• Aucun résultat trouvé

Proteomics of secretory vesicles, perspectives:

CHAPITRE IV: Proteomics of secretory vesicles

2. PROTEOMIC ANALYSIS OF SECRETORY VESICLES:

2.8. Proteomics of secretory vesicles, perspectives:

As shown in this review, the different proteomic analysis of secretory vesicles performed so far, allowed to identify several new proteins for each type of secretory vesicles. This approach has permitted to obtain new information concerning the biogenesis and the exocytosis mechanisms of secretory vesicles. The study of quantitative or qualitative modifications of secretory vesicle proteome after treatment with drugs or secretagogues could also be an interesting alternative strategy to highlight new proteins involved in the modulation of cell secretion.

2.8.1. Protein quantitation:

Recently, several labelling methods have been developed to directly perform relative quantitation of identified proteins in mass spectrometry [130, 131]. SILAC has been specifically developed for in vivo labelling of cultured cells [132]. This method

consists to cultivate two similar cell populations either in normal cell culture condition or with a culture media where one essential amino acid is replaced by an isotopically heavy form that is incorporated in nascent proteins. In a next step, each cell population can be exposed to different stimuli. The two cell populations are mixed and secretory vesicles can be isolated through one of the previously discussed isolation methods. Secretory vesicle proteins can be then identified by mass spectrometry. In addition, heavy and light amino acids can be easily distinguished in mass spectrometry, which allow to detect any modification of protein expression, due to cell stimuli. So far, SILAC approach has been very rarely used to quantitatively compare the proteome of secretory vesicles isolated from cells treated with different stimuli, probably because this method is relatively new and require the acquisition of expensive mass spectrometers. However, SILAC and mass spectrometry represent extraordinary tools to compare in a quantitative manner the proteome of secretory vesicles, exposed to different cell culture conditions. It will thus allow to test the effect of different drugs and secretagogues on the modification of the proteome of secretory vesicles, which could potentially explain the modulation of secretion induced by these compounds.

Other types of protein labelling have been developed for protein quantitation, such as iTRAQ [133] or TMT [134]. In this case, each protein population is chemically labelled with an organic compound of a known mass. The addition of these compounds does not affect the behaviour of the labelled proteins in a mass spectrometer. The fragmentation of the labelled protein in MS/MS mode induces the release of the protein tag corresponding to one specific protein population. The comparison of the intensity of the fragmented tags from different protein populations allows to compare the expression of proteins coming from different biological samples. The main

advantage of this type of protein quantitation is that it enables to compare up to 10 different samples.

2.8.2. Protein post translational modifications:

It is known that post-translational modifications (PTM) are able to modulate activity of most eukaryotic proteins [135]. Therefore, PTM mass spectrometry detection of secretory vesicle proteins after drug or secretagogue cell stimulation, could certainly allow to better understand some of the mechanisms governing modulation of cell secretion. So far, many different PTM have been highlighted, such as phosphorylation, glycation, glycosylation, acylation or ubiquitination [136]. As peptides triggering PTM have more difficulties to be analyzed by mass spectrometry compared to non-PTM peptides, it is often necessary to enriched proteins with a specific PTM, for their efficient detection in mass spectrometry. Several chromatographic columns have been developed to specifically enriched proteins and/or peptides with one particular PTM, such as phosphorylation with IMAC [137]

and TiO2 columns [138] or glycation with boronate columns [139]. The detection of PTM by mass spectrometry is based on the loss of the PTM during peptide fragmentation, which corresponds to a specific ǻmass. Once detected, peptides triggering PTM can be identified. It is also possible to quantify proteins with PTM by using the SILAC strategy. This approach has been used to identify and quantify phopshoproteins from unstimulated and EGF-stimulated cells [136]

