全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Advantageous Uses of Mass Spectrometry for the Quantification of Proteins

DOI: 10.1155/2013/219452

Full-Text   Cite this paper   Add to My Lib

Abstract:

Quantitative protein measurements by mass spectrometry have gained wide acceptance in research settings. However, clinical uptake of mass spectrometric protein assays has not followed suit. In part, this is due to the long-standing acceptance by regulatory agencies of immunological assays such as ELISA assays. In most cases, ELISAs provide highly accurate, sensitive, relatively inexpensive, and simple assays for many analytes. The barrier to acceptance of mass spectrometry in these situations will remain high. However, mass spectrometry provides solutions to certain protein measurements that are difficult, if not impossible, to accomplish by immunological methods. Cases where mass spectrometry can provide solutions to difficult assay development include distinguishing between very closely related protein species and monitoring biological and analytical variability due to sample handling and very high multiplexing capacity. This paper will highlight several examples where mass spectrometry has made certain protein measurements possible where immunological techniques have had a great difficulty. 1. Introduction Quantitative mass spectrometry of proteins has evolved dramatically over the last decade. Early methods involved labeling proteins with reagents enriched in stable isotopes in order to introduce mass tags into proteins of interest for relative quantification of proteins [1, 2]. These reagents have been refined over the years and have found widespread use in the form of the ITRAQ reagent [3]. Metabolic labeling of proteins with stable isotope-enriched amino acids has also been used as a technique for the relative quantification of proteins in cell culture systems [4]. Additionally, “label-free” methods have been developed for relative quantification of proteins in complex mixtures [5, 6]. As mentioned, these methods were developed for relative quantification of proteins, that is, comparing two or more samples and determining whether levels of proteins increased or decreased in response to some perturbation. Isotopically labeled peptides have been used as standards for the absolute quantification of proteins in complex mixtures. Variations of this approach include the AQUA and SISCAPA methods [7, 8]. Among the advantages of these techniques is that a quantitative assay may be developed for a given protein without the need for an antibody [7]. Alternatively an antibody to a synthetic peptide may be used [8]. This greatly simplifies the development of assays from immunological formats, such as ELISA, where well-characterized antibodies are needed.

