MicroPOTS Analysis of Barrett’s Esophageal Cell Line Models Identifies Proteomic Changes after Physiologic and Radiation StressClick to copy article linkArticle link copied!
- Kenneth WekeKenneth WekeUniversity of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, PolandMore by Kenneth Weke
- Ashita SinghAshita SinghInstitute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K.Research Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, 656 53 Brno, Czech RepublicMore by Ashita Singh
- Naomi UwugiarenNaomi UwugiarenUniversity of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, PolandMore by Naomi Uwugiaren
- Javier A. AlfaroJavier A. AlfaroUniversity of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, PolandInstitute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K.More by Javier A. Alfaro
- Tongjie WangTongjie WangInstitute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K.More by Tongjie Wang
- Ted R. HuppTed R. HuppUniversity of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, PolandInstitute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K.More by Ted R. Hupp
- J. Robert O’NeillJ. Robert O’NeillInstitute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland EH4 2XR, U.K.Cambridge Oesophagogastric Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, U.K.More by J. Robert O’Neill
- Borek VojtesekBorek VojtesekResearch Centre for Applied Molecular Oncology (RECAMO), Masaryk Memorial Cancer Institute, 656 53 Brno, Czech RepublicMore by Borek Vojtesek
- David R. Goodlett*David R. Goodlett*Email: [email protected]University of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, PolandUniversity of Victoria − Genome British Columbia Proteomics Centre, Victoria, BC V8Z 7X8, CanadaDepartment of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8P 5C2, CanadaMore by David R. Goodlett
- Sarah M. WilliamsSarah M. WilliamsEnvironmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United StatesMore by Sarah M. Williams
- Mowei ZhouMowei ZhouEnvironmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United StatesMore by Mowei Zhou
- Ryan T. KellyRyan T. KellyEnvironmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United StatesMore by Ryan T. Kelly
- Ying ZhuYing ZhuEnvironmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99354, United StatesMore by Ying Zhu
- Irena Dapic*Irena Dapic*Email: [email protected]University of Gdansk, International Centre for Cancer Vaccine Science, ul. Kładki 24, 80-822 Gdansk, PolandMore by Irena Dapic
Abstract
Moving from macroscale preparative systems in proteomics to micro- and nanotechnologies offers researchers the ability to deeply profile smaller numbers of cells that are more likely to be encountered in clinical settings. Herein a recently developed microscale proteomic method, microdroplet processing in one pot for trace samples (microPOTS), was employed to identify proteomic changes in ∼200 Barrett’s esophageal cells following physiologic and radiation stress exposure. From this small population of cells, microPOTS confidently identified >1500 protein groups, and achieved a high reproducibility with a Pearson’s correlation coefficient value of R > 0.9 and over 50% protein overlap from replicates. A Barrett’s cell line model treated with either lithocholic acid (LCA) or X-ray had 21 (e.g., ASNS, RALY, FAM120A, UBE2M, IDH1, ESD) and 32 (e.g., GLUL, CALU, SH3BGRL3, S100A9, FKBP3, AGR2) overexpressed proteins, respectively, compared to the untreated set. These results demonstrate the ability of microPOTS to routinely identify and quantify differentially expressed proteins from limited numbers of cells.
1. Introduction
2. Experimental Section
2.1. Materials
2.2. Cell Culture
2.3. Cells Treatment with LCA and X-ray
2.4. Proteomic Sample Preparation in Microwells
2.5. LC-MS/MS Analysis
2.6. Data and Statistical Analysis
2.7. Protein Annotation and Assessment of Physicochemical Aspects
3. Results and Discussion
3.1. Analyzing Barrett’s Esophageal Cell Samples by MicroPOTS
3.2. Protein Identification from Different CP-A Genotypes and Stress Conditions
3.3. Evaluation of Protein Extraction Efficiency and Reproducibility
3.4. Comparison of MicroPOTS Data to Bulk Proteomics Data Set
3.5. Comparison of Physicochemical Aspects of Identified Proteins
3.6. Effect of Stress on Protein Expression
Conclusion
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jproteome.0c00629.
Figure S1: Venn diagram illustrating the shared number of identified proteins among different sample types, as well as proteins that are unique to each sample type; Figure S2: Number of missed cleavages are shown for all replicates and are expressed in percentage; Figure S3: Qualitative assessment of reproducibility of the microPOTS system; Figure S4: Quantitative assessment of reproducibility of the microPOTS system; Table S1: Median coefficient of variation (CV) for quantile normalized protein LFQ values for each sample type; Box S1: Sample preparation and LC-MS analysis for bulk proteomics data set; Box S2: Bulk data analysis and comparison to microPOTS data; Figure S5: Scatter plot with associated Pearson’s correlation coefficient (R = 0.625) between microPOTS and bulk proteomics for all overlapping 1066 proteins that were identified; Figure S6: Assessment of physicochemical characteristics; Figure S7: Assessment of physicochemical characteristics; Figure S8: Subcellular localization; Table S2: List of differentially expressed proteins between CP-A dKO cells with or without LCA treatment; Table S3: List of differentially expressed proteins between CP-A dKO cells with or without X-ray treatment (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This work was funded by the International Research Agenda’s Program of the Foundation for Polish Science (MAB/2017/03). The “International Centre for Cancer Vaccine Science” project is carried out within the International Research Agendas programme of the Foundation for Polish Science cofinanced by the European Union under the European Regional Development Fund. The authors would also like to thank the CI-TASK, Gdansk and the PL-Grid Infrastructure, Poland for providing their hardware and software resources. The University of Victoria-Genome BC Proteomics Centre is grateful to Genome Canada and Genome British Columbia for financial support for Genomics Technology Platforms (GTP) funding for operations and technology development (264PRO). The experiment was performed using EMSL (grid.436923.9), a DOE Office of Science User Facility sponsored by the Office of Biological and Environmental Research and located at PNNL. The Brno CP-A study was supported by the European Regional Development Fund – Project ENOCH (No. CZ.02.1.01/0.0/0.0/16_019/0000868) and by the Ministry of Health Development of Research Organization, MH CZ – DRO (MMCI, 00209805).
References
This article references 57 other publications.
- 1Mann, M.; Hendrickson, R. C.; Pandey, A. Analysis of Proteins and Proteomes by Mass Spectrometry. Annu. Rev. Biochem. 2001, 70, 437– 473, DOI: 10.1146/annurev.biochem.70.1.437Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXlsVehtL4%253D&md5=1bd749a47bfc1ec30226dce9a820aef6Analysis of proteins and proteomes by mass spectrometryMann, Matthias; Hendrickson, Ronald C.; Pandey, AkhileshAnnual Review of Biochemistry (2001), 70 (), 437-473CODEN: ARBOAW; ISSN:0066-4154. (Annual Reviews Inc.)A review with 91 refs. A decade after the discovery of electrospray and matrix-assisted laser desorption ionization (MALDI), methods that finally allowed gentle ionization of large biomols., mass spectrometry has become a powerful tool in protein anal. and the key technol. in the emerging field of proteomics. The success of mass spectrometry is driven both by innovative instrumentation designs, esp. those operating on the time-of-flight or ion-trapping principles, and by large-scale biochem. strategies, which use mass spectrometry to detect the isolated proteins. Any human protein can now be identified directly from genome databases on the basis of minimal data derived by mass spectrometry. As has already happened in genomics, increased automation of sample handling, anal., and the interpretation of results will generate an avalanche of qual. and quant. proteomic data. Protein-protein interactions can be analyzed directly by pptn. of a tagged bait followed by mass spectrometric identification of its binding partners. By these and similar strategies, entire protein complexes, signaling pathways, and whole organelles are being characterized. Posttranslational modifications remain difficult to analyze but are starting to yield to generic strategies.
- 2Wasinger, V. C.; Cordwell, S. J.; Cerpa-Poljak, A.; Yan, J. X.; Gooley, A. A.; Wilkins, M. R.; Duncan, M. W.; Harris, R.; Williams, K. L.; Humphery-Smith, I. Progress with Gene-Product Mapping of the Mollicutes: Mycoplasma Genitalium. Electrophoresis 1995, 16 (7), 1090– 1094, DOI: 10.1002/elps.11501601185Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXntF2lsLg%253D&md5=11e7acd04dc0eaf17ec7f99996a0c654Progress with gene-product mapping of the Mollicutes: Mycoplasma genitaliumWasinger, Valerie C.; Cordwell, Stuart J.; Cerpa-Poljak, Anne; Yan, Jun X.; Gooley, Andrew A.; Wilkins, Marc R.; Duncan, Mark W.; Harris, Ray; Williams, Keith L.; Humphery-Smith, IanElectrophoresis (1995), 16 (7), 1090-4CODEN: ELCTDN; ISSN:0173-0835. (VCH)A protein map of the smallest known self-replicating organism Mycoplasma genitalium (Class: Mollicutes) revealed a high proportion of acidic proteins. Amino acid compn. was used to putatively identify or provide unique parameters for 50 gene products sepd. by two-dimensional gel electrophoresis. A further 19 proteins were subjected to peptide-mass fingerprinting using matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry and 4 were subjected to N-terminal Edman degrdn. The majority of M. genitalium proteins remain uncharacterized. However, the combined approach of amino acid anal. and peptide-mass fingerprinting allowed gene products to be linked to homologous genes in a variety of organisms. This allowed proteins to be identified prior to detection of their resp. genes via the M. genitalium sequencing initiative. The principle of 'hierarchical' anal. for the mass screening of proteins and the anal. of microbial genomes via their protein complement or 'proteome' is detailed. Here, characterization of gene products depends upon the quickest and most economical technols. being employed initially to det. if a large no. of proteins are already present in both homologous and heterologous species databases. Initial screening, which lends itself to automation and robotics, can then be followed by more time and cost intensive procedures, when necessary.
