a differentially pumped momentumseparator. The PB interface allows flow-rates from 0.1–0.5 mL/min. Mostanalytes that are amenable to

PB LC–MS can be analysed using GC–MSas well.

Atmospheric Pressure Ionization

The earliest LC–MS techniques (DLI, TSP, moving belt, PB), although

commercialized, were often difficult touse, had limited sensitivity and were notrobust; however, they were very useful forspecific applications. The overwhelmingincrease in LC–MS applications is mainlythe result of the sensitivity and

ruggedness of atmospheric pressure

ionization (API) LC–MS techniques. API isa general name for all ionization

techniques in which the ions are formedat atmospheric pressure. Though verypopular today, ionization processes atatmospheric pressure (flames, dischargesetc.) have been studied using massspectrometers for many years (18–20).In modern LC–MS applications we findtwo major techniques: ES and APCI.Electrospray can be subdivided into

techniques such as pneumatic-assisted ES,ES, multiple sprayer ES etc., that differmainly in the formation of a spray fromthe LC flow. However, all ES variants relyon the same mechanism(s) to form ionsfrom the droplets at atmospheric pressure.The ions formed at atmospheric pressureare transported from the source to thevacuum of the analyser through one ormore differentially pumped stages

separated by skimmers (Figure 5). The ionsare focused and guided through the

skimmer openings into the MS by applyingappropriate electric fields. Various sourcedesigns, ion optics configurations,

pumping systems and other experimentalparameters (21) are used, but the basicfeatures can be found in all instruments.Where ES has its optimal performance atlow flow-rates (nL/min range) APCI

operates happily using mL/min flow-rates.ES and APCI perform differently underdifferent chromatographic modes.

The advantages of API were summarizedby Voyksner (22) in four points:

1. “API approaches can handle volumesof liquid typically used in LC”

2. “API is suitable for the analysis of non-volatile, polar and thermally unstablecompounds typically analysed by LC”3. “API-MS systems are sensitive, offeringcomparable or better detection limitsthan achieved by GC–MS”

4. “API systems are very rugged and relatively easy to use.”

Figure 9: ES spectrum of the -chain of bovine haemoglobin.

biomolecules such as peptides, proteins,oligonucleotides etc., with molecular

weights of tens and hundred of thousandsamu. Unlike most ionization techniquesthat yield monocharged ions, ES of thesebiopolymers yields multiply charged ions.Because MS measures a mass-over-chargeration, these large masses can be

measured using standard MS equipmentwith a limited mass range (quadrupole4000 amu, TOF 20000 amu) (Figure 9).Although these multiple charge spectralook somewhat odd at first, molecularweight information can be obtained usingsome simple mathematics.

Unless special additives are used, mostmolecules including peptides and proteinsare charged by (de)protonation. Thisimplies that successive peaks (M 1/Z 1and M 2/Z 2) in the envelope of multiply chargedions differ by 1 charge unit and the massof 1 proton. Therefore, we know that forthese peaks the following holds:

M 1Z 1 ϭ M ϩ 1.0079*Z 1[1]M 2Z 2 ϭ M ϩ 1.0079*Z 2

[2]

where M is the mass of the unchargedmolecule. For the electric charges we knowthat

Z 2 ϭ Z1 ϩ 1

[3]

This allows us to calculate the chargestate of the ion at M 1/Z 1

Z 1 ϭ (M2 Ϫ 1.0079)/(M 2 Ϫ M 1)

[4]

Once we know the charge status themass of the multiply charged ion can becalculated and together with the chargestate (equals number of protons) themolecular weight of the analyte can beestablished.

Another highly uncommon characteristicof ES is its ‘softness’; that is, very labilestructures can be carried as ions into thegas phase without disrupting their

structures. ES can be used to study proteinfolding status, non-covalent bonding, DNAduplexes etc. (38). For the same reason ESspectra contain little or no structuralinformation because of the absence offragmentation. Molecular weightinformation is obtained in the first

instance. If more structure information isneeded, for example, sequence

information of peptides, fragmentationmust be induced. This is most convenientlydone by applying tandem MS. Startingfrom doubly charged peptides product ion

spectra can be obtained from which theamino acid sequence can be deduced(Figure 10).