2.8.3. Bioinformatics:

Performant mass spectrometry software are necessary for the quantification of proteins and detection of PTMs [140]. However, the main mass spectrometry softwares such as Mascot, Phenyx, Profound or Sequest have been initially developed only to obtain identification of proteins either by PMF or MS/MS. The recent development of efficient mass spectrometers allowing the detection of PTM and/or the quantification of proteins opened an urgent need for the development of new bioinformatics tools allowing the management of PTM and protein quantitation after mass spectrometry analysis. recently, several mass spectrometry software have began to be upgraded in order to be able to automatically detect mass shifts corresponding to a particular PTM or to the labelling of peptides with isobaric tag or isotopic amino acids [141]. However, due to the large numbers of different PTM and quantification methods, the development of more specific bioinformatics tools will be necessary in the future.

3. CONCLUSION:

The comprehension of the mechanism governing cell secretion is fundamental since this process allows the release of endocrine, paracrine or autocrine compounds.

Secretory vesicles are the key organelles of this secretory pathway. Due to the high number of different secretory vesicles, only few of them have been studied in detail so far. However, as described in this review, the combination of subcellular fractionation, proteins separation and mass spectrometry analysis, recently allowed to establish the partial proteome of a wide range of secretory vesicles. All proteomic studies of secretory vesicles allowed to identify several hundreds of proteins that are often present in different types of secretory vesicles. However the knowledge of the

specific protein content of each secretory vesicle may represent the starting point for further biological analysis and could potentially succeed to obtain a better comprehension of the cell secretion mechanisms of each type of cell. In addition, quantitative and/or qualitative comparisons of the secretory vesicle proteome isolated from cells that have been treated with different drugs or secretagogues, will also certainly allow to better understand some impairments of cell secretion.

References:

[1] Normann, T. C., Calcium-dependence of neurosecretion by exocytosis. J Exp Biol 1974, 61, 401-409.

[2] Burgoyne, R. D., Morgan, A., Secretory granule exocytosis. Physiol Rev 2003, 83, 581-632.

[3] Jena, B. P., Cell secretion machinery: studies using the AFM. Ultramicroscopy 2006, 106, 663-669.

[4] Tooze, S. A., Biogenesis of secretory granules. Implications arising from the immature secretory granule in the regulated pathway of secretion. FEBS Lett 1991, 285, 220-224.

[5] Farquhar, M. G., Palade, G. E., The Golgi apparatus (complex)-(1954-1981)-from artifact to center stage. J Cell Biol 1981, 91, 77s-103s.

[6] Leyte, A., Barr, F. A., Kehlenbach, R. H., Huttner, W. B., Multiple trimeric G-proteins on the trans-Golgi network exert stimulatory and inhibitory effects on secretory vesicle

formation. Embo J 1992, 11, 4795-4804.

[7] Ohashi, M., Huttner, W. B., An elevation of cytosolic protein phosphorylation modulates trimeric G-protein regulation of secretory vesicle formation from the trans-Golgi network. J Biol Chem 1994, 269, 24897-24905.

[8] Barr, F. A., Huttner, W. B., A role for ADP-ribosylation factor 1, but not COP I, in secretory vesicle biogenesis from the trans-Golgi network. FEBS Lett 1996, 384, 65-70.

[9] Chen, Y. G., Shields, D., ADP-ribosylation factor-1 stimulates formation of nascent secretory vesicles from the trans-Golgi network of endocrine cells. J Biol Chem 1996, 271, 5297-5300.

[10] Kornfeld, S., Mellman, I., The biogenesis of lysosomes. Annu Rev Cell Biol 1989, 5, 483-525.

[11] Trowbridge, I. S., Collawn, J. F., Hopkins, C. R., Signal-dependent membrane protein trafficking in the endocytic pathway. Annu Rev Cell Biol 1993, 9, 129-161.

[12] Huttner, W. B., Gerdes, H. H., Rosa, P., The granin (chromogranin/secretogranin) family. Trends Biochem Sci 1991, 16, 27-30.