References

[1]  S. P. Gygi, B. Rist, S. A. Gerber, F. Turecek, M. H. Gelb, and R. Aebersold, “Quantitative analysis of complex protein mixtures using isotope-coded affinity tags,” Nature Biotechnology, vol. 17, no. 10, pp. 994–999, 1999.
[2]  A. Chakraborty and F. E. Regnier, “Global internal standard technology for comparative proteomics,” Journal of Chromatography A, vol. 949, no. 1-2, pp. 173–184, 2002.
[3]  L. DeSouza, G. Diehl, M. J. Rodrigues et al., “Search for cancer markers from endometrial tissues using differentially labeled tags iTRAQ and cICAT with multidimensional liquid chromatography and tandem mass spectrometry,” Journal of Proteome Research, vol. 4, no. 2, pp. 377–386, 2005.
[4]  S. E. Ong, B. Blagoev, I. Kratchmarova et al., “Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics,” Molecular & Cellular Proteomics, vol. 1, no. 5, pp. 376–386, 2002.
[5]  P. V. Bondarenko, D. Chelius, and T. A. Shaler, “Identification and relative quantitation of protein mixtures by enzymatic digestion followed by capillary reversed-phase liquid chromatography—tandem mass spectrometry,” Analytical Chemistry, vol. 74, no. 18, pp. 4741–4749, 2002.
[6]  W. M. Old, K. Meyer-Arendt, L. Aveline-Wolf et al., “Comparison of label-free methods for quantifying human proteins by shotgun proteomics,” Molecular & Cellular Proteomics, vol. 4, no. 10, pp. 1487–1502, 2005.
[7]  S. A. Gerber, J. Rush, O. Stemman, M. W. Kirschner, and S. P. Gygi, “Absolute quantification of proteins and phosphoproteins from cell lysates by tandem MS,” Proceedings of the National Academy of Sciences of the United States of America, vol. 100, no. 12, pp. 6940–6945, 2003.
[8]  N. L. Anderson, N. G. Anderson, L. R. Haines, D. B. Hardie, R. W. Olafson, and T. W. Pearson, “Mass spectrometric quantitation of peptides and proteins using Stable Isotope Standards and Capture by Anti-Peptide Antibodies (SISCAPA),” Journal of Proteome Research, vol. 3, no. 2, pp. 235–244, 2004.
[9]  M. Bantscheff, M. Schirle, G. Sweetman, J. Rick, and B. Kuster, “Quantitative mass spectrometry in proteomics: a critical review,” Analytical and Bioanalytical Chemistry, vol. 389, no. 4, pp. 1017–1031, 2007.
[10]  J. Poirier, J. Davignon, D. Bouthillier, S. Kogan, P. Bertrand, and S. Gauthier, “Apolipoprotein E polymorphism and Alzheimer's disease,” The Lancet, vol. 342, no. 8873, pp. 697–699, 1993.
[11]  J. E. Eichner, S. T. Dunn, G. Perveen, D. M. Thompson, K. E. Stewart, and B. C. Stroehla, “Apolipoprotein E polymorphism and cardiovascular disease: a HuGE review,” American Journal of Epidemiology, vol. 155, no. 6, pp. 487–495, 2002.
[12]  K. R. Wildsmith, B. Han, and R. J. Bateman, “Method for the simultaneous quantitation of apolipoprotein E isoforms using tandem mass spectrometry,” Analytical Biochemistry, vol. 395, no. 1, pp. 116–118, 2009.
[13]  J. R. Murrell, R. G. Schoner, J. J. Liepnieks, H. N. Rosen, A. C. Moses, and M. D. Benson, “Production and functional analysis of normal and variant recombinant human transthyretin proteins,” The Journal of Biological Chemistry, vol. 267, no. 23, pp. 16595–16600, 1992.
[14]  A. Lim, T. Prokaeva, M. E. McComb et al., “Characterization of transthyretin variants in familial transthyretin amyloidosis by mass spectrometric peptide mapping and DNA sequence analysis,” Analytical Chemistry, vol. 74, no. 4, pp. 741–751, 2002.
[15]  Y. Sekijima, R. L. Wiseman, J. Matteson, et al., “The biological and chemical basis for tissue-selective amyloid disease. A mutation in a secreted protein can lead to decreased folding efficiency within the ER and reduced exporting, such as the disulfide isomerases,” Cell, vol. 121, pp. 73–85, 2005.
[16]  L. Lobato and A. Rocha, “Transthyretin amyloidosis and the kidney,” Clinical Journal of the American Society of Nephrology, no. 8, pp. 1337–1346, 2012.
[17]  A. Lim, T. Prokaeva, M. E. McComb, L. H. Connors, M. Skinner, and C. E. Costello, “Identification of S-sulfonation and S-thiolation of a novel transthyretin Phe33Cys variant from a patient diagnosed with familial transthyretin amyloidosis,” Protein Science, vol. 12, no. 8, pp. 1775–1785, 2003.
[18]  U. A. Kiernan, K. A. Tubbs, K. Gruber et al., “High-throughput protein characterization using mass spectrometric immunoassay,” Analytical Biochemistry, vol. 301, no. 1, pp. 49–56, 2002.
[19]  O. Trenchevska, E. Kamcheva, and D. Nedelkov, “Mass spectrometric immunoassay for quantitative determination of transthyretin and its variants,” Proteomics, vol. 11, pp. 3633–3641, 2011.
[20]  Z. Yang, M. Hayes, X. Fang, M. P. Daley, S. Effenberg, and F. L. S. Tse, “LC-MS/MS approach for quantification of therapeutic proteins in plasma using a protein internal standard and 2D-solid-phase extraction cleanup,” Analytical Chemistry, vol. 79, no. 24, pp. 9294–9301, 2007.
[21]  O. Heudi, S. Barteau, D. Zimmer et al., “Towards absolute quantification of therapeutic monoclonal antibody in serum by LC-MS/MS using isotope-labeled antibody standard and protein cleavage isotope dilution mass spectrometry,” Analytical Chemistry, vol. 80, no. 11, pp. 4200–4207, 2008.
