Proteogenomic analysis reveals exosomes are more oncogenic than ectosomes

Shivakumar Keerthikumar, Lahiru Gangoda, Michael Liem, Pamali Fonseka, Ishara Atukorala, Cemil Ozcitti, Adam Mechler, Christopher G Adda, Ching-Seng Ang, Suresh Mathivanan, Shivakumar Keerthikumar, Lahiru Gangoda, Michael Liem, Pamali Fonseka, Ishara Atukorala, Cemil Ozcitti, Adam Mechler, Christopher G Adda, Ching-Seng Ang, Suresh Mathivanan

Abstract

Extracellular vesicles (EVs) include the exosomes (30-100 nm) that are produced through the endocytic pathway via the multivesicular bodies and the ectosomes (100-1000 nm) that are released through the budding of the plasma membrane. Despite the differences in the mode of biogenesis and size, reliable markers that can distinguish between exosomes and ectosomes are non-existent. Moreover, the precise functional differences between exosomes and ectosomes remains poorly characterised. Here, using label-free quantitative proteomics, we highlight proteins that could be exploited as markers to discriminate between exosomes and ectosomes. For the first time, a global proteogenomics analysis unveiled the secretion of mutant proteins that are implicated in cancer progression through tumor-derived EVs. Follow up integrated bioinformatics analysis highlighted the enrichment of oncogenic cargo in exosomes and ectosomes. Interestingly, exosomes induced significant cell proliferation and migration in recipient cells compared to ectosomes confirming the oncogenic nature of exosomes. These findings ascertain that cancer cells facilitate oncogenesis by the secretion of mutant and oncoproteins into the tumor microenvironment via exosomes and ectosomes. The integrative proteogenomics approach utilized in this study has the potential to identify disease biomarker candidates which can be later assayed in liquid biopsies obtained from cancer patients.

Keywords: ectosomes; exosomes; extracellular vesicles; integrated OMICs analysis; proteogenomics.