- 3Zhang, Y.; Fonslow, B. R.; Shan, B.; Baek, M.-C.; Yates, J. R. Protein Analysis by Shotgun/Bottom-up Proteomics. Chem. Rev. 2013, 113 (4), 2343– 2394, DOI: 10.1021/cr3003533Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjtVClu7g%253D&md5=a27c72f72bc6fa2f827725d2e71df241Protein Analysis by Shotgun/Bottom-up ProteomicsZhang, Yaoyang; Fonslow, Bryan R.; Shan, Bing; Baek, Moon-Chang; Yates, John R.Chemical Reviews (Washington, DC, United States) (2013), 113 (4), 2343-2394CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review.
- 4Mayne, J.; Ning, Z.; Zhang, X.; Starr, A. E.; Chen, R.; Deeke, S.; Chiang, C.-K.; Xu, B.; Wen, M.; Cheng, K.; Seebun, D.; Star, A.; Moore, J. I.; Figeys, D. Bottom-Up Proteomics (2013–2015): Keeping up in the Era of Systems Biology. Anal. Chem. 2016, 88 (1), 95– 121, DOI: 10.1021/acs.analchem.5b04230Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvVSlsLvJ&md5=291844fae9e91fc1b1d219948ef7d3c4Bottom-Up Proteomics (2013-2015): Keeping up in the Era of Systems BiologyMayne, Janice; Ning, Zhibin; Zhang, Xu; Starr, Amanda E.; Chen, Rui; Deeke, Shelley; Chiang, Cheng-Kang; Xu, Bo; Wen, Ming; Cheng, Kai; Seebun, Deeptee; Star, Alexandra; Moore, Jasmine I.; Figeys, DanielAnalytical Chemistry (Washington, DC, United States) (2016), 88 (1), 95-121CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A review. Over 17,000 papers related to proteomics have been published since the authors' 2011-2013 review. A subset are true gems reporting amazing applications of proteomics to better understand protein interactions, pathways, dynamic signaling and the human proteome, to name a few. While the authors have clearly seen more attention paid to data quality, and the proper exptl. design in manuscripts reporting biol. applications of proteomics, more improvements are needed. Technol. development still remains a strong driving force of proteomics, making up a large portion of these papers. As in any maturing field, some areas of technol. development in proteomics are satg. However, many new areas in proteomics are burgeoning. Examples include the development of enrichment materials to study different protein modifications, approaches for protein quantification, data independent acquisition (DIA) methods, bioinformatics and the rapid developments in metaproteomics. The acceptance of proteomics in the biol. community really depends on its applications. For instance, protein interaction mapping, post-translational modification (PTM) analyses and protein Atlas are routinely used by the biol. community. However, other aspects of proteomics are still under appreciated including biomarker discovery and studies of diseases from tissues. In part, this is due to previous and, to some extent, still ongoing overstatements from underpowered studies that reported unsubstantiated biol. conclusions. Very large-scale, good quality, quant. proteomic studies are now tech. feasible and ongoing in a few labs. around the world. However, few bioinformatics tools are capable of handling these large studies. Moreover, proteomics is increasingly integrated in expts. involving multiple -omics datasets obtained across large nos. of biol. replicates. "Big data" challenges are clearly appearing in proteomics. This biannual review is focused on technol. trends, which the authors believe are important in proteomics. The authors continue to foresee that technol. and bioinformatics development will remain important areas of research in proteomics.
- 5Fagerberg, L.; Strömberg, S.; El-Obeid, A.; Gry, M.; Nilsson, K.; Uhlen, M.; Ponten, F.; Asplund, A. Large-Scale Protein Profiling in Human Cell Lines Using Antibody-Based Proteomics. J. Proteome Res. 2011, 10 (9), 4066– 4075, DOI: 10.1021/pr200259vGoogle Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXptFKlsr4%253D&md5=38f0615cccc16390ae4f6be6fd806c85Large-Scale Protein Profiling in Human Cell Lines Using Antibody-Based ProteomicsFagerberg, Linn; Stromberg, Sara; El-Obeid, Adila; Gry, Marcus; Nilsson, Kenneth; Uhlen, Mathias; Ponten, Fredrik; Asplund, AnnaJournal of Proteome Research (2011), 10 (9), 4066-4075CODEN: JPROBS; ISSN:1535-3893. (American Chemical Society)Human cancer cell lines grown in vitro are frequently used to decipher basic cell biol. phenomena and to also specifically study different forms of cancer. Here the authors present the first large-scale study of protein expression patterns in cell lines using an antibody-based proteomics approach. The authors analyzed the expression pattern of 5436 proteins in 45 different cell lines using hierarchical clustering, principal component anal., and two-group comparisons for the identification of differentially expressed proteins. The authors' results show that immunohistochem. detd. protein profiles can categorize cell lines into groups that overall reflect the tumor tissue of origin and that hematol. cell lines appear to retain their protein profiles to a higher degree than cell lines established from solid tumors. The two-group comparisons reveal well-characterized proteins as well as previously unstudied proteins that could be of potential interest for further investigations. Moreover, multiple myeloma cells and cells of myeloid origin were found to share a protein profile, relative to the protein profile of lymphoid leukemia and lymphoma cells, possibly reflecting their common dependency of bone marrow microenvironment. This work also provides an extensive list of antibodies, for which high-resoln. images as well as validation data are available on the Human Protein Atlas (www.proteinatlas.org), that are of potential use in cell line studies.
- 6Branca, R. M. M.; Orre, L. M.; Johansson, H. J.; Granholm, V.; Huss, M.; Pérez-Bercoff, Å.; Forshed, J.; Käll, L.; Lehtiö, J. HiRIEF LC-MS Enables Deep Proteome Coverage and Unbiased Proteogenomics. Nat. Methods 2014, 11 (1), 59– 62, DOI: 10.1038/nmeth.2732Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslygsLnE&md5=acdad94163f52313a336a41ccfed44c4HiRIEF LC-MS enables deep proteome coverage and unbiased proteogenomicsBranca, Rui M. M.; Orre, Lukas M.; Johansson, Henrik J.; Granholm, Viktor; Huss, Mikael; Perez-Bercoff, Aasa; Forshed, Jenny; Kaell, Lukas; Lehtioe, JanneNature Methods (2014), 11 (1), 59-62CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)We present a liq. chromatog.-mass spectrometry (LC-MS)-based method permitting unbiased (gene prediction-independent) genome-wide discovery of protein-coding loci in higher eukaryotes. Using high-resoln. isoelec. focusing (HiRIEF) at the peptide level in the 3.7-5.0 pH range and accurate peptide isoelec. point (pI) prediction, we probed the six-reading-frame translation of the human and mouse genomes and identified 98 and 52 previously undiscovered protein-coding loci, resp. The method also enabled deep proteome coverage, identifying 13,078 human and 10,637 mouse proteins.
- 7Wang, G.; Brennan, C.; Rook, M.; Wolfe, J. L.; Leo, C.; Chin, L.; Pan, H.; Liu, W.-H.; Price, B.; Makrigiorgos, G. M. Balanced-PCR Amplification Allows Unbiased Identification of Genomic Copy Changes in Minute Cell and Tissue Samples. Nucleic Acids Res. 2004, 32 (9), e76, DOI: 10.1093/nar/gnh070Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksVahsLk%253D&md5=e87e9779bb0d6186644d4f7b394d1448Balanced-PCR amplification allows unbiased identification of genomic copy changes in minute cell and tissue samplesWang, Gang; Brennan, Cameron; Rook, Martha; Wolfe, Jia Liu; Leo, Christopher; Chin, Lynda; Pan, Hongjie; Liu, Wei-Hua; Price, Brendan; Makrigiorgos, G. MikeNucleic Acids Research (2004), 32 (9), e76/1-e76/10CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Anal. of genomic DNA derived from cells and fresh or fixed tissues often requires whole genome amplification prior to microarray screening. Tech. hurdles to this process are the introduction of amplification bias and/or the inhibitory effects of formalin fixation on DNA amplification. Here we demonstrate a balanced-PCR procedure that allows unbiased amplification of genomic DNA from fresh or modestly degraded paraffin-embedded DNA samples. Following digestion and ligation of a target and a control genome with distinct linkers, the two are mixed and amplified in a single PCR, thereby avoiding biases assocd. with PCR satn. and impurities. We demonstrate genome-wide retention of allelic differences following balanced-PCR amplification of DNA from breast cancer and normal human cells and genomic profiling by array-CGH (cDNA arrays, 100 kb resoln.) and by real-time PCR (single gene resoln.). Comparison of balanced-PCR with multiple displacement amplification (MDA) demonstrates equiv. performance between the two when intact genomic DNA is used. When DNA from paraffin-embedded samples is used, balanced PCR overcomes problems assocd. with modest DNA degrdn. and produces unbiased amplification whereas MDA does not. Balanced-PCR allows amplification and recovery of modestly degraded genomic DNA for subsequent retrospective anal. of human tumors with known outcomes.