Atmospheric Pressure ChemicalIonization

Using APCI the liquid flow from the LC issprayed and rapidly evaporated by a coaxialnitrogen stream and heating the nebulizerto high temperature (350–500 °C).

Although these temperatures may degradethe analytes, the high flow-rates and

coaxial N2-flow prevent breakdown of themolecules. Ions already present in solutioncan be carried into the gas phase,

however, additional ionization is achievedusing a corona discharge (3–6 kV) in thespray. This discharge can ionize not onlythe analyte molecules, but also the solventmolecules. These solvent ions can reactwith the analytes in the gas phase in thesame way samples are ionized in a CIsource by the reagent gas. In positive ionmode protonated molecules and adductsare formed; in negative ion mode ions areformed by deprotonation, combinationswith anions or electron-capture. Theionization efficiency is better comparedwith CI because it occurs at high pressure(atmospheric) so the collision frequency ishigh compared with the process in astandard CI source, and ionization

efficiency is higher. The different ionizationmodes can be exploited to further enhancethe sensitivity of the technique. One of themost sensitive GC–MS techniques iselectron capture negative ion CI. Theanalytes are derivatized with an electron-capturing group, generally a

pentafluorobenzyl derivative. Becauseelectrons are produced in the coronadischarge used in APCI it was anticipatedthat these derivatives would give goodsensitivity using APCI as well. For a series of biomolecules and drugs it isshown that one can obtain detection limits in the attomole (femtogram) range (41).

Unlike in ES, the solvent-evaporation andion-formation processes are separated inAPCI. This allows the use of solvents thatare unfavourable for ion formation. Theselow-polarity solvents are commonly used innormal-phase chromatography with lowpolarity samples that can generally beevaporated for APCI ionization. Anothermajor difference between APCI and ES canbe found in the LC flow-rates that areused. APCI is a technique with optimalperformance at high flow-rates (1 mL/minand higher). Lower flow-rates can be used;however, when flow-rates are too low thestability of the corona discharge may

Figure 10: ES product ion spectrum of a tryptic digest peptide Glu-Fibrino peptide B showingthe sequence ions.

Figure 11: Basic elements of a cf-FAB probe.

Future Developments

A very clear goal since the beginnings ofLC–MS and still an important trend in

newly developed instruments is robustness.Both separation science and mass

spectrometry are very specialized researchdomains and often scientists are focusedon only one of them. Thus, when applyingthe hyphenated LC–MS techniques theother half “just has to work”. The

chromatographer wants MS to work as areliable detector that can be hooked up toa column (no matter what flow-rate orkind of separation), whilst the massspectrometrist needs a system forintroducing these liquid samples

(sometimes mixtures) containing polar,thermolabile, involatile biomolecules,pharmaceuticals, environmentalcontaminants, pesticides etc.

As sample availability and sensitivity arealways an issue in analytical (bio)chemistry,miniaturization will be a continuing trendin LC–MS. Miniaturization of the

separation techniques and consequentdevelopment of the appropriate interfaceswill proceed, including chip-basedtechnology for both separation andinterfacing to MS. Techniques that aretoday still considered as off-line

techniques, for example, MALDI, 2D gelelectrophoresis etc., will be modified andnew techniques developed to couple withexisting MS and LC–MS systems (49, 50).References

(1)P .J. Arpino, Trends Anal. Chem., 1, 154–158,

(1982).

(2)R.P .W. Scott et al., J. Chromatogr., 99,

395–405, (1974).

(3)Y . Ito et al.,J. Chromatogr., 346, 161–166, (1985).(4)R.M. Caprioli, T. Fan and J.S. Cotrell,

Anal.Chem. , 58(14), 2949–2953, (1986).(5)G.V . Tal’roze et al., J. Phys. Chem., 42,

1658–1664, (1968).

(6)C. Siethoff et al., J. Mass Spectrom., 34,

412–426, (1999).

(7)L.R. Snyder and J.J Kirkland, Introduction to

Modern Liquid Chromatography, (Wiley, NewYork, USA, 1974).

(8)E.L. Esmans et al.,Biomed.Mass Spectrom.,

12(5), 241–245, (1985).