Conflict of interest statement

CONFLICTS OF INTEREST

None declared

Figures

Figure 1. Isolation and characterization of EVs
Figure 1. Isolation and characterization of EVs
(a) Western blot analysis (10 μg) of fractions of increasing density obtained by OptiPrepTM density gradient centrifugation (from SH-SY5Y cells) showed the presence of Alix and TSG101 in fractions 3-9 with a clear enrichment in fraction 7 (1.10 g/mL). Pellet obtained from 10,000 g also contained detectable amount of Alix and TSG101. (b) Western blot of GM130 (Golgi marker) and Alix in EVs isolated from 1.10 and 1.14 g/mL densities, 10,000 g pellet and WCL is shown. The absence of GM130 in fractions 7, 9 and 10K confirms the depletion of contaminating vesicles arising from apoptosis. (c) TEM images of EVs isolated from SH-SY5Y cells by OptiPrepTM gradient corresponding to the density 1.10 g/mL showed vesicles in the range of 30-100 nm diameter consistent with exosomes. (d) TEM images of EVs isolated from SH-SY5Y cells by OptiPrepTM gradient corresponding to the density 1.14-1.20 g/mL showed vesicles more than 200 nm in diameter. (e) TEM images of vesicles recovered from 10,000 g pellet (10K) showed aggregates and large EVs secreted by SH-SY5Y neuroblastoma and (f) LIM1215 colorectal cancer cells.
Figure 2. AFM imaging based characterization of…
Figure 2. AFM imaging based characterization of EVs isolated by OptiPrep™ density gradient centrifugation
(a) AFM images of fraction 7 (1.10 g/mL) isolated by OptiPrepTM density gradient centrifugation showed vesicles in the range of 30-70 nm. Four profile images (1-4) of the vesicle diameter are also depicted. (b) AFM images of EVs obtained by OptiPrepTM density gradient centrifugation corresponding to 1.14-1.20 g/mL showed enrichment of larger vesicles. Eight profile (1-8) images of the vesicle diameter are also depicted.
Figure 3. Vesicle size distribution and Venn…
Figure 3. Vesicle size distribution and Venn diagram of total and differentially abundant proteins in exosomes, ectosomes and WCL
(a) Pie chart representing the size distribution of vesicles in fraction 7 (1.10 g/mL). (b) Pie chart representing the size distribution of vesicles in fraction 9 (1.14-1.20 g/mL). (c) Label-free quantitative mass spectrometry-based proteomics analysis was performed on WCL, exosomes (1.10 g/mL) and ectosomes (1.14-1.20 g/mL). Venn diagram of differentially expressed proteins in exosomes and (d) ectosomes in comparison to WCL is displayed. (e) Venn diagram of differentially abundant proteins identified in exosomes and ectosomes showed 693 proteins enriched more than 2-fold in exosomes compared to ectosomes. On the contrast, 770 proteins were enriched more than 2-fold ectosomes compared to exosomes.
Figure 4. Fold change of proteins known…
Figure 4. Fold change of proteins known to be involved in EV biogenesis, trafficking and membrane fusion
Asterisk (*) represents proteins that are not detected in ectosomes and/or 10K while # represents proteins that are not detected in exosomes. Dotted line represents 2-fold cut off. (a) Histogram of proteins involved in ESCRT machinery showed that proteins involved in ESCRT were more than 2-fold abundant (except VPS37D) in exosomes compared to ectosomes. (b) Histogram of tetraspanins shows that exosomes are enriched with tetraspanins CD81, TSPAN14 and TSPAN9 compared to the ectosomes and 10K. CD63, CD9 and TSPAN6 were not enriched more than 2-fold in any data set (even though detected in exosomes with one peptide identification from multiple MS/MS spectra). (c) Among the RAB GTPases, subsets of them were uniquely identified in exosomes (*) while some others in ectosomes and/or 10K (#). The same set of RAB GTPases was uniquely present in both ectosomes and 10K while not detected in exosomes. (d) Proteins known to be involved in trafficking and membrane fusion were all enriched in exosomes except FGA and HLA-A. (e) A literature survey was carried out to identify proteins either involved in ectosome biogenesis or identified in ectosomes. The manually curated protein list was plotted as a histogram based on the proteomics data. Except MMP2, other proteins were not enriched in ectosomes or 10K.
Figure 5. Western blotting analysis of exosomes…
Figure 5. Western blotting analysis of exosomes and ectosomes and polar histogram of top 50 proteins abundant in exosomes and ectosomes
(a) Western blot analysis of exosomes, ectosomes, 10K and WCL. CD81 is exclusively detected in exosomes while MMP2 is unique to ectosomes and 10K. CD63 was enriched in exosomes but was also detected in ectosomes. GM130 is absent in exosomes, ectosomes and 10K confirming the depletion of contaminating vesicles arising from apoptosis. (b) Normalised spectral counts of top 50 abundant proteins are displayed. The list is sorted by the number of times the particular protein is identified in Vesiclepedia. The numbers (outer circle) correspond to the number of studies reported in Vesiclepedia. The color scale represents the protein abundance level in terms of normalised spectral count. Top 50 abundant proteins in exosomes include CD81, flotillins and ADAM10 which are some of the proteins identified more often in Vesiclepedia. (c) Top 50 abundant proteins in ectosomes include GSTP1, RAB(s) and ribosomal proteins.