- 8Wang, N.; Xu, M.; Wang, P.; Li, L. Development of Mass Spectrometry-Based Shotgun Method for Proteome Analysis of 500 to 5000 Cancer Cells. Anal. Chem. 2010, 82 (6), 2262– 2271, DOI: 10.1021/ac9023022Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptlGrtA%253D%253D&md5=89ac83bbe62a4e164961925b870a13e4Development of Mass Spectrometry-Based Shotgun Method for Proteome Analysis of 500 to 5000 Cancer CellsWang, Nan; Xu, Mingguo; Wang, Peng; Li, LiangAnalytical Chemistry (Washington, DC, United States) (2010), 82 (6), 2262-2271CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A shotgun proteome anal. method and its performance for protein identification from 500 to 5000 cells are described. Sample prepn., which was done in one tube, involved the use of a surfactant (NP-40) for cell lysis, followed by acetone pptn. of the proteins. The resulting protein pellet was washed with cold acetone to remove remaining surfactant, and the pellet was then solubilized in NH4HCO3. After trypsin digestion of the proteins, the digest was analyzed by the use of nanoflow liq. chromatog. (LC) quadrupole time-of-flight mass spectrometry (QTOF MS). Sample injection and gradient speed in running LC QTOF MS were optimized. It was shown that this method could identify an av. (n = 3) of 167±21, 237±30, 491±63, and 619±59 proteins from 500, 1000, 2500, and 5000 MCF-7 breast cancer cells, resp. To demonstrate the potential use of this method for generating proteome profile from circulating tumor cells (CTCs) isolated from human blood, a healthy human blood sample was spiked with MCF-7 cells, and this mixt. was processed and then subjected to antibody tagging of the MCF-7 cells. The tagged cells were sorted and collected using flow cytometry. The proteome profiles of small nos. of cells isolated in this way were similar to those of the original MCF-7 cells, suggesting the possibility of the use of this method for cell typing of CTCs.
- 9Parapatics, K.; Huber, M. L.; Lehmann, D.; Knoll, C.; Superti-Furga, G.; Bennett, K. L.; Rudashevskaya, E. L. Proteomic Analysis of Low Quantities of Cellular Material in the Range Obtainable from Scarce Patient Samples. J. Integr. OMICS 2015, 5 (1), 30– 43–43, DOI: 10.5584/jiomics.v5i1.172Google ScholarThere is no corresponding record for this reference.
- 10Clair, G.; Piehowski, P. D.; Nicola, T.; Kitzmiller, J. A.; Huang, E. L.; Zink, E. M.; Sontag, R. L.; Orton, D. J.; Moore, R. J.; Carson, J. P.; Smith, R. D.; Whitsett, J. A.; Corley, R. A.; Ambalavanan, N.; Ansong, C. Spatially-Resolved Proteomics: Rapid Quantitative Analysis of Laser Capture Microdissected Alveolar Tissue Samples. Sci. Rep. 2016, 6, 6, DOI: 10.1038/srep39223Google ScholarThere is no corresponding record for this reference.
- 11Coscia, F.; Doll, S.; Bech, J. M.; Mund, A.; Lengyel, E.; Lindebjerg, J.; Madsen, G. I.; Moreira, J. M. A.; Mann, M. A Streamlined Mass Spectrometry-Based Proteomics Workflow for Large Scale FFPE Tissue Analysis. bioRxiv , Sep 23, 2019, 779009. DOI: 10.1101/779009 .Google ScholarThere is no corresponding record for this reference.
- 12Martin, J. G.; Rejtar, T.; Martin, S. A. Integrated Microscale Analysis System for Targeted Liquid Chromatography Mass Spectrometry Proteomics on Limited Amounts of Enriched Cell Populations. Anal. Chem. 2013, 85 (22), 10680– 10685, DOI: 10.1021/ac401937cGoogle Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFGkurbN&md5=7fd20d18e145d970a56b1aadfe651f89Integrated Microscale Analysis System for Targeted Liquid Chromatography Mass Spectrometry Proteomics on Limited Amounts of Enriched Cell PopulationsMartin, Jeffrey G.; Rejtar, Tomas; Martin, Stephen A.Analytical Chemistry (Washington, DC, United States) (2013), 85 (22), 10680-10685CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Limited samples, such as those that are in vivo sourced via biopsy, are closely representative of biol. systems and contain valuable information for drug discovery. However, these precious samples are often heterogeneous and require cellular prefractionation prior to proteomic anal. to isolate specific subpopulations of interest. Enriched cells from in vivo samples are often very limited (<104 cells) and pose a significant challenge to proteomic nanoliquid chromatog. mass spectrometry (nanoLCMS) sample prepn. To enable the streamlined anal. of these limited samples, the authors have developed an online cell enrichment, microscale sample prepn., nanoLCMS proteomics workflow by integrating fluorescence activated cell sorting (FACS), focused ultrasonication, microfluidics, immobilized trypsin digestion, and nanoLCMS. To assess the performance of the online FACS-Chip-LCMS workflow, 5000 fluorescent labeled cells were enriched from a 5% heterogeneous cell population and processed for LCMS proteomics in <2 h. Within these 5000 enriched cells, 30 peptides corresponding to 17 proteins spanning >4 orders of magnitude of cellular abundance were quantified using a QExactive MS. The results from the online FACS-Chip-LCMS workflow starting from 5000 enriched cells were directly compared to results from a traditional macroscale sample prepn. workflow starting from 2.0 × 106 cells. The microscale FACS-Chip-LCMS workflow demonstrated high cellular enrichment efficiency and high peptide recovery across the wide dynamic range of targeted peptides. Overall the microscale FACS-Chip-LCMS workflow showed effectiveness in efficiently prepg. limited amts. of FACS enriched cells in an online manner for proteomic LCMS.
- 13Yamaguchi, H.; Miyazaki, M.; Honda, T.; Briones-Nagata, M. P.; Arima, K.; Maeda, H. Rapid and Efficient Proteolysis for Proteomic Analysis by Protease-Immobilized Microreactor. Electrophoresis 2009, 30 (18), 3257– 3264, DOI: 10.1002/elps.200900134Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFGqsLjK&md5=9870da1a12d77e6e302a53e76019fd79Rapid and efficient proteolysis for proteomic analysis by protease-immobilized microreactorYamaguchi, Hiroshi; Miyazaki, Masaya; Honda, Takeshi; Briones-Nagata, Maria Portia; Arima, Kazunari; Maeda, HideakiElectrophoresis (2009), 30 (18), 3257-3264CODEN: ELCTDN; ISSN:0173-0835. (Wiley-VCH Verlag GmbH & Co. KGaA)Proteolysis is an important part of protein identification in proteomics anal. The conventional method of in-soln. digestion of proteins is time-consuming and has limited sensitivity. In this study, trypsin- or α-chymotrypsin-immobilized microreactors prepd. by a microfluidics-based enzyme-immobilization technique were studied for rapid sample prepn. in proteomic anal. The kinetic studies for hydrolysis of substrate by microreactors revealed that immobilized proteases had higher hydrolytic efficiency than those performed by in-soln. digestion. The performance of the microreactors was evaluated by digesting cytochrome c and bovine serum albumin (BSA). Protein digestion was achieved within a short period of time (∼5 min) at 30° without any complicated redn. and alkylation procedures. The efficiency of digestion by trypsin-immobilized reactor was evaluated by analyzing the sequence coverage, which was 47 and 12% for cytochrome c and BSA, resp. These values were higher than those performed by in-soln. digestion. Besides, because of higher stability against high concn. of denaturant, the microreactors can be useful for immediate digestion of the denatured protein. In the present study, the authors propose a protease-immobilized microreactor digestion method, which can utilize as a proteome technique for biol. and clin. research.
- 14Myers, S. A.; Rhoads, A.; Cocco, A. R.; Peckner, R.; Haber, A. L.; Schweitzer, L. D.; Krug, K.; Mani, D. R.; Clauser, K. R.; Rozenblatt-Rosen, O.; Hacohen, N.; Regev, A.; Carr, S. A. Streamlined Protocol for Deep Proteomic Profiling of FAC-Sorted Cells and Its Application to Freshly Isolated Murine Immune Cells. Mol. Cell. Proteomics 2019, 18 (5), 995– 1009, DOI: 10.1074/mcp.RA118.001259Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1OhsbbP&md5=c4a2707f8e65bcc5fd9462e4952b5e06Streamlined protocol for deep proteomic profiling of FAC-sorted cells and its application to freshly isolated murine immune cellsMyers, Samuel A.; Rhoads, Andrew; Cocco, Alexandra R.; Peckner, Ryan; Haber, Adam L.; Schweitzer, Lawrence D.; Krug, Karsten; Mani, D. R.; Clauser, Karl R.; Rozenblatt-Rosen, Orit; Hacohen, Nir; Regev, Aviv; Carr, Steven A.Molecular & Cellular Proteomics (2019), 18 (5), 995-1009CODEN: MCPOBS; ISSN:1535-9484. (American Society for Biochemistry and Molecular Biology)Proteomic profiling describes the mol. landscape of proteins in cells immediately available to sense, transduce, and enact the appropriate responses to extracellular queues. Transcriptional profiling has proven invaluable to our understanding of cellular responses; however, insights may be lost as mounting evidence suggests transcript levels only moderately correlate with protein levels in steady state cells. Mass spectrometry-based quant. proteomics is a well-suited and widely used anal. tool for studying global protein abundances. Typical proteomic workflows are often limited by the amt. of sample input that is required for deep and quant. proteome profiling. This is esp. true if the cells of interest need to be purified by fluorescence-activated cell sorting (FACS) and one wants to avoid ex vivo culturing. To address this need, we developed an easy to implement, streamlined workflow that enables quant. proteome profiling from roughly 2μg of protein input per exptl. condition. Utilizing a combination of facile cell collection from cell sorting, solid-state isobaric labeling and multiplexing of peptides, and small-scale fractionation, we profiled the proteomes of 12 freshly isolated, primary murine immune cell types. Analyzing half of the 3e5 cells collected per cell type, we quantified over 7000 proteins across 12 key immune cell populations directly from their resident tissues. We show that low input proteomics is precise, and the data generated accurately reflects many aspects of known immunol., while expanding the list of cell-type specific proteins across the cell types profiled. The low input proteomics methods we developed are readily adaptable and broadly applicable to any cell or sample types and should enable proteome profiling in systems previously unattainable.