[13] Shennan, K. I., Taylor, N. A., Docherty, K., Calcium- and pH-dependent aggregation and membrane association of the precursor of the prohormone convertase PC2. J Biol Chem 1994, 269, 18646-18650.

[14] Colomer, V., Kicska, G. A., Rindler, M. J., Secretory granule content proteins and the luminal domains of granule membrane proteins aggregate in vitro at mildly acidic pH. J Biol Chem 1996, 271, 48-55.

[15] Cool, D. R., Fenger, M., Snell, C. R., Loh, Y. P., Identification of the sorting signal motif within pro-opiomelanocortin for the regulated secretory pathway. J Biol Chem 1995, 270, 8723-8729.

[16] Thiele, C., Huttner, W. B., The disulfide-bonded loop of chromogranins, which is essential for sorting to secretory granules, mediates homodimerization. J Biol Chem 1998, 273, 1223-1231.

[17] Chanat, E., Weiss, U., Huttner, W. B., Tooze, S. A., Reduction of the disulfide bond of chromogranin B (secretogranin I) in the trans-Golgi network causes its missorting to the constitutive secretory pathways. EMBO J 1993, 12, 2159-2168.

[18] Arvan, P., Halban, P. A., Sorting ourselves out: seeking consensus on trafficking in the beta-cell. Traffic 2004, 5, 53-61.

[19] Blazquez, M., Shennan, K. I., Basic mechanisms of secretion: sorting into the regulated secretory pathway. Biochem Cell Biol 2000, 78, 181-191.

[20] Olofsson, C. S., Gopel, S. O., Barg, S., Galvanovskis, J., et al., Fast insulin secretion reflects exocytosis of docked granules in mouse pancreatic B-cells. Pflugers Arch 2002, 444, 43-51.

[21] Cheviet, S., Coppola, T., Haynes, L. P., Burgoyne, R. D., Regazzi, R., The Rab-binding protein Noc2 is associated with insulin-containing secretory granules and is essential for pancreatic beta-cell exocytosis. Mol Endocrinol 2004, 18, 117-126.

[22] Martin, T. F., Prime movers of synaptic vesicle exocytosis. Neuron 2002, 34, 9-12.

[23] Rorsman, P., Renstrom, E., Insulin granule dynamics in pancreatic beta cells.

Diabetologia 2003, 46, 1029-1045.

[24] Gillis, K. D., Chow, R. H., Kinetics of exocytosis in adrenal chromaffin cells. Semin Cell Dev Biol 1997, 8, 133-140.

[25] Sedej, S., Rose, T., Rupnik, M., cAMP increases Ca2+-dependent exocytosis through both PKA and Epac2 in mouse melanotrophs from pituitary tissue slices. J Physiol 2005, 567, 799-813.

[26] Sorensen, J. B., Formation, stabilisation and fusion of the readily releasable pool of secretory vesicles. Pflugers Arch 2004, 448, 347-362.

[27] Duncan, R. R., Greaves, J., Wiegand, U. K., Matskevich, I., et al., Functional and spatial segregation of secretory vesicle pools according to vesicle age. Nature 2003, 422, 176-180.

[28] Winkler, H., Fischer-Colbrie, R., Regulation of the biosynthesis of large dense-core vesicles in chromaffin cells and neurons. Cell Mol Neurobiol 1998, 18, 193-209.

[29] Bi, G. Q., Morris, R. L., Liao, G., Alderton, J. M., et al., Kinesin- and myosin-driven steps of vesicle recruitment for Ca2+-regulated exocytosis. J Cell Biol 1997, 138, 999-1008.

[30] Varadi, A., Tsuboi, T., Rutter, G. A., Myosin Va transports dense core secretory vesicles in pancreatic MIN6 beta-cells. Mol Biol Cell 2005, 16, 2670-2680.