Figure 6. Functional enrichment analysis of exosomes…
Figure 6. Functional enrichment analysis of exosomes and ectosomes using FunRich
(a) Biological pathways enriched in proteins that are more than 2-fold abundant in exosomes compared to ectosomes are displayed. Proteins implicated in ESCRT, syndecan signaling and membrane trafficking are enriched in exosomes. When proteins that are more than 2-fold abundant in ectosomes compared to exosomes were analysed using FunRich, proteins implicated in gene expression and translation were enriched in ectosomes. (b) Gene Ontology-based biological processes that are enriched and depleted in proteins differentially abundant in exosomes compared to ectosomes are displayed. (c) Gene Ontology-based molecular functions that are enriched and depleted in proteins differentially abundant in ectosomes compared to exosome are displayed.
Figure 7. Integrated genomics and proteomics workflow
Figure 7. Integrated genomics and proteomics workflow
This figure depicts the overview of the proteogenomics analysis involving genomic data from exome sequencing and proteomic data derived from mass-spectrometry. Exosomes and ectosomes were isolated from SH-SY5Y cells and were subjected to label-free quantitative proteomics analysis. Exome sequencing was carried out in SH-SY5Y cells and the SNVs and INDELs detected were used to create a customised mutant protein database. Proteomics analysis was also carried on SH-SY5Y WCL samples. MS/MS spectra from exosomes, ectosomes and WCL samples were searched against the wild-type and mutant database. Two-way Venn diagrams were plotted to depict the overlap of wild-type and mutant proteins identified in two respective samples. A total of 60 and 71 mutant proteins were identified in exosomes and ectosomes, respectively. Venn diagram depicting the wild-type proteins represents all identified proteins and relative abundance is not taken into account.
Figure 8. Proteogenomics landscape of exosomes, ectosomes…
Figure 8. Proteogenomics landscape of exosomes, ectosomes and WCL of SH-SY5Y cells
This figure depicts the overview of the genomic data derived from exome sequencing and proteomic data obtained by mass spectrometry. (1) The chromosomes represent the human ideogram. (2) Represents histogram density (every 1 Mb size) of single nucleotide variations (SNVs). (3) Represents histogram density (every 1 Mb size) of insertions and deletions (INDELs) identified using exome sequencing data. (4-6) Represents mutant proteins identified by mass spectrometry in exosomes, ectosomes and WCL, respectively, when searched against mutant databases containing the SNVs and INDELS. (7) and (8) represents heat map of differentially expressed proteins in exosomes and ectosomes in comparison to WCL, whereas (9) represents heat map of differentially expressed proteins in exosomes compared to ectosomes.
Figure 9. Oncogenic potential of exosomes and…
Figure 9. Oncogenic potential of exosomes and ectosomes
(a) Genes significantly enriched in COSMIC database (p<0.05, chi-square test in different human cancer tissue types (except neuroblastoma which is not significantly enriched-represented by *)) were compared against proteins differentially abundant in exosomes and ectosomes. The normalized spectral counts of the enriched proteins were plotted as a box plot and grouped by the human cancer tissue types. The X-axis represents normalized spectral counts of significantly enriched (p<0.05, chi-square test) proteins whereas Y-axis represents cancer tissue types from COSMIC. Proteins identified in exosomes were implicated in many cancer types as compared to ectosomal proteins. (b) Genes enriched in EST database (NCBI UniGene; p<0.05, chi-square test) were compared against proteins differentially abundant in exosomes and ectosomes. The normalized spectral counts of the proteins were plotted as a box plot and grouped by the tissue types. The Y-axis represents normalized spectral counts of significantly enriched proteins whereas X-axis represents tissue types from EST database. Proteins identified in exosomes were implicated in a wide range of cancer types as compared to ectosomal proteins.
Figure 10. Cell proliferation and migration potential…
Figure 10. Cell proliferation and migration potential of exosomes and ectosomes
(a) MTS cell proliferation assay was performed with SK-N-BE2 neuroblastoma cells treated with exosomes and ectosomes derived from SH-SY5Y cells for 24 h. As a control, untreated SK-N-BE2 cells were grown. Exosomes induced a 2-fold proliferation of SK-N-BE2 cells. On the contrary, ectosomes did not induce any significant proliferation of SK-N-BE2 cells. Error bars represent standard error of the mean, n=3, * denotes significance (p<0.05). (b) Wound healing assay of neuroblastoma cell line SK-N-BE2 is displayed. Wound was created post reaching 100% confluence, and cells were treated with either exosomes or ectosomes for 24 h. Migration was assessed at 24 h after wounding. Images were taken under the 4x objective of the light microscope. Quantification of wound closure showed that exosomes induced more migration compared to ectosomes. Error bars represent standard error of the mean, n=3, * denotes significance (p<0.05). Student's t-test was used to evaluate statistically significant differences between the values.