- 15Gao, W.; Li, H.; Liu, L.; Huang, P.; Wang, Z.; Chen, W.; Ye, M.; Yu, X.; Tian, R. An Integrated Strategy for High-Sensitive and Multi-Level Glycoproteome Analysis from Low Micrograms of Protein Samples. J. Chromatogr. A 2019, 1600, 46– 54, DOI: 10.1016/j.chroma.2019.04.041Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXosVyjsrg%253D&md5=d483a4c2b76420673a2b34063171c9b4An integrated strategy for high-sensitive and multi-level glycoproteome analysis from low micrograms of protein samplesGao, Weina; Li, Hongjie; Liu, Liping; Huang, Peiwu; Wang, Zhikun; Chen, Wendong; Ye, Mingliang; Yu, Xiaofang; Tian, RuijunJournal of Chromatography A (2019), 1600 (), 46-54CODEN: JCRAEY; ISSN:0021-9673. (Elsevier B.V.)Glycosylation, as a biol. important protein post-translational modification, often alters on both glycosites and glycans, simultaneously. However, most of current approaches focused on biased profiling of either glycosites or glycans, and limited by time-consuming process and milligrams of starting protein material. We describe here a simple and integrated spintip-based glycoproteomics technol. (termed Glyco-SISPROT) for achieving a comprehensive view of glycoproteome with shorter sample processing time and low microgram starting material. By carefully integrating and optimizing SCX, C18 and Con A (Con A) packing material and their combination in spintip format, both predigested peptides and protein lysates could be processed by Glyco-SISPROT with high efficiency. More importantly, deglycopeptide, intact glycopeptide and glycans released by multiple glycosidases could be readily collected from the same Glyco-SISPROT workflow for LC-MS anal. In total, above 1850 glycosites in ∼1770 unique deglycopeptides were characterized from mouse liver by using either 100 μg of predigested peptides or directly using 100 μg of protein lysates, in which about 30% of glycosites were released by both PNGase F and Endos. To the best of our knowledge, this approach should be one of the most comprehensive glycoproteomic approaches by using limited protein starting material. One significant benefit of Glyco-SISPROT is that whole processing time is dramatically reduced from a few days to less than 6 h with good reproducibility when protein lysates were directly processed by Glyco-SISPROT. We expect that this method will be suitable for multi-level glycoproteome anal. of rare biol. samples with high sensitivity.
- 16Chen, W.; Wang, S.; Adhikari, S.; Deng, Z.; Wang, L.; Chen, L.; Ke, M.; Yang, P.; Tian, R. Simple and Integrated Spintip-Based Technology Applied for Deep Proteome Profiling. Anal. Chem. 2016, 88 (9), 4864– 4871, DOI: 10.1021/acs.analchem.6b00631Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xls1Kis7w%253D&md5=8d60eba33022626272064cf060d9fa1eSimple and Integrated Spintip-Based Technology Applied for Deep Proteome ProfilingChen, Wendong; Wang, Shuai; Adhikari, Subash; Deng, Zuhui; Wang, Lingjue; Chen, Lan; Ke, Mi; Yang, Pengyuan; Tian, RuijunAnalytical Chemistry (Washington, DC, United States) (2016), 88 (9), 4864-4871CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Great efforts have been taken for developing high-sensitive mass spectrometry (MS)-based proteomic technologies, among which sample prepn. is one of the major focus. Here, a simple and integrated spintip-based proteomics technol. (SISPROT) consisting of strong cation exchange beads and C18 disk in one pipet tip was developed. Both proteomics sample prepn. steps, including protein preconcn., redn., alkylation, and digestion, and reversed phase (RP)-based desalting and high-pH RP-based peptide fractionation can be achieved in a fully integrated manner for the first time. This easy-to-use technol. achieved high sensitivity with negligible sample loss. Proteomic anal. of 2000 HEK 293 cells readily identified 1270 proteins within 1.4 h of MS time, while 7826 proteins were identified when 100000 cells were processed and analyzed within only 22 h of MS time. More importantly, the SISPROT can be easily multiplexed on a std. centrifuge with good reproducibility (Pearson correlation coeff. > 0.98) for both single-shot anal. and deep proteome profiling with five-step high-pH RP fractionation. The SISPROT was exemplified by the triplicate anal. of 100000 stem cells from human exfoliated deciduous teeth (SHED). This led to the identification of 9078 proteins contg. 3771 annotated membrane proteins, which was the largest proteome data set for dental stem cells reported to date. We expect that the SISPROT will be well suited for deep proteome profiling for fewer than 100000 cells and applied for translational studies where multiplexed technol. with good label-free quantification precision is required.
- 17Dou, M.; Zhu, Y.; Liyu, A.; Liang, Y.; Chen, J.; Piehowski, P. D.; Xu, K.; Zhao, R.; Moore, R. J.; Atkinson, M. A.; Mathews, C. E.; Qian, W.-J.; Kelly, R. T. Nanowell-Mediated Two-Dimensional Liquid Chromatography Enables Deep Proteome Profiling of < 1000 Mammalian Cells. Chem. Sci. 2018, 9 (34), 6944– 6951, DOI: 10.1039/C8SC02680GGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlCmtbbF&md5=1ec9556a474d130acc404d70f7a6a838Nanowell-mediated two-dimensional liquid chromatography enables deep proteome profiling of <1000 mammalian cellsDou, Maowei; Zhu, Ying; Liyu, Andrey; Liang, Yiran; Chen, Jing; Piehowski, Paul D.; Xu, Kerui; Zhao, Rui; Moore, Ronald J.; Atkinson, Mark A.; Mathews, Clayton E.; Qian, Wei-Jun; Kelly, Ryan T.Chemical Science (2018), 9 (34), 6944-6951CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Multidimensional peptide sepns. can greatly increase the depth of coverage in proteome profiling. However, a major challenge for multidimensional sepns. is the requirement of large biol. samples, often contg. milligram amts. of protein. We have developed nanowell-mediated two-dimensional (2D) reversed-phase nanoflow liq. chromatog. (LC) sepns. for in-depth proteome profiling of low-nanogram samples. Peptides are first sepd. using high-pH LC and the effluent is concatenated into 4 or 12 nanowells. The contents of each nanowell are reconstituted in LC buffer and collected for subsequent sepn. and anal. by low-pH nanoLC-MS/MS. The nanowell platform minimizes peptide losses to surfaces in offline 2D LC fractionation, enabling >5800 proteins to be confidently identified from just 50 ng of HeLa digest. Furthermore, in combination with a recently developed nanowell-based sample prepn. workflow, we demonstrated deep proteome profiling of >6000 protein groups from small populations of cells, including ∼650 HeLa cells and 10 single human pancreatic islet thin sections (∼1000 cells) from a pre-symptomatic type 1 diabetic donor.
- 18Zhu, Y.; Scheibinger, M.; Ellwanger, D. C.; Krey, J. F.; Choi, D.; Kelly, R. T.; Heller, S.; Barr-Gillespie, P. G. Single-Cell Proteomics Reveals Changes in Expression during Hair-Cell Development. eLife 2019, 8, e50777, DOI: 10.7554/eLife.50777Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlamurbN&md5=66e9a7e7876b6b48af6f2fb378ccd1d1Single-cell proteomics reveals changes in expression during hair-cell developmentZhu, Ying; Scheibinger, Mirko; Ellwanger, Daniel Christian; Krey, Jocelyn F.; Choi, Dongseok; Kelly, Ryan T.; Heller, Stefan; Barr-Gillespie, Peter G.eLife (2019), 8 (), e50777CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Hearing and balance rely on small sensory hair cells that reside in the inner ear. To explore dynamic changes in the abundant proteins present in differentiating hair cells, we used nanoliter-scale shotgun mass spectrometry of single cells, each ~ 1 pL, from utricles of embryonic day 15 chickens. We identified unique constellations of proteins or protein groups from presumptive hair cells and from progenitor cells. The single-cell proteomes enabled the de novo reconstruction of a developmental trajectory using protein expression levels, revealing proteins that greatly increased in expression during differentiation of hair cells (e.g., OCM, CRABP1, GPX2, AK1, GSTO1) and those that decreased during differentiation (e.g., TMSB4X, AGR3). Complementary single-cell transcriptome profiling showed corresponding changes in mRNA during maturation of hair cells. Single-cell proteomics data thus can be mined to reveal features of cellular development that may be missed with transcriptomics.