(9)D.E. Games, Biomed. Mass Spectrom., 8,

454–462, (1981).

(10)M.L. Vestal, Science , 226, 275–281, (1984).(11)M.L. Vestal and G.J Fergusson, Anal. Chem.,

557, 2373–2378, (1985).

(12)C.R. Blakley, M.J. McAdams and M.L. Vestal,

J. Chromatogr., 158, 261–276, (1978).(13)C.R. Blakley, J.J. Carmody and M.L. Vestal,

Anal. Chem., 52, 1636–1641, (1980).

(14)P . Arpino, Mass Spec. Rev., 9, 631–669, (1990).(15)F . Lemière et al., J. Chromatogr., 647(2),

211–218 (1993).

(16)K. Vanhoutte et al., J. Mass Spectrometry, 30,

1453–1461, (1995).

(17)R.C. Willoughby and R.F. Browner, Anal. Chem.,

56, 2625–2631, (1984).

(18)P .F . Knewstubb and S.B. Sugden, Proc. Roy. Sec.

A , 255, 520, (1966).

(19)M.M. Shahin, J. Chem. Phys., 45, 2600, (1966).

(20)E.C. Horning et al., J. Chromatogr. Sci., 12,

725, (1974).

(21)A.P . Bruins, TRAC, 13(1), (1994).

(22)R.D. Voyksner, in “Electrospray Ionization Mass

Spectrometry: Fundamentals, Instrumentation,and Applications”, Ed. R.B. Cole, (Wiley-Interscience, New York, USA, 1997).

(23)K. Tang and A. Gomez, J. Aerosol Sci., 25(6),

1237–1249, (1994).

(24)S. Zafran, J.C. Beynon and P.W. Kidd, Prototype

One–Millipound Colloid Thruster System, AIAA11th Electric Propulsion Conf., New Orleans,Louisiana, USA, Paper No. 75–392, (1975).(25)C.J. McNeal, R.D. Mcfarlane and E.L. Thurston,

Anal. Chem., 51, 2036, (1979).

(26)N.J. Felici, Recent Developments and Future

Trends in Electrostatic Generation., DirectCurrent, 4, 3–12, (1959).

(27)J.J. Thomson, Rays of Positive Electricity and

their Application to Chemical Analysis, (Longmans Green, London, UK, 1913).(28)J. Zeleny, Phys. Rev., 10, 1–6, (1917).

(29)A.P . Bruins, T.R. Covey and J.D. Henion, Anal.

Chem. , 59, 2642–2646, (1987).

(30)G.A. Clegg and M. Dole, Biopolymers , 10,

2240–2249, (1971).

(31)B.A. Thomson and J.V. Iribarne, J. Chem. Phys.,

71, 4451–4463, (1979).

(32)J.V . Iribarne, P.J. Dziedzic and B.A. Thomson,

Int. J. Mass Spectrom. Ion Phys., 50, 331–347,(1983).

(33)J.V . Iribarne and B.A. Thomson, J. Chem. Phys.,

64, 2287, (1976).

(34)M. Dole, L.L. Mack and R.L. Hines, J. Chem.

Phys. , 49(5), (1968).

(35)M. Yamashita and J.B. Fenn, J. Phys. Chem., 88,

4451–4459, (1984).

(36)M. Yamashita and J.B. Fenn, J. Phys. Chem., 88,

4671–4675, (1984).

(37)M.L. Aleskandrov et al., Dokl. Akad. Nauk.

SSSR , 277, 379–383, (1984).

(38)‘Mass Spectrometry of Biological Materials’, Ed.

B.S. Larsen and C.N. McEwen, (Marcel DekkerInc., New York, USA, 1998.)

(39)K. Vanhoutte et al., Anal.Chem. , 69,

3161–3168, (1997).

(40)C.G. Edmonds et al., J. Chromatogr., 474,

21–37, (1989).

(41)G. Singh et al., Anal.Chem. , 72, 3007–3013,

(2000).

(42)S. Perez and D. Barcelo, Chromatographia ,

53(9–10), 475–480, (2001).

(43)B.A. Ingelse et al., J. Chromatogr. A, 918(1),

67–78, (2001).

(44)M. Barber et al., J. Chem. Soc. Chem.

Commun. , 325, (1981).