References

    1. Bissell MJ, Radisky D. Putting tumours in context. Nat Rev Cancer. 2001;1:46–54.
    1. Thery C, Ostrowski M, Segura E. Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 2009;9:581–593.
    1. Mathivanan S, Ji H, Simpson RJ. Exosomes: Extracellular organelles important in intercellular communication. J Proteomics. 2010;73:1907–1920.
    1. Barry OP, Pratico D, Savani RC, FitzGerald GA. Modulation of monocyte-endothelial cell interactions by platelet microparticles. J Clin Invest. 1998;102:136–144.
    1. Cossetti C, Iraci N, Mercer TR, Leonardi T, Alpi E, Drago D, Alfaro-Cervello C, Saini HK, Davis MP, Schaeffer J, Vega B, Stefanini M, Zhao C, Muller W, Garcia-Verdugo JM, Mathivanan S, et al. Extracellular vesicles from neural stem cells transfer IFN-gamma via Ifngr1 to activate Stat1 signaling in target cells. Mol Cell. 2014;56:193–204.
    1. Mathivanan S. Exosomes and Shedding Microvesicles are Mediators of Intercellular Communication: How do they Communicate with the Target Cells? J Biotechnol Biomater. 2012:2.
    1. Kalra H, Simpson RJ, Ji H, Aikawa E, Altevogt P, Askenase P, Bond VC, Borras FE, Breakefield X, Budnik V, Buzas E, Camussi G, Clayton A, Cocucci E, Falcon-Perez JM, Gabrielsson S, et al. Vesiclepedia: a compendium for extracellular vesicles with continuous community annotation. PLoS Biol. 2012;10:e1001450.
    1. Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. The Journal of cell biology. 2013;200:373–383.
    1. Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19:43–51.
    1. Muralidharan-Chari V, Clancy J, Plou C, Romao M, Chavrier P, Raposo G, D'Souza-Schorey C. ARF6-regulated shedding of tumor cell-derived plasma membrane microvesicles. Curr Biol. 2009;19:1875–1885.
    1. Simpson RJ, Mathivanan S. Extracellular Microvesicles: The Need for Internationally Recognised Nomenclature and Stringent Purification Criteria. J Proteomics Bioinform. 2012;5:ii–ii.
    1. Lotvall J, Hill AF, Hochberg F, Buzas EI, Di Vizio D, Gardiner C, Gho YS, Kurochkin IV, Mathivanan S, Quesenberry P, Sahoo S, Tahara H, Wauben MH, Witwer KW, Thery C. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 2014;3:26913.
    1. Simpson RJ, Kalra H, Mathivanan S. ExoCarta as a resource for exosomal research. J Extracell Vesicles. 2012;1:18374.
    1. Gangoda L, Keerthikumar S, Fonseka P, Edgington L, Ang C, Ozcitti C, Bogyo M, Parker B, Mathivanan S. Inhibition of cathepsin proteases attenuates migration and sensitizes aggressive N-Myc amplified human neuroblastoma cells to doxorubicin. Oncotarget. 2015:5.
    1. Johnstone RM. Exosomes biological significance: A concise review. Blood Cells Mol Dis. 2006;36:315–321.
    1. Keller S, Sanderson MP, Stoeck A, Altevogt P. Exosomes: from biogenesis and secretion to biological function. Immunol Lett. 2006;107:102–108.
    1. Gangoda L, Boukouris S, Liem M, Kalra H, Mathivanan S. Extracellular vesicles including exosomes are mediators of signal transduction: Are they protective or pathogenic? Proteomics. 2015;15:260–271.
    1. Williams RL, Urbe S. The emerging shape of the ESCRT machinery. Nat Rev Mol Cell Biol. 2007;8:355–368.
    1. Simpson RJ, Lim JW, Moritz RL, Mathivanan S. Exosomes: proteomic insights and diagnostic potential. Expert Rev Proteomics. 2009;6:267–283.
    1. Mathivanan S, Lim JW, Tauro BJ, Ji H, Moritz RL, Simpson RJ. Proteomics analysis of A33 immunoaffinity-purified exosomes released from the human colon tumor cell line LIM1215 reveals a tissue-specific protein signature. Mol Cell Proteomics. 2010;9:197–208.
    1. Tauro BJ, Greening DW, Mathias RA, Ji H, Mathivanan S, Scott AM, Simpson RJ. Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods. 2012;56:293–304.
    1. Zoller M. Tetraspanins: push and pull in suppressing and promoting metastasis. Nat Rev Cancer. 2009;9:40–55.
    1. Andreu Z, Yanez-Mo M. Tetraspanins in extracellular vesicle formation and function. Front Immunol. 2014;5:442.