- 19Shao, X.; Zhang, X. Design of Five-Layer Gold Nanoparticles Self-Assembled in a Liquid Open Tubular Column for Ultrasensitive Nano-LC-MS/MS Proteomic Analysis of 80 Living Cells. Proteomics 2017, 17 (8), 1600463, DOI: 10.1002/pmic.201600463Google ScholarThere is no corresponding record for this reference.
- 20Chen, Q.; Yan, G.; Gao, M.; Zhang, X. Ultrasensitive Proteome Profiling for 100 Living Cells by Direct Cell Injection, Online Digestion and Nano-LC-MS/MS Analysis. Anal. Chem. 2015, 87 (13), 6674– 6680, DOI: 10.1021/acs.analchem.5b00808Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVaku77P&md5=1b3f40134e4afd0f68bbec6483dd7455Ultrasensitive Proteome Profiling for 100 Living Cells by Direct Cell Injection, Online Digestion and Nano-LC-MS/MS AnalysisChen, Qi; Yan, Guoquan; Gao, Mingxia; Zhang, XiangminAnalytical Chemistry (Washington, DC, United States) (2015), 87 (13), 6674-6680CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Single-cell proteome anal. has always been an exciting goal because it provides crucial information about cellular heterogeneity and dynamic change. Here the authors presented an integrated proteome anal. device (iPAD) for 100 living cells (iPAD-100) that might be suitable for single-cell anal. Once cells were cultured, the iPAD-100 could be applied to inject 100 living cells, to transform the living cells into peptides, and to produce protein identification results with total automation. Due to the major obstacle for detection limit of mass spectrometry, the authors applied the iPAD-100 to analyze the proteome of 100 cells. In total, 813 proteins were identified in a DLD-cell proteome by three duplicate runs. Gene Ontol. anal. revealed that proteins from different cellular compartments were well-represented, including membrane proteins. The iPAD-100 greatly simplified the sampling process, reduced sample loss, and prevented contamination. As a result, proteins whose copy nos. were <1000 were identified from 100-cell samples with the iPAD-100, showing that a detection limit of 200 zmol was achieved. With increased sensitivity of mass spectrometry, the iPAD-100 may be able to reveal bountiful proteome information from a single cell in the near future.
- 21Zhu, Y.; Clair, G.; Chrisler, W. B.; Shen, Y.; Zhao, R.; Shukla, A. K.; Moore, R. J.; Misra, R. S.; Pryhuber, G. S.; Smith, R. D.; Ansong, C.; Kelly, R. T. Proteomic Analysis of Single Mammalian Cells Enabled by Microfluidic Nanodroplet Sample Preparation and Ultrasensitive NanoLC-MS. Angew. Chem., Int. Ed. 2018, 57 (38), 12370– 12374, DOI: 10.1002/anie.201802843Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFemur7N&md5=1c6276ba46c3866bfaeb163aee051663Proteomic Analysis of Single Mammalian Cells Enabled by Microfluidic Nanodroplet Sample Preparation and Ultrasensitive NanoLC-MSZhu, Ying; Clair, Geremy; Chrisler, William B.; Shen, Yufeng; Zhao, Rui; Shukla, Anil K.; Moore, Ronald J.; Misra, Ravi S.; Pryhuber, Gloria S.; Smith, Richard D.; Ansong, Charles; Kelly, Ryan T.Angewandte Chemie, International Edition (2018), 57 (38), 12370-12374CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The authors report on the quant. proteomic anal. of single mammalian cells. Fluorescence-activated cell sorting was employed to deposit cells into a newly developed nanodroplet sample processing chip, after which samples were analyzed by ultrasensitive nanoLC-MS. An av. of circa 670 protein groups were confidently identified from single HeLa cells, which is a far greater level of proteome coverage for single cells than has been previously reported. The single-cell proteomics platform can be used to differentiate cell types from enzyme-dissocd. human lung primary cells and identify specific protein markers for epithelial and mesenchymal cells.
- 22Shao, X.; Wang, X.; Guan, S.; Lin, H.; Yan, G.; Gao, M.; Deng, C.; Zhang, X. Integrated Proteome Analysis Device for Fast Single-Cell Protein Profiling. Anal. Chem. 2018, 90 (23), 14003– 14010, DOI: 10.1021/acs.analchem.8b03692Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitVart7%252FO&md5=f7f84b752d5638b2765d395b56eeecf6Integrated Proteome Analysis Device for Fast Single-Cell Protein ProfilingShao, Xi; Wang, Xuantang; Guan, Sheng; Lin, Haizhu; Yan, Guoquan; Gao, Mingxia; Deng, Chunhui; Zhang, XiangminAnalytical Chemistry (Washington, DC, United States) (2018), 90 (23), 14003-14010CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)In the authors' previous work, the authors demonstrated an integrated proteome anal. device (iPAD-100) to analyze proteomes from 100 cells. For the first time, a novel integrated device for single-cell anal. (iPAD-1) was developed to profile proteins in a single cell within 1 h. In the iPAD-1, a selected single cell was directly sucked into a 22 μm internal diam. capillary. Then the cell lysis and protein digestion were simultaneously accomplished in the capillary in a 2 nL vol., which could prevent protein loss and excessive diln. Digestion was accelerated by using elevated temp. with ultrasonication. The whole time of cell treatment was 30 min. After that, single-cell digest peptides were transferred into an LC column directly through a true zero dead vol. union, to minimize protein transfer loss. A homemade 22 μm internal diam. nano-LC packing column with 3 μm internal diam. ESI tip was used in the device to achieve ultrasensitive detection. A 30 min elution program was applied to anal. of the single-cell proteome. Therefore, the total time needed for a single-cell anal. was only 1 h. In an anal. of 10 single HeLa cells, a max. of 328 proteins were identified in one cell by using an Orbitrap Fusion Tribrid MS instrument, and the detection limit was estd. at ∼1.7-170 zmol. Such a sensitivity of the iPAD-1 was ∼120-fold higher than that of the authors' previously developed iPAD-100 system. Prominent cellular heterogeneity in protein expressive profiling was obsd. Furthermore, the authors roughly estd. the phases of the cell cycle of tested HeLa cells by the amt. of core histone proteins.
- 23Zhu, Y.; Piehowski, P. D.; Zhao, R.; Chen, J.; Shen, Y.; Moore, R. J.; Shukla, A. K.; Petyuk, V. A.; Campbell-Thompson, M.; Mathews, C. E.; Smith, R. D.; Qian, W.-J.; Kelly, R. T. Nanodroplet Processing Platform for Deep and Quantitative Proteome Profiling of 10–100 Mammalian Cells. Nat. Commun. 2018, 9 (1), 882, DOI: 10.1038/s41467-018-03367-wGoogle Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MrmtlyntQ%253D%253D&md5=ed52404cccbcdc4f8b9081ede1d092cbNanodroplet processing platform for deep and quantitative proteome profiling of 10-100 mammalian cellsZhu Ying; Zhao Rui; Kelly Ryan T; Piehowski Paul D; Shen Yufeng; Moore Ronald J; Shukla Anil K; Petyuk Vladislav A; Smith Richard D; Qian Wei-Jun; Chen Jing; Campbell-Thompson Martha; Mathews Clayton ENature communications (2018), 9 (1), 882 ISSN:.Nanoscale or single-cell technologies are critical for biomedical applications. However, current mass spectrometry (MS)-based proteomic approaches require samples comprising a minimum of thousands of cells to provide in-depth profiling. Here, we report the development of a nanoPOTS (nanodroplet processing in one pot for trace samples) platform for small cell population proteomics analysis. NanoPOTS enhances the efficiency and recovery of sample processing by downscaling processing volumes to <200 nL to minimize surface losses. When combined with ultrasensitive liquid chromatography-MS, nanoPOTS allows identification of ~1500 to ~3000 proteins from ~10 to ~140 cells, respectively. By incorporating the Match Between Runs algorithm of MaxQuant, >3000 proteins are consistently identified from as few as 10 cells. Furthermore, we demonstrate quantification of ~2400 proteins from single human pancreatic islet thin sections from type 1 diabetic and control donors, illustrating the application of nanoPOTS for spatially resolved proteome measurements from clinical tissues.