[22]  C. Hagman, D. Ricke, S. Ewert, S. Bek, R. Falchetto, and F. Bitsch, “Absolute quantification of monoclonal antibodies in biofluids by liquid chromatography-tandem mass spectrometry,” Analytical Chemistry, vol. 80, no. 4, pp. 1290–1296, 2008.
[23]  L. J. M. Dekker, L. Zeneyedpour, E. Brouwer, M. M. van Duijn, P. A. E. Sillevis Smitt, and T. M. Luider, “An antibody-based biomarker discovery method by mass spectrometry sequencing of complementarity determining regions,” Analytical and Bioanalytical Chemistry, vol. 399, no. 3, pp. 1081–1091, 2011.
[24]  J. J. Pesavento, C. A. Mizzen, and N. L. Kelleher, “Quantitative analysis of modified proteins and their positional isomers by tandem mass spectrometry: human histone H4,” Analytical Chemistry, vol. 78, no. 13, pp. 4271–4280, 2006.
[25]  B. A. Garcia, J. J. Pesavento, C. A. Mizzen, and N. L. Kelleher, “Pervasive combinatorial modification of histone H3 in human cells,” Nature Methods, vol. 4, no. 6, pp. 487–489, 2007.
[26]  D. Phanstiel, J. Brumbaugh, W. T. Berggren et al., “Mass spectrometry identifies and quantifies 74 unique histone H4 isoforms in differentiating human embryonic stem cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 11, pp. 4093–4098, 2008.
[27]  T. Jenuwein and C. D. Allis, “Translating the histone code,” Science, vol. 293, no. 5532, pp. 1074–1080, 2001.
[28]  M. Kojima, H. Hosoda, Y. Date, M. Nakazato, H. Matsuo, and K. Kangawa, “Ghrelin is a growth-hormone-releasing acylated peptide from stomach,” Nature, vol. 402, no. 6762, pp. 656–660, 1999.
[29]  M. Tschop, D. L. Smiley, and M. L. Heiman, “Ghrelin induces adiposity in rodents,” Nature, vol. 407, no. 6806, pp. 908–913, 2000.
[30]  Y. Date, M. Nakazato, S. Hashiguchi et al., “Ghrelin is present in pancreatic α-cells of humans and rats and stimulates insulin secretion,” Diabetes, vol. 51, no. 1, pp. 124–129, 2002.
[31]  J. Liu, C. E. Prudom, R. Nass et al., “Novel ghrelin assays provide evidence for independent regulation of ghrelin acylation and secretion in healthy young men,” The Journal of Clinical Endocrinology & Metabolism, vol. 93, no. 5, pp. 1980–1987, 2008.
[32]  C. Prudom, J. Liu, J. Patrie et al., “Comparison of competitive radioimmunoassays and two-site sandwich assays for the measurement and interpretation of plasma ghrelin levels,” The Journal of Clinical Endocrinology & Metabolism, vol. 95, no. 5, pp. 2351–2358, 2010.
[33]  J. A. Gutierrez, J. A. Willency, M. D. Knierman, et al., “From Ghrelin to Ghrelin's O-acyl transferase,” Methods in Enzymology, vol. 514, pp. 129–146, 2012.
[34]  J. Hardy and D. J. Selkoe, “The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics,” Science, vol. 297, no. 5580, pp. 353–356, 2002.
[35]  T. Jonsson, J. K. Atwal, S. Steinberg, et al., “A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline,” Nature, vol. 488, no. 7409, pp. 96–99, 2012.
[36]  E. Portelius, A. Westman-Brinkmalm, H. Zetterberg, and K. Blennow, “Determination of β-amyloid peptide signatures in cerebrospinal fluid using immunoprecipitation-mass spectrometry,” Journal of Proteome Research, vol. 5, no. 4, pp. 1010–1016, 2006.
[37]  R. B. DeMattos, M. M. Racke, V. Gelfanova, et al., “Identification, characterization, and comparison of amino-terminally truncated Aβ42 peptides in Alzheimer's disease brain tissue and in plasma from Alzheimer's patients receiving solanezumab immunotherapy treatment,” Alzheimer's & Dementia, vol. 5, no. 4, supplement, pp. P156–P157, 2009.
[38]  J. R. Cirrito, P. C. May, M. A. O'Dell et al., “In vivo assessment of brain interstitial fluid with microdialysis reveals plaque-associated changes in amyloid-β metabolism and half-life,” Journal of Neuroscience, vol. 23, no. 26, pp. 8844–8853, 2003.
[39]  T. Oe, B. L. Ackermann, K. Inoue et al., “Quantitative analysis of amyloid β peptides in cerebrospinal fluid of Alzheimer's disease patients by immunoaffinity purification and stable isotope dilution liquid chromatography/negative electrospray ionization tandem mass spectrometry,” Rapid Communications in Mass Spectrometry, vol. 20, no. 24, pp. 3723–3735, 2006.
[40]  J. Legleiter, D. L. Czilli, B. Gitter, R. B. DeMattos, D. M. Holtzman, and T. Kowalewski, “Effect of differentanti-Abeta antibodies on Abeta fibrillogenesis asassessed by atomic force microscopy,” Journal of Molecular Biology, vol. 335, no. 4, pp. 997–1006, 2004.
[41]  V. Gelfanova, R. E. Higgs, R. A. Dean et al., “Quantitative analysis of amyloid-β peptides in cerebrospinal fluid using immunoprecipitation and MALDI-Tof mass spectrometry,” Briefings in Functional Genomics & Proteomics, vol. 6, no. 2, pp. 149–158, 2007.
[42]  M. J. Ford, J. L. Cantone, C. Polson, J. H. Toyn, J. E. Meredith, and D. M. Drexler, “Qualitative and quantitative characterization of the amyloid β peptide (Aβ) population in biological matrices using an immunoprecipitation-LC/MS assay,” Journal of Neuroscience Methods, vol. 168, no. 2, pp. 465–474, 2008.
[43]  A. M. Tabert, M. P. Goodwin, J. S. Duncan, C. D. Fico, and R. G. Cooks, “Multiplexed rectilinear ion trap mass spectrometer for high-throughput analysis,” Analytical Chemistry, vol. 78, no. 14, pp. 4830–4838, 2006.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133