    1. Kowal J, Tkach M, Thery C. Biogenesis and secretion of exosomes. Curr Opin Cell Biol. 2014;29:116–125.
    1. Thery C, Boussac M, Veron P, Ricciardi-Castagnoli P, Raposo G, Garin J, Amigorena S. Proteomic analysis of dendritic cell-derived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J Immunol. 2001;166:7309–7318.
    1. Taraboletti G, D'Ascenzo S, Borsotti P, Giavazzi R, Pavan A, Dolo V. Shedding of the matrix metalloproteinases MMP-2, MMP-9, and MT1-MMP as membrane vesicle-associated components by endothelial cells. The American journal of pathology. 2002;160:673–680.
    1. Stoeck A, Keller S, Riedle S, Sanderson MP, Runz S, Le Naour F, Gutwein P, Ludwig A, Rubinstein E, Altevogt P. A role for exosomes in the constitutive and stimulus-induced ectodomain cleavage of L1 and CD44. Biochem J. 2006;393:609–618.
    1. Ochieng J, Pratap S, Khatua AK, Sakwe AM. Anchorage-independent growth of breast carcinoma cells is mediated by serum exosomes. Exp Cell Res. 2009;315:1875–1888.
    1. Demory Beckler M, Higginbotham JN, Franklin JL, Ham AJ, Halvey PJ, Imasuen IE, Whitwell C, Li M, Liebler DC, Coffey RJ. Proteomic analysis of exosomes from mutant KRAS colon cancer cells identifies intercellular transfer of mutant KRAS. Molecular & cellular proteomics : MCP. 2013;12:343–355.
    1. Al-Nedawi K, Meehan B, Micallef J, Lhotak V, May L, Guha A, Rak J. Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells. Nat Cell Biol. 2008;10:619–624.
    1. Cooke IR, Jones D, Bowen JK, Deng C, Faou P, Hall NE, Jayachandran V, Liem M, Taranto AP, Plummer KM, Mathivanan S. Proteogenomic analysis of the Venturia pirina (Pear Scab Fungus) secretome reveals potential effectors. J Proteome Res. 2014;13:3635–3644.
    1. Zhang B, Wang J, Wang X, Zhu J, Liu Q, Shi Z, Chambers MC, Zimmerman LJ, Shaddox KF, Kim S, Davies SR, Wang S, Wang P, Kinsinger CR, Rivers RC, Rodriguez H, et al. Proteogenomic characterization of human colon and rectal cancer. Nature. 2014;513:382–387.
    1. Mathivanan S, Ji H, Tauro BJ, Chen YS, Simpson RJ. Identifying mutated proteins secreted by colon cancer cell lines using mass spectrometry. J Proteomics. 2012;76 Spec No:141–149.
    1. Pugh TJ, Morozova O, Attiyeh EF, Asgharzadeh S, Wei JS, Auclair D, Carter SL, Cibulskis K, Hanna M, Kiezun A, Kim J, Lawrence MS, Lichenstein L, McKenna A, Pedamallu CS, Ramos AH, et al. The genetic landscape of high-risk neuroblastoma. Nat Genet. 2013;45:279–284.
    1. Xu H, Zhang Y, Altomare D, Pena MM, Wan F, Pirisi L, Creek KE. Six1 promotes epithelial-mesenchymal transition and malignant conversion in human papillomavirus type 16-immortalized human keratinocytes. Carcinogenesis. 2014;35:1379–1388.
    1. Li Z, Tian T, Hu X, Zhang X, Li L, Nan F, Chang Y, Wang X, Sun Z, Lv F, Zhang M. Targeting Six1 by lentivirus-mediated RNA interference inhibits colorectal cancer cell growth and invasion. Int J Clin Exp Pathol. 2014;7:631–639.
    1. Li Z, Tian T, Hu X, Zhang X, Nan F, Chang Y, Lv F, Zhang M. Six1 mediates resistance to paclitaxel in breast cancer cells. Biochem Biophys Res Commun. 2013;441:538–543.
    1. Lv H, Cui A, Sun F, Zhang Y, Li Y, Li L, Lin Z. Sineoculis homeobox homolog 1 protein as an independent biomarker for gastric adenocarcinoma. Exp Mol Pathol. 2014;97:74–80.
    1. Jin A, Xu Y, Liu S, Jin T, Li Z, Jin H, Lin L, Lin Z. Sineoculis homeobox homolog 1 protein overexpression as an independent biomarker for pancreatic ductal adenocarcinoma. Exp Mol Pathol. 2014;96:54–60.
    1. Beierle EA, Dai W, Langham MR, Jr, Copeland EM, 3rd, Chen MK. VEGF receptors are differentially expressed by neuroblastoma cells in culture. J Pediatr Surg. 2003;38:514–521.
    1. Valencia T, Joseph A, Kachroo N, Darby S, Meakin S, Gnanapragasam VJ. Role and expression of FRS2 and FRS3 in prostate cancer. BMC Cancer. 2011;11:484.