- 24Zhou, M.; Uwugiaren, N.; Williams, S. M.; Moore, R. J.; Zhao, R.; Goodlett, D.; Dapic, I.; Paša-Tolić, L.; Zhu, Y. Sensitive Top-Down Proteomics Analysis of a Low Number of Mammalian Cells Using a Nanodroplet Sample Processing Platform. Anal. Chem. 2020, 92 (10), 7087– 7095, DOI: 10.1021/acs.analchem.0c00467Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXosFWnsrg%253D&md5=2472ebd1f191b4f3419913f0d5999fcaSensitive Top-Down Proteomics Analysis of a Low Number of Mammalian Cells Using a Nanodroplet Sample Processing PlatformZhou, Mowei; Uwugiaren, Naomi; Williams, Sarah M.; Moore, Ronald J.; Zhao, Rui; Goodlett, David; Dapic, Irena; Pasa-Tolic, Ljiljana; Zhu, YingAnalytical Chemistry (Washington, DC, United States) (2020), 92 (10), 7087-7095CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Top-down proteomics is a powerful tool for characterizing genetic variations and post-translational modifications at intact protein level. However, one significant tech. gap of top-down proteomics is the inability to analyze a low amt. of biol. samples, which limits its access to isolated rare cells, fine needle aspiration biopsies, and tissue substructures. Herein, we developed an ultrasensitive top-down platform by incorporating a microfluidic sample prepn. system, termed nanoPOTS (nanodroplet processing in one pot for trace samples), into a top-down proteomic workflow. A unique combination of a nonionic detergent dodecyl-β-D-maltopyranoside (DDM) with urea as protein extn. buffer significantly improved both protein extn. efficiency and sample recovery. We hypothesize that the DDM detergent improves protein recovery by efficiently reducing nonspecific adsorption of intact proteins on container surfaces, while urea serves as a strong denaturant to disrupt noncovalent complexes and release intact proteins for downstream anal. The nanoPOTS-based top-down platform reproducibly and quant. identified ~ 170 to ~ 620 proteoforms from ~ 70 to ~ 770 HeLa cells contg. ~ 10 to ~ 115 ng of total protein. A variety of post-translational modifications including acetylation, myristoylation, and iron binding were identified using only less than 800 cells. We anticipate the nanoPOTS top-down proteomics platform will be broadly applicable in biomedical research, particularly where clin. specimens are not available in amts. amenable to std. workflows.
- 25Xu, K.; Liang, Y.; Piehowski, P. D.; Dou, M.; Schwarz, K. C.; Zhao, R.; Sontag, R. L.; Moore, R. J.; Zhu, Y.; Kelly, R. T. Benchtop-Compatible Sample Processing Workflow for Proteome Profiling of < 100 Mammalian Cells. Anal. Bioanal. Chem. 2019, 411 (19), 4587– 4596, DOI: 10.1007/s00216-018-1493-9Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXit12ls7nJ&md5=5f57393c6315af49f67303e080dfe016Benchtop-compatible sample processing workflow for proteome profiling of < 100 mammalian cellsXu, Kerui; Liang, Yiran; Piehowski, Paul D.; Dou, Maowei; Schwarz, Kaitlynn C.; Zhao, Rui; Sontag, Ryan L.; Moore, Ronald J.; Zhu, Ying; Kelly, Ryan T.Analytical and Bioanalytical Chemistry (2019), 411 (19), 4587-4596CODEN: ABCNBP; ISSN:1618-2642. (Springer)Extending proteomics to smaller samples can enable the mapping of protein expression across tissues with high spatial resoln. and can reveal sub-group heterogeneity. However, despite the continually improving sensitivity of LC-MS instrumentation, in-depth profiling of samples contg. low-nanogram amts. of protein has remained challenging due to analyte losses incurred during prepn. and anal. To address this, the authors recently developed nanodroplet processing in one pot for trace samples (nanoPOTS), a robotic/microfluidic platform that generates ready-to-analyze peptides from cellular material in ∼200 nL droplets with greatly reduced sample losses. In combination with ultrasensitive LC-MS, nanoPOTS has enabled >3000 proteins to be confidently identified from as few as 10 cultured human cells and ∼700 proteins from single cells. However, the nanoPOTS platform requires a highly skilled operator and a costly inhouse-built robotic nanopipetting instrument. The authors sought to evaluate the extent to which the benefits of nanodroplet processing could be preserved when upscaling reagent dispensing vols. by a factor of 10 to those addressable by com. micropipette. The authors characterized the resulting platform, termed microdroplet processing in one pot for trace samples (μPOTS), for the anal. of as few as ∼25 cultured HeLa cells (4 ng total protein) or 50 μm square mouse liver tissue thin sections and found that ∼1800 and ∼1200 unique proteins were resp. identified with high reproducibility. The reduced equipment requirements should facilitate broad dissemination of nanoproteomics workflows by obviating the need for a capital-intensive custom liq. handling system.
- 26Ross-Innes, C. S.; Becq, J.; Warren, A.; Cheetham, R. K.; Northen, H.; O’Donovan, M.; Malhotra, S.; di Pietro, M.; Ivakhno, S.; He, M.; Weaver, J. M. J.; Lynch, A. G.; Kingsbury, Z.; Ross, M.; Humphray, S.; Bentley, D.; Fitzgerald, R. C. Whole-Genome Sequencing Provides New Insights into the Clonal Architecture of Barrett’s Esophagus and Esophageal Adenocarcinoma. Nat. Genet. 2015, 47 (9), 1038– 1046, DOI: 10.1038/ng.3357Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1WltLjN&md5=a60d7a9bfd4221c3838bdfdffc50ee2aWhole-genome sequencing provides new insights into the clonal architecture of Barrett's esophagus and esophageal adenocarcinomaRoss-Innes, Caryn S.; Becq, Jennifer; Warren, Andrew; Cheetham, R. Keira; Northen, Helen; O'Donovan, Maria; Malhotra, Shalini; di Pietro, Massimiliano; Ivakhno, Sergii; He, Miao; Weaver, Jamie M. J.; Lynch, Andy G.; Kingsbury, Zoya; Ross, Mark; Humphray, Sean; Bentley, David; Fitzgerald, Rebecca C.; Hayes, Stephen J.; Ang, Yeng; Welch, Ian; Preston, Shaun; Oakes, Sarah; Save, Vicki; Skipworth, Richard; Tucker, Olga; Davies, Jim; Crichton, Charles; Schusterreiter, Christian; Underwood, Tim; Noble, Fergus; Stacey, Bernard; Kelly, Jamie; Byrne, James; Haydon, Annette; Sharland, Donna; Owsley, Jack; Barr, Hugh; Lagergren, Jesper; Gossage, James; Davies, Andrew; Mason, Robert; Chang, Fuju; Zylstra, Janine; Sanders, Grant; Wheatley, Tim; Berrisford, Richard; Bracey, Tim; Harden, Catherine; Bunting, David; Roques, Tom; Nobes, Jenny; Loo, Suat; Lewis, Mike; Cheong, Ed; Priest, Oliver; Parsons, Simon L.; Soomro, Irshad; Kaye, Philip; Saunders, John; Pang, Vincent; Welch, Neil T.; Catton, James A.; Duffy, John P.; Ragunath, Krish; Lovat, Laurence; Haidry, Rehan; Miah, Haroon; Kerr, Sarah; Eneh, Victor; Butawan, Rommel; Roques, Tom; Lewis, Michael; Cheong, Edward; Kumar, Bhasker; Igali, Laszlo; Walton, Sharon; Dann, Adela; Safranek, Peter; Hindmarsh, Andy; Sudjendran, Vijayendran; Scott, Michael; Cluroe, Alison; Miremadi, Ahmad; Mahler-Araujo, Betania; Nutzinger, Barbara; Peters, Chris; Abdullahi, Zarah; Crawte, Jason; MacRae, Shona; Noorani, Ayesha; Elliott, Rachael Fels; Bower, Lawrence; Edwards, Paul; Tavare, Simon; Eldridge, Matthew; Bornschein, Jan; Secrier, Maria; Yang, Tsun-Po; O'Neill, J. Robert; Adamczuk, Kasia; Lao-Sirieix, Pierre; Grehan, Nicola; Smith, Laura; Lishman, Suzy; Beardsmore, Duncan; Dawson, SarahNature Genetics (2015), 47 (9), 1038-1046CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)The mol. genetic relationship between esophageal adenocarcinoma (EAC) and its precursor lesion, Barrett's esophagus, is poorly understood. Using whole-genome sequencing on 23 paired Barrett's esophagus and EAC samples, together with one in-depth Barrett's esophagus case study sampled over time and space, we have provided the following new insights: (i) Barrett's esophagus is polyclonal and highly mutated even in the absence of dysplasia; (ii) when cancer develops, copy no. increases and heterogeneity persists such that the spectrum of mutations often shows surprisingly little overlap between EAC and adjacent Barrett's esophagus; and (iii) despite differences in specific coding mutations, the mutational context suggests a common causative insult underlying these two conditions. From a clin. perspective, the histopathol. assessment of dysplasia appears to be a poor reflection of the mol. disarray within the Barrett's epithelium, and a mol. Cytosponge technique overcomes sampling bias and has the capacity to reflect the entire clonal architecture.
- 27Peters, Y.; Al-Kaabi, A.; Shaheen, N. J.; Chak, A.; Blum, A.; Souza, R. F.; Di Pietro, M.; Iyer, P. G.; Pech, O.; Fitzgerald, R. C.; Siersema, P. D. Barrett Oesophagus. Nat. Rev. Dis. Primer 2019, 5 (1), 35, DOI: 10.1038/s41572-019-0086-zGoogle ScholarThere is no corresponding record for this reference.