[31] Abu-Hamdah, R., Cho, W. J., Horber, J. K., Jena, B. P., Secretory vesicles in live cells are not free-floating but tethered to filamentous structures: a study using photonic force microscopy. Ultramicroscopy 2006, 106, 670-673.

[32] Neco, P., Giner, D., del Mar Frances, M., Viniegra, S., Gutierrez, L. M., Differential participation of actin- and tubulin-based vesicle transport systems during secretion in bovine chromaffin cells. Eur J Neurosci 2003, 18, 733-742.

[33] Malacombe, M., Bader, M. F., Gasman, S., Exocytosis in neuroendocrine cells: new tasks for actin. Biochim Biophys Acta 2006, 1763, 1175-1183.

[34] Giner, D., Neco, P., Frances Mdel, M., Lopez, I., et al., Real-time dynamics of the F-actin cytoskeleton during secretion from chromaffin cells. J Cell Sci 2005, 118, 2871-2880.

[35] Sokac, A. M., Bement, W. M., Kiss-and-coat and compartment mixing: coupling

exocytosis to signal generation and local actin assembly. Mol Biol Cell 2006, 17, 1495-1502.

[36] Watanabe, M., Nomura, K., Ohyama, A., Ishikawa, R., et al., Myosin-Va regulates exocytosis through the submicromolar Ca2+-dependent binding of syntaxin-1A. Mol Biol Cell 2005, 16, 4519-4530.

[37] Larina, O., Bhat, P., Pickett, J. A., Launikonis, B. S., et al., Dynamic regulation of the large exocytotic fusion pore in pancreatic acinar cells. Mol Biol Cell 2007, 18, 3502-3511.

[38] Izumi, T., Kasai, K., Gomi, H., Secretory vesicle docking to the plasma membrane:

molecular mechanism and functional significance. Diabetes Obes Metab 2007, 9 Suppl 2, 109-117.

[39] Voets, T., Toonen, R. F., Brian, E. C., de Wit, H., et al., Munc18-1 promotes large dense-core vesicle docking. Neuron 2001, 31, 581-591.

[40] Korteweg, N., Maia, A. S., Thompson, B., Roubos, E. W., et al., The role of Munc18-1 in docking and exocytosis of peptide hormone vesicles in the anterior pituitary. Biol Cell 2005, 97, 445-455.

[41] Verhage, M., Maia, A. S., Plomp, J. J., Brussaard, A. B., et al., Synaptic assembly of the brain in the absence of neurotransmitter secretion. Science 2000, 287, 864-869.

[42] Klenchin, V. A., Martin, T. F., Priming in exocytosis: attaining fusion-competence after vesicle docking. Biochimie 2000, 82, 399-407.

[43] Stojilkovic, S. S., Ca2+-regulated exocytosis and SNARE function. Trends Endocrinol Metab 2005, 16, 81-83.

[44] Blank, P. S., Vogel, S. S., Malley, J. D., Zimmerberg, J., A kinetic analysis of calcium-triggered exocytosis. J Gen Physiol 2001, 118, 145-156.

[45] Sollner, T., Whiteheart, S. W., Brunner, M., Erdjument-Bromage, H., et al., SNAP receptors implicated in vesicle targeting and fusion. Nature 1993, 362, 318-324.

[46] Gerst, J. E., SNAREs and SNARE regulators in membrane fusion and exocytosis. Cell Mol Life Sci 1999, 55, 707-734.

[47] Washbourne, P., Pellizzari, R., Baldini, G., Wilson, M. C., Montecucco, C., Botulinum neurotoxin types A and E require the SNARE motif in SNAP-25 for proteolysis. FEBS Lett 1997, 418, 1-5.

[48] Gutierrez, L. M., Quintanar, J. L., Viniegra, S., Salinas, E., et al., Anti-syntaxin

antibodies inhibit calcium-dependent catecholamine secretion from permeabilized chromaffin cells. Biochem Biophys Res Commun 1995, 206, 1-7.