    1. Ward Y, Yap SF, Ravichandran V, Matsumura F, Ito M, Spinelli B, Kelly K. The GTP binding proteins Gem and Rad are negative regulators of the Rho-Rho kinase pathway. The Journal of cell biology. 2002;157:291–302.
    1. Leone A, Mitsiades N, Ward Y, Spinelli B, Poulaki V, Tsokos M, Kelly K. The Gem GTP-binding protein promotes morphological differentiation in neuroblastoma. Oncogene. 2001;20:3217–3225.
    1. Cantilena S, Pastorino F, Pezzolo A, Chayka O, Pistoia V, Ponzoni M, Sala A. Frizzled receptor 6 marks rare, highly tumourigenic stem-like cells in mouse and human neuroblastomas. Oncotarget. 2011;2:976–983.
    1. Dasgupta A, Alvarado CS, Xu Z, Findley HW. Expression and functional role of inhibitor-of-apoptosis protein livin (BIRC7) in neuroblastoma. Biochem Biophys Res Commun. 2010;400:53–59.
    1. Diergaarde B, Brand R, Lamb J, Cheong SY, Stello K, Barmada MM, Feingold E, Whitcomb DC. Pooling-based genome-wide association study implicates gamma-glutamyltransferase 1 (GGT1) gene in pancreatic carcinogenesis. Pancreatology. 2010;10:194–200.
    1. Lee SJ, Chae YS, Kim JG, Kim WW, Jung JH, Park HY, Jeong JY, Park JY, Jung HJ, Kwon TH. AQP5 expression predicts survival in patients with early breast cancer. Ann Surg Oncol. 2014;21:375–383.
    1. Arnason T, Pino MS, Yilmaz O, Kirley SD, Rueda BR, Chung DC, Zukerberg LR. Cables1 is a tumor suppressor gene that regulates intestinal tumor progression in Apc(Min) mice. Cancer Biol Ther. 2013;14:672–678.
    1. Wang Z, Shen D, Parsons DW, Bardelli A, Sager J, Szabo S, Ptak J, Silliman N, Peters BA, van der Heijden MS, Parmigiani G, Yan H, Wang TL, Riggins G, Powell SM, Willson JK, et al. Mutational analysis of the tyrosine phosphatome in colorectal cancers. Science. 2004;304:1164–1166.
    1. Fan Y, Cheng J, Vasudevan SA, Patel RH, Liang L, Xu X, Zhao Y, Jia W, Lu F, Zhang H, Nuchtern JG, Kim ES, Yang J. TAK1 inhibitor 5Z-7-oxozeaenol sensitizes neuroblastoma to chemotherapy. Apoptosis : an international journal on programmed cell death. 2013;18:1224–1234.
    1. Muralidharan-Chari V, Clancy JW, Sedgwick A, D'Souza-Schorey C. Microvesicles: mediators of extracellular communication during cancer progression. J Cell Sci. 2010;123:1603–1611.
    1. Boukouris S, Mathivanan S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteomics Clin Appl. 2015
    1. Mathivanan S. Quest for Cancer Biomarkers: Assaying Mutant Proteins and RNA that Provides the Much Needed Specificity. J Proteomics Bioinform. 2012;5:xiii–xvii.
    1. Kalra H, Adda CG, Liem M, Ang CS, Mechler A, Simpson RJ, Hulett MD, Mathivanan S. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. Proteomics. 2013;13:3354–3364.
    1. Pruitt KD, Tatusova T, Maglott DR. NCBI reference sequences (RefSeq): a curated non-redundant sequence database of genomes, transcripts and proteins. Nucleic Acids Res. 2007;35(Database issue):D61–65.
    1. Craig R, Beavis RC. TANDEM: matching proteins with tandem mass spectra. Bioinformatics. 2004;20:1466–1467.
    1. Chen YS, Mathias RA, Mathivanan S, Kapp EA, Moritz RL, Zhu HJ, Simpson RJ. Proteomics profiling of Madin-Darby canine kidney plasma membranes reveals Wnt-5a involvement during oncogenic H-Ras/TGF-beta-mediated epithelial-mesenchymal transition. Mol Cell Proteomics. 2011;10:M110 001131.
    1. Forbes SA, Beare D, Gunasekaran P, Leung K, Bindal N, Boutselakis H, Ding M, Bamford S, Cole C, Ward S, Kok CY, Jia M, De T, Teague JW, Stratton MR, McDermott U, et al. COSMIC: exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015;43(Database issue):D805–811.
    1. Oortveld MA, Keerthikumar S, Oti M, Nijhof B, Fernandes AC, Kochinke K, Castells-Nobau A, van Engelen E, Ellenkamp T, Eshuis L, Galy A, van Bokhoven H, Habermann B, Brunner HG, Zweier C, Verstreken P, et al. Human intellectual disability genes form conserved functional modules in Drosophila. PLoS Genet. 2013;9:e1003911.

Source: PubMed

3
Subscribe