- 28Hofmann, A. F. The Continuing Importance of Bile Acids in Liver and Intestinal Disease. Arch. Intern. Med. 1999, 159 (22), 2647– 2658, DOI: 10.1001/archinte.159.22.2647Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3c%252FmsVKhtw%253D%253D&md5=4f017c841da85f0723c292fb31e70037The continuing importance of bile acids in liver and intestinal diseaseHofmann A FArchives of internal medicine (1999), 159 (22), 2647-58 ISSN:0003-9926.Bile acids, the water-soluble, amphipathic end products of cholesterol metabolism, are involved in liver, biliary, and intestinal disease. Formed in the liver, bile acids are absorbed actively from the small intestine, with each molecule undergoing multiple enterohepatic circulations before being excreted. After their synthesis from cholesterol, bile acids are conjugated with glycine or taurine, a process that makes them impermeable to cell membranes and permits high concentrations to persist in bile and intestinal content. The relation between the chemical structure and the multiple physiological functions of bile acids is reviewed. Bile acids induce biliary lipid secretion and solubilize cholesterol in bile, promoting its elimination. In the small intestine, bile acids solubilize dietary lipids promoting their absorption. Bile acids are cytotoxic when present in abnormally high concentrations. This may occur intracellularly, as occurs in the hepatocyte in cholestasis, or extracellularly, as occurs in the colon in patients with bile acid malabsorption. Disturbances in bile acid metabolism can be caused by (1) defective biosynthesis from cholesterol or defective conjugation, (2) defective membrane transport in the hepatocyte or ileal enterocyte, (3) defective transport between organs or biliary diversion, and (4) increased bacterial degradation during enterohepatic cycling. Bile acid therapy involves bile acid replacement in deficiency states or bile acid displacement by ursodeoxycholic acid, a noncytotoxic bile acid. In cholestatic liver disease, administration of ursodeoxycholic acid decreases hepatocyte injury by retained bile acids, improving liver tests, and slowing disease progression. Bile acid malabsorption may lead to high concentrations of bile acids in the colon and impaired colonic mucosal function; bile acid sequestrants provide symptomatic benefit for diarrhea. A knowledge of bile acid physiology and the perturbations of bile acid metabolism in liver and digestive disease should be useful for the internist.
- 29Fitzgerald, R. C.; di Pietro, M.; O’Donovan, M.; Maroni, R.; Muldrew, B.; Debiram-Beecham, I.; Gehrung, M.; Offman, J.; Tripathi, M.; Smith, S. G.; Aigret, B.; Walter, F. M.; Rubin, G.; Bagewadi, A.; Patrick, A.; Shenoy, A.; Redmond, A.; Muddu, A.; Northrop, A.; Groves, A.; Shiner, A.; Heer, A.; Takhar, A.; Bowles, A.; Jarman, A.; Wong, A.; Lucas, A.; Gibbons, A.; Dhar, A.; Curry, A.; Lalonde, A.; Swinburn, A.; Turner, A.; Lydon, A.-M.; Gunstone, A.; Lee, A.; Nambi, A.; Ariyarathenam, A.; Elden, A.; Wilson, A.; Donepudi, B.; Campbell, B.; Uszycka, B.; Bowers, B.; Coghill, B.; de Quadros, B.; Cheah, C.; Bratten, C.; Brown, C.; Moorbey, C.; Clisby, C.; Gordon, C.; Schramm, C.; Castle, C.; Newark, C.; Norris, C.; A’Court, C.; Graham, C.; Fletcher, C.; Grocott, C.; Rees, C.; Bakker, C.; Paschalides, C.; Vickery, C.; Schembri, D.; Morris, D.; Hagan, D.; Cronk, D.; Goddard, D.; Graham, D.; Phillips, D.; Prabhu, D.; Kejariwal, D.; Garg, D.; Lonsdale, D.; Butterworth, D.; Clements, D.; Bradman, D.; Blake, D.; Mather, E.; O’Farrell, E.; Markowetz, F.; Adams, F.; Pesola, F.; Forbes, G.; Taylor, G.; Collins, G.; Irvine, G.; Fourie, G.; Doyle, H.; Barnes, H.; Bowyer, H.; Whiting, H.; Beales, I.; Binnian, I.; Bremner, I.; Jennings, I.; Troiceanu, I.; Modelell, I.; Emmerson, I.; Ortiz, J.; Lilley, J.; Harvey, J.; Vicars, J.; Takhar, J.; Larcombe, J.; Bornschein, J.; Aldegather, J.; Johnson, J.; Ducker, J.; Skinner, J.; Dash, J.; Walsh, J.; Miralles, J.; Ridgway, J.; Ince, J.; Kennedy, J.; Hampson, K.; Milne, K.; Ellerby, K.; Priddis, K.; Rainsbury, K.; Powell, K.; Gunner, K.; Ragunath, K.; Knox, K.; Baseley, L.; White, L.; Lovat, L.; Berney, L.; Crockett, L.; Murray, L.; Westwood, L.; Chalkley, L.; Leggett, L.; Dale, L.; Scovell, L.; Brooks, L.; Saunders, L.; Owen, L.; Dilwershah, M.; Baldry, M.; Corcoran, M.; Roy, M.; Macedo, M.; Attah, M.; Anson, M.-J.; Rutter, M.; Wallard, M.; Gaw, M.; Hunt, M.; Lea-Hagerty, M.; Penacerrada, M.; Bianchi, M.; Baker-Moffatt, M.; Czajkowski, M.; Sleeth, M.; Brewer, N.; Wooding, N.; Todd, N.; Millen, N.; Zolle, O.; Whitehead, O.; Ojechi, P.; Moore, P.; Banim, P.; Spellar, P.; Bhandari, P.; Kant, P.; Nixon, R.; Russell, R.; Roberts, R.; Skule, R.; West, R.; Fox, R.; Beesley, R.; Gibbins, R.; Osborne, R.; Thiagarajan, S.; Bastiman, S.; Warburton, S.; Pai, S.; Leith-Russell, S.; Utting, S.; Watson, S.; Wytrykowski, S.; Singh, S.; Malhotra, S.; Woods, S.; Conway, S.; Mateer, S.; Macrae, S.; Singh, S.; Fourie, S.; Campbell, S.; Parslow-Williams, S.; Goel, S.; Dellar, S.; Jones, S.; Knight, S.; Mackay-Thomas, S.; Mukherjee, S.; Allen, S.; Henry, S.; Evans, T.; Leighton, T.; Bray, T.; Shackleton, T.; Santosh, V.; Glover, V.; Chandraraj, V.; Elson, W.; Briggs, W.; Barron, Z.; Khan, Z.; Sasieni, P. Cytosponge-Trefoil Factor 3 versus Usual Care to Identify Barrett’s Oesophagus in a Primary Care Setting: A Multicentre, Pragmatic, Randomised Controlled Trial. Lancet 2020, 396 (10247), 333– 344, DOI: 10.1016/S0140-6736(20)31099-0Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38jpvVGrtQ%253D%253D&md5=b0b2f86fdf8f90356abf375b51307abeCytosponge-trefoil factor 3 versus usual care to identify Barrett's oesophagus in a primary care setting: a multicentre, pragmatic, randomised controlled trialFitzgerald Rebecca C; di Pietro Massimiliano; O'Donovan Maria; Tripathi Monika; Maroni Roberta; Muldrew Beth; Aigret Benoit; Sasieni Peter; Debiram-Beecham Irene; Gehrung Marcel; Offman Judith; Smith Samuel G; Walter Fiona M; Rubin GregLancet (London, England) (2020), 396 (10247), 333-344 ISSN:.BACKGROUND: Treatment of dysplastic Barrett's oesophagus prevents progression to adenocarcinoma; however, the optimal diagnostic strategy for Barrett's oesophagus is unclear. The Cytosponge-trefoil factor 3 (TFF3) is a non-endoscopic test for Barrett's oesophagus. The aim of this study was to investigate whether offering this test to patients on medication for gastro-oesophageal reflux would increase the detection of Barrett's oesophagus compared with standard management. METHODS: This multicentre, pragmatic, randomised controlled trial was done in 109 socio-demographically diverse general practice clinics in England. Randomisation was done both at the general practice clinic level (cluster randomisation) and at the individual patient level, and the results for each type of randomisation were analysed separately before being combined. Patients were eligible if they were aged 50 years or older, had been taking acid-suppressants for symptoms of gastro-oesophageal reflux for more than 6 months, and had not undergone an endoscopy procedure within the past 5 years. General practice clinics were selected by the local clinical research network and invited to participate in the trial. For cluster randomisation, clinics were randomly assigned (1:1) by the trial statistician using a computer-generated randomisation sequence; for individual patient-level randomisation, patients were randomly assigned (1:1) by the general practice clinics using a centrally prepared computer-generated randomisation sequence. After randomisation, participants received either standard management of gastro-oesophageal reflux (usual care group), in which participants only received an endoscopy if required by their general practitioner, or usual care plus an offer of the Cytosponge-TFF3 procedure, with a subsequent endoscopy if the procedure identified TFF3-positive cells (intervention group). The primary outcome was the diagnosis of Barrett's oesophagus at 12 months after enrolment, expressed as a rate per 1000 person-years, in all participants in the intervention group (regardless of whether they had accepted the offer of the Cytosponge-TFF3 procedure) compared with all participants in the usual care group. Analyses were intention-to-treat. The trial is registered with the ISRCTN registry, ISRCTN68382401, and is completed. FINDINGS: Between March 20, 2017, and March 21, 2019, 113 general practice clinics were enrolled, but four clinics dropped out shortly after randomisation. Using an automated search of the electronic prescribing records of the remaining 109 clinics, we identified 13 657 eligible patients who were sent an introductory letter with 14 days to opt out. 13 514 of these patients were randomly assigned (per practice or at the individual patient level) to the usual care group (n=6531) or the intervention group (n=6983). Following randomisation, 149 (2%) of 6983 participants in the intervention group and 143 (2%) of 6531 participants in the usual care group, on further scrutiny, did not meet all eligibility criteria or withdrew from the study. Of the remaining 6834 participants in the intervention group, 2679 (39%) expressed an interest in undergoing the Cytosponge-TFF3 procedure. Of these, 1750 (65%) met all of the eligibility criteria on telephone screening and underwent the procedure. Most of these participants (1654 [95%]; median age 69 years) swallowed the Cytosponge successfully and produced a sample. 231 (3%) of 6834 participants had a positive Cytosponge-TFF3 result and were referred for an endoscopy. Patients who declined the offer of the Cytosponge-TFF3 procedure and all participants in the usual care group only had an endoscopy if deemed necessary by their general practitioner. During an average of 12 months of follow-up, 140 (2%) of 6834 participants in the intervention group and 13 (<1%) of 6388 participants in the usual care group were diagnosed with Barrett's oesophagus (absolute difference 18·3 per 1000 person-years [95% CI 14·8-21·8]; rate ratio adjusted for cluster randomisation 10·6 [95% CI 6·0-18·8], p<0·0001). Nine (<1%) of 6834 participants were diagnosed with dysplastic Barrett's oesophagus (n=4) or stage I oesophago-gastric cancer (n=5) in the intervention group, whereas no participants were diagnosed with dysplastic Barrett's oesophagus or stage I gastro-oesophageal junction cancer in the usual care group. Among 1654 participants in the intervention group who swallowed the Cytosponge device successfully, 221 (13%) underwent endoscopy after testing positive for TFF3 and 131 (8%, corresponding to 59% of those having an endoscopy) were diagnosed with Barrett's oesophagus or cancer. One patient had a detachment of the Cytosponge from the thread requiring endoscopic removal, and the most common side-effect was a sore throat in 63 (4%) of 1654 participants. INTERPRETATION: In patients with gastro-oesophageal reflux, the offer of Cytosponge-TFF3 testing results in improved detection of Barrett's oesophagus. Cytosponge-TFF3 testing could also lead to the diagnosis of treatable dysplasia and early cancer. This strategy will lead to additional endoscopies with some false positive results. FUNDING: Cancer Research UK, National Institute for Health Research, the UK National Health Service, Medtronic, and the Medical Research Council.