[49] Sutton, R. B., Fasshauer, D., Jahn, R., Brunger, A. T., Crystal structure of a SNARE complex involved in synaptic exocytosis at 2.4 A resolution. Nature 1998, 395, 347-353.

[50] Jahn, R., Sudhof, T. C., Membrane fusion and exocytosis. Annu Rev Biochem 1999, 68, 863-911.

[51] Lin, R. C., Scheller, R. H., Structural organization of the synaptic exocytosis core complex. Neuron 1997, 19, 1087-1094.

[52] Perin, M. S., Fried, V. A., Mignery, G. A., Jahn, R., Sudhof, T. C., Phospholipid binding by a synaptic vesicle protein homologous to the regulatory region of protein kinase C. Nature 1990, 345, 260-263.

[53] Perin, M. S., Johnston, P. A., Ozcelik, T., Jahn, R., et al., Structural and functional conservation of synaptotagmin (p65) in Drosophila and humans. J Biol Chem 1991, 266, 615-622.

[54] Chapman, E. R., Synaptotagmin: a Ca(2+) sensor that triggers exocytosis? Nat Rev Mol Cell Biol 2002, 3, 498-508.

[55] Butz, S., Fernandez-Chacon, R., Schmitz, F., Jahn, R., Sudhof, T. C., The subcellular localizations of atypical synaptotagmins III and VI. Synaptotagmin III is enriched in synapses and synaptic plasma membranes but not in synaptic vesicles. J Biol Chem 1999, 274, 18290-18296.

[56] Gut, A., Kiraly, C. E., Fukuda, M., Mikoshiba, K., et al., Expression and localisation of synaptotagmin isoforms in endocrine beta-cells: their function in insulin exocytosis. J Cell Sci 2001, 114, 1709-1716.

[57] Koh, T. W., Bellen, H. J., Synaptotagmin I, a Ca2+ sensor for neurotransmitter release.

Trends Neurosci 2003, 26, 413-422.

[58] Liu, J., Guo, T., Wu, J., Bai, X., et al., Overexpression of complexin in PC12 cells inhibits exocytosis by preventing SNARE complex recycling. Biochemistry (Mosc) 2007, 72, 439-444.

[59] Melia, T. J., Jr., Putting the clamps on membrane fusion: how complexin sets the stage for calcium-mediated exocytosis. FEBS Lett 2007, 581, 2131-2139.

[60] Bracher, A., Kadlec, J., Betz, H., Weissenhorn, W., X-ray structure of a neuronal complexin-SNARE complex from squid. J Biol Chem 2002, 277, 26517-26523.

[61] Tang, J., Maximov, A., Shin, O. H., Dai, H., et al., A complexin/synaptotagmin 1 switch controls fast synaptic vesicle exocytosis. Cell 2006, 126, 1175-1187.

[62] Zhao, C., Slevin, J. T., Whiteheart, S. W., Cellular functions of NSF: not just SNAPs and SNAREs. FEBS Lett 2007, 581, 2140-2149.

[63] Stenbeck, G., Soluble NSF-attachment proteins. Int J Biochem Cell Biol 1998, 30, 573-577.

[64] Weidenhaupt, M., Bruckert, F., Louwagie, M., Garin, J., Satre, M., Functional and molecular identification of novel members of the ubiquitous membrane fusion proteins alpha- and gamma-SNAP (soluble N-ethylmaleimide-sensitive factor-attachment proteins) families in Dictyostelium discoideum. Eur J Biochem 2000, 267, 2062-2070.

[65] Seabra, M. C., Mules, E. H., Hume, A. N., Rab GTPases, intracellular traffic and disease.

Trends Mol Med 2002, 8, 23-30.

[66] Lang, T., Jahn, R., Core proteins of the secretory machinery. Handb Exp Pharmacol 2008, 107-127.