(45)Y. Ito et al., J.Chromatogr. , 346, 161, (1985).(46)S.M. Wolf et al., J. Am. Soc. Mass Spectrom., 3,

757–761, (1992).

(47)L.J. Deterding et al., Anal.Chem. , 64(21),

2636–2641, (1992).

(48)M.J.F . Suter and R.M. Caprioli, J. Am. Soc. Mass

Spectrom. , 3, 198–206, (1992).

(49)J. Preisler et al.,Anal. Chem., 72, 4785–4795,

(2000).

(50)B. Zhang et al., Anal. Chem., 72, 1015, (2000).

Dr Filip Lemière works in the NucleosideResearch and Mass Spectrometry Unit,

Department of Chemistry at the University ofAntwerp, Belgium. His research interestsinclude the miniaturization of LC–MSprocedures particularly for the analysis ofDNA adducts.


相关内容

  • 超高效液相色谱_线性离子阱_静电_省略_质谱快速筛查化妆品中的24种激素_李兆永
    2014年5 月May 2014 Chinese Journal of Chromatography Vol.32N o.5 477-484 研究论文 DO I :10.3724/SP.J.1123.2013.12004 超高效液相色谱-线 ...
  • 高效液相色谱技术及其应用
    目录 1.高效液相色谱技术..................................................................................................... 1 1.1 ...
  • 环境激素物质监测分析方法的应用与研究
    第5期 环境激素物质监测分析方法的应用与研究欂欂氊 欂欂欂欂欂欂欂欂氊 欂欂氊 23 专题与评述 1 环境激素物质的种类 目前已知的环境激素约有7广泛存在于大0种,气.水.土壤等环境介质中.几乎所有的环境激素都 2] .环境激素物质无处不在 ...
  • 纺织品禁用偶氮染料的检测
    2007年第6期 纺织科技进展 ・7・ 纺织品禁用偶氮染料的检测 阿 阳 (国家服装质量监督检验中心(上海) , 上海200336) 摘 要:介绍了纺织品禁用偶氮染料的致癌性.检测标准.部分主要标准的要求, 详述了用气2质联用仪(GC 2M ...
  • 大蒜挥发油的分离提取工艺研究进展
    DOI:10.3969/j.issn.1001-8972.2011.14.048 大蒜挥发油的分离提取工艺研究进展 徐丽萍1 张亚非2 吴正勇1 1.四川化工职业技术学院,四川泸州6460052.重庆市北碚区农业委员会,重庆400700 护 ...
  • LC-MS使用经验教训-离子对试剂
    LC-MS使用经验教训-离子对试剂 液质联用仪使用经验(教训) 一做液质,加离子诱导剂得是挥发性的,生物碱用甲酸或乙酸 二我认为要维护好仪器 首先流速不能过大,液质是不能承载过大流速的: 其次电压不要加到极限,尤其是正负离子转换时要适当调整 ...
  • 072 食品中甲醛的检测方法
    ・813・ methylated2CpGislandsincancer[J].LabInvest,2005,85(9):117221180. 国外医学卫生学分册 2008年 第35卷 第5期 tissues[J].Electrophores ...
  • 现代分析技术课程总结
    成绩: 2010-2011 学年第二学期硕士研究生 <现代分析技术>课程总结(100分) 专业: 微生物 学号: [1**********]51 姓名: 王涛 1.专题论文(50分) 根据讲述过的知识,从学校图书馆专业学术文献数 ...
  • 最新仪器分析与实验思考题与习题
    仪器分析与实验思考题与习题 第一章绪论 1 什么是仪器分析?有哪些特点? 2 在常见的仪器分析法种哪几种方法适用于测定元素含量与样品组成?哪几种方法适用于混合物的测定,哪几种分析方法适用于有机化合物分析?哪几种方法适用于物质的结构分析? 仪 ...
  • 室内环境污染物及检测技术
    室内环境污染物及检测技术文献综述 系别:化材系 专业:化学 姓名:汤婷学号:11130225 摘要:介绍中国住宅室内和公共室内环境污染问题的现状,阐述了室内环境中的主要污染物(甲醛.苯.挥发性有机化合物等)对人体健康产生的危害,主要从普通住 ...