[67] Tanaka, M., Miyoshi, J., Ishizaki, H., Togawa, A., et al., Role of Rab3 GDP/GTP exchange protein in synaptic vesicle trafficking at the mouse neuromuscular junction. Mol Biol Cell 2001, 12, 1421-1430.

[68] Fischer von Mollard, G., Mignery, G. A., Baumert, M., Perin, M. S., et al., rab3 is a small GTP-binding protein exclusively localized to synaptic vesicles. Proc Natl Acad Sci U S A 1990, 87, 1988-1992.

[69] Thoma, N. H., Niculae, A., Goody, R. S., Alexandrov, K., Double prenylation by RabGGTase can proceed without dissociation of the mono-prenylated intermediate. J Biol Chem 2001, 276, 48631-48636.

[70] Chou, J. H., Jahn, R., Binding of Rab3A to synaptic vesicles. J Biol Chem 2000, 275, 9433-9440.

[71] Jordens, I., Marsman, M., Kuijl, C., Neefjes, J., Rab proteins, connecting transport and vesicle fusion. Traffic 2005, 6, 1070-1077.

[72] Darchen, F., Goud, B., Multiple aspects of Rab protein action in the secretory pathway:

focus on Rab3 and Rab6. Biochimie 2000, 82, 375-384.

[73] Cheviet, S., Waselle, L., Regazzi, R., Noc-king out exocrine and endocrine secretion.

Trends Cell Biol 2004, 14, 525-528.

[74] Tolmachova, T., Anders, R., Stinchcombe, J., Bossi, G., et al., A general role for Rab27a in secretory cells. Mol Biol Cell 2004, 15, 332-344.

[75] Izumi, T., Gomi, H., Kasai, K., Mizutani, S., Torii, S., The roles of Rab27 and its effectors in the regulated secretory pathways. Cell Struct Funct 2003, 28, 465-474.

[76] Lindau, M., Almers, W., Structure and function of fusion pores in exocytosis and ectoplasmic membrane fusion. Curr Opin Cell Biol 1995, 7, 509-517.

[77] Jackson, M. B., In search of the fusion pore of exocytosis. Biophys Chem 2007, 126, 201-208.

[78] Heuser, J. E., Reese, T. S., Structural changes after transmitter release at the frog neuromuscular junction. J Cell Biol 1981, 88, 564-580.

[79] Harata, N. C., Aravanis, A. M., Tsien, R. W., Kiss-and-run and full-collapse fusion as modes of exo-endocytosis in neurosecretion. J Neurochem 2006, 97, 1546-1570.

[80] Valtorta, F., Meldolesi, J., Fesce, R., Synaptic vesicles: is kissing a matter of competence? Trends Cell Biol 2001, 11, 324-328.

[81] Fernandez-Peruchena, C., Navas, S., Montes, M. A., Alvarez de Toledo, G., Fusion pore regulation of transmitter release. Brain Res Brain Res Rev 2005, 49, 406-415.

[82] Stevens, C. F., Williams, J. H., "Kiss and run" exocytosis at hippocampal synapses. Proc Natl Acad Sci U S A 2000, 97, 12828-12833.

[83] Dvorak, J. A., The application of atomic force microscopy to the study of living vertebrate cells in culture. Methods 2003, 29, 86-96.

[84] Schneider, S. W., Sritharan, K. C., Geibel, J. P., Oberleithner, H., Jena, B. P., Surface dynamics in living acinar cells imaged by atomic force microscopy: identification of plasma membrane structures involved in exocytosis. Proc Natl Acad Sci U S A 1997, 94, 316-321.

[85] Jena, B. P., Molecular machinery and mechanism of cell secretion. Exp Biol Med (Maywood) 2005, 230, 307-319.

[86] Cho, S. J., Quinn, A. S., Stromer, M. H., Dash, S., et al., Structure and dynamics of the fusion pore in live cells. Cell Biol Int 2002, 26, 35-42.

[87] Cho, S. J., Jeftinija, K., Glavaski, A., Jeftinija, S., et al., Structure and dynamics of the fusion pores in live GH-secreting cells revealed using atomic force microscopy.

Endocrinology 2002, 143, 1144-1148.

[88] Kelly, M. L., Cho, W. J., Jeremic, A., Abu-Hamdah, R., Jena, B. P., Vesicle swelling regulates content expulsion during secretion. Cell Biol Int 2004, 28, 709-716.

[89] Cho, S. J., Sattar, A. K., Jeong, E. H., Satchi, M., et al., Aquaporin 1 regulates GTP-induced rapid gating of water in secretory vesicles. Proc Natl Acad Sci U S A 2002, 99, 4720-4724.

[90] Kakimoto, M., Inoguchi, T., Sonta, T., Yu, H. Y., et al., Accumulation of 8-hydroxy-2'-deoxyguanosine and mitochondrial DNA deletion in kidney of diabetic rats. Diabetes 2002, 51, 1588-1595.

[91] Huber, L. A., Pfaller, K., Vietor, I., Organelle proteomics: implications for subcellular fractionation in proteomics. Circ Res 2003, 92, 962-968.

[92] Graham, J. M., Ford, T., Rickwood, D., Isolation of the major subcellular organelles from mouse liver using Nycodenz gradients without the use of an ultracentrifuge. Anal Biochem 1990, 187, 318-323.

[93] Brunner, Y., Coute, Y., Iezzi, M., Foti, M., et al., Proteomics analysis of insulin secretory granules. Mol Cell Proteomics 2007, 6, 1007-1017.

[94] Watarai, H., Inagaki, Y., Kubota, N., Fuju, K., et al., Proteomic approach to the identification of cell membrane proteins. Electrophoresis 2000, 21, 460-464.

[95] Iezzi, M., Kouri, G., Fukuda, M., Wollheim, C. B., Synaptotagmin V and IX isoforms control Ca2+ -dependent insulin exocytosis. J Cell Sci 2004, 117, 3119-3127.

[96] Kjeldsen, L., Sengelov, H., Borregaard, N., Subcellular fractionation of human neutrophils on Percoll density gradients. J Immunol Methods 1999, 232, 131-143.

[97] Murayama, K., Fujimura, T., Morita, M., Shindo, N., One-step subcellular fractionation of rat liver tissue using a Nycodenz density gradient prepared by freezing-thawing and two-dimensional sodium dodecyl sulfate electrophoresis profiles of the main fraction of

organelles. Electrophoresis 2001, 22, 2872-2880.

[98] Mura, C. V., Becker, M. I., Orellana, A., Wolff, D., Immunopurification of Golgi vesicles by magnetic sorting. J Immunol Methods 2002, 260, 263-271.

[99] Morciano, M., Burre, J., Corvey, C., Karas, M., et al., Immunoisolation of two synaptic vesicle pools from synaptosomes: a proteomics analysis. J Neurochem 2005, 95, 1732-1745.

[100] Jethwaney, D., Islam, M. R., Leidal, K. G., de Bernabe, D. B., et al., Proteomic analysis of plasma membrane and secretory vesicles from human neutrophils. Proteome Sci 2007, 5, 12.

[101] Casey, T. M., Meade, J. L., Hewitt, E. W., Organelle proteomics: identification of the exocytic machinery associated with the natural killer cell secretory lysosome. Mol Cell Proteomics 2007, 6, 767-780.

[102] Wegrzyn, J., Lee, J., Neveu, J. M., Lane, W. S., Hook, V., Proteomics of

neuroendocrine secretory vesicles reveal distinct functional systems for biosynthesis and exocytosis of peptide hormones and neurotransmitters. J Proteome Res 2007, 6, 1652-1665.

[103] Barile, M., Pisitkun, T., Yu, M. J., Chou, C. L., et al., Large scale protein identification in intracellular aquaporin-2 vesicles from renal inner medullary collecting duct. Mol Cell Proteomics 2005, 4, 1095-1106.

[104] Muth, E., Driscoll, W. J., Smalstig, A., Goping, G., Mueller, G. P., Proteomic analysis of rat atrial secretory granules: a platform for testable hypotheses. Biochim Biophys Acta 2004, 1699, 263-275.

[105] Rindler, M. J., Xu, C. F., Gumper, I., Smith, N. N., Neubert, T. A., Proteomic analysis of pancreatic zymogen granules: identification of new granule proteins. J Proteome Res 2007, 6, 2978-2992.

[106] Lominadze, G., Powell, D. W., Luerman, G. C., Link, A. J., et al., Proteomic analysis of human neutrophil granules. Mol Cell Proteomics 2005, 4, 1503-1521.

[107] Chen, X., Walker, A. K., Strahler, J. R., Simon, E. S., et al., Organellar proteomics:

analysis of pancreatic zymogen granule membranes. Mol Cell Proteomics 2006, 5, 306-312.

[108] Choe, L. H., Werner, B. G., Lee, K. H., Two-dimensional protein electrophoresis: from molecular pathway discovery to biomarker discovery in neurological disorders. NeuroRx 2006, 3, 327-335.

[109] Braun, R. J., Kinkl, N., Beer, M., Ueffing, M., Two-dimensional electrophoresis of membrane proteins. Anal Bioanal Chem 2007, 389, 1033-1045.

[110] Tribl, F., Gerlach, M., Marcus, K., Asan, E., et al., "Subcellular proteomics" of

neuromelanin granules isolated from the human brain. Mol Cell Proteomics 2005, 4, 945-957.

[111] Coughenour, H. D., Spaulding, R. S., Thompson, C. M., The synaptic vesicle proteome:

a comparative study in membrane protein identification. Proteomics 2004, 4, 3141-3155.

[112] Burre, J., Beckhaus, T., Schagger, H., Corvey, C., et al., Analysis of the synaptic vesicle proteome using three gel-based protein separation techniques. Proteomics 2006, 6, 6250-6262.

[113] Nagele, E., Vollmer, M., Horth, P., Vad, C., 2D-LC/MS techniques for the

identification of proteins in highly complex mixtures. Expert Rev Proteomics 2004, 1, 37-46.

[114] Guerrera, I. C., Kleiner, O., Application of mass spectrometry in proteomics. Biosci Rep 2005, 25, 71-93.

[115] Karas, M., Hillenkamp, F., Laser desorption ionization of proteins with molecular masses exceeding 10,000 daltons. Anal Chem 1988, 60, 2299-2301.

[116] Hernandez-Ruiz, L., Valverde, F., Jimenez-Nunez, M. D., Ocana, E., et al., Organellar proteomics of human platelet dense granules reveals that 14-3-3zeta is a granule protein related to atherosclerosis. J Proteome Res 2007, 6, 4449-4457.

[117] Fenn, J. B., Mann, M., Meng, C. K., Wong, S. F., Whitehouse, C. M., Electrospray ionization for mass spectrometry of large biomolecules. Science 1989, 246, 64-71.

[118] Cotter, R. J., Griffith, W., Jelinek, C., Tandem time-of-flight (TOF/TOF) mass

spectrometry and the curved-field reflectron. J Chromatogr B Analyt Technol Biomed Life Sci 2007, 855, 2-13.

[119] Medzihradszky, K. F., Campbell, J. M., Baldwin, M. A., Falick, A. M., et al., The characteristics of peptide collision-induced dissociation using a high-performance MALDI-TOF/TOF tandem mass spectrometer. Anal Chem 2000, 72, 552-558.

[119] Medzihradszky, K. F., Campbell, J. M., Baldwin, M. A., Falick, A. M., et al., The characteristics of peptide collision-induced dissociation using a high-performance MALDI-TOF/TOF tandem mass spectrometer. Anal Chem 2000, 72, 552-558.