- 30Ross-Innes, C. S.; Chettouh, H.; Achilleos, A.; Galeano-Dalmau, N.; Debiram-Beecham, I.; MacRae, S.; Fessas, P.; Walker, E.; Varghese, S.; Evan, T.; Lao-Sirieix, P. S.; O’Donovan, M.; Malhotra, S.; Novelli, M.; Disep, B.; Kaye, P. V.; Lovat, L. B.; Haidry, R.; Griffin, M.; Ragunath, K.; Bhandari, P.; Haycock, A.; Morris, D.; Attwood, S.; Dhar, A.; Rees, C.; Rutter, M. D.; Ostler, R.; Aigret, B.; Sasieni, P. D.; Fitzgerald, R. C. Risk Stratification of Barrett’s Oesophagus Using a Non-Endoscopic Sampling Method Coupled with a Biomarker Panel: A Cohort Study. Lancet Gastroenterol. Hepatol. 2017, 2 (1), 23– 31, DOI: 10.1016/S2468-1253(16)30118-2Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cvnt1OmtQ%253D%253D&md5=e51955fc3160fbf1f2d64146831713ddRisk stratification of Barrett's oesophagus using a non-endoscopic sampling method coupled with a biomarker panel: a cohort studyRoss-Innes Caryn S; Chettouh Hamza; Achilleos Achilleas; Galeano-Dalmau Nuria; Debiram-Beecham Irene; MacRae Shona; Fessas Petros; Walker Elaine; Varghese Sibu; Evan Theodore; Lao-Sirieix Pierre S; O'Donovan Maria; Malhotra Shalini; Novelli Marco; Lovat Laurence B; Haidry Rehan; Disep Babett; Griffin Michael; Kaye Phillip V; Ragunath Krish; Bhandari Pradeep; Haycock Adam; Morris Danielle; Attwood Stephen; Dhar Anjan; Rees Colin; Rutter Matt D; Ostler Richard; Aigret Benoit; Sasieni Peter D; Fitzgerald Rebecca CThe lancet. Gastroenterology & hepatology (2017), 2 (1), 23-31 ISSN:.BACKGROUND: Barrett's oesophagus predisposes to adenocarcinoma. However, most patients with Barrett's oesophagus will not progress and endoscopic surveillance is invasive, expensive, and fraught by issues of sampling bias and the subjective assessment of dysplasia. We investigated whether a non-endoscopic device, the Cytosponge, could be coupled with clinical and molecular biomarkers to identify a group of patients with low risk of progression suitable for non-endoscopic follow-up. METHODS: In this multicentre cohort study (BEST2), patients with Barrett's oesophagus underwent the Cytosponge test before their surveillance endoscopy. We collected clinical and demographic data and tested Cytosponge samples for a molecular biomarker panel including three protein biomarkers (P53, c-Myc, and Aurora kinase A), two methylation markers (MYOD1 and RUNX3), glandular atypia, and TP53 mutation status. We used a multivariable logistic regression model to compute the conditional probability of dysplasia status. We selected a simple model with high classification accuracy and applied it to an independent validation cohort. The BEST2 study is registered with ISRCTN, number 12730505. FINDINGS: The discovery cohort consisted of 468 patients with Barrett's oesophagus and intestinal metaplasia. Of these, 376 had no dysplasia and 22 had high-grade dysplasia or intramucosal adenocarcinoma. In the discovery cohort, a model with high classification accuracy consisted of glandular atypia, P53 abnormality, and Aurora kinase A positivity, and the interaction of age, waist-to-hip ratio, and length of the Barrett's oesophagus segment. 162 (35%) of 468 of patients fell into the low-risk category and the probability of being a true non-dysplastic patient was 100% (99% CI 96-100) and the probability of having high-grade dysplasia or intramucosal adenocarcinoma was 0% (0-4). 238 (51%) of participants were classified as of moderate risk; the probability of having high-grade dysplasia was 14% (9-21). 58 (12%) of participants were classified as high-risk; the probability of having non-dysplastic endoscopic biopsies was 13% (5-27), whereas the probability of having high-grade dysplasia or intramucosal adenocarcinoma was 87% (73-95). In the validation cohort (65 patients), 51 were non-dysplastic and 14 had high-grade dysplasia. In this cohort, 25 (38%) of 65 patients were classified as being low-risk, and the probability of being non-dysplastic was 96·0% (99% CI 73·80-99·99). The moderate-risk group comprised 27 non-dysplastic and eight high-grade dysplasia cases, whereas the high-risk group (8% of the cohort) had no non-dysplastic cases and five patients with high-grade dysplasia. INTERPRETATION: A combination of biomarker assays from a single Cytosponge sample can be used to determine a group of patients at low risk of progression, for whom endoscopy could be avoided. This strategy could help to avoid overdiagnosis and overtreatment in patients with Barrett's oesophagus. FUNDING: Cancer Research UK.
- 31Williams, S. M.; Liyu, A. V.; Tsai, C.-F.; Moore, R. J.; Orton, D. J.; Chrisler, W. B.; Gaffrey, M. J.; Liu, T.; Smith, R. D.; Kelly, R. T.; Pasa-Tolic, L.; Zhu, Y. Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography–Mass Spectrometry for High-Throughput Single-Cell Proteomics. Anal. Chem. 2020, 92 (15), 10588– 10596, DOI: 10.1021/acs.analchem.0c01551Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlCmtL3O&md5=11a723a01a532b3fd5dbb8f43fb00134Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography-Mass Spectrometry for High-Throughput Single-Cell ProteomicsWilliams, Sarah M.; Liyu, Andrey V.; Tsai, Chia-Feng; Moore, Ronald J.; Orton, Daniel J.; Chrisler, William B.; Gaffrey, Matthew J.; Liu, Tao; Smith, Richard D.; Kelly, Ryan T.; Pasa-Tolic, Ljiljana; Zhu, YingAnalytical Chemistry (Washington, DC, United States) (2020), 92 (15), 10588-10596CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Single-cell proteomics can provide crit. biol. insight into the cellular heterogeneity that is masked by bulk-scale anal. The authors have developed a nanoPOTS (nanodroplet processing in one pot for trace samples) platform and demonstrated its broad applicability for single-cell proteomics. However, because of nanoliter-scale sample vols., the nanoPOTS platform is not compatible with automated LC-MS systems, which significantly limits sample throughput and robustness. To address this challenge, the authors have developed a nanoPOTS autosampler allowing fully automated sample injection from nanowells to LC-MS systems. The authors also developed a sample drying, extn., and loading workflow to enable reproducible and reliable sample injection. The sequential anal. of 20 samples contg. 10 ng tryptic peptides demonstrated high reproducibility with correlation coeffs. of >0.995 between any two samples. The nanoPOTS autosampler can provide anal. throughput of 9.6, 16, and 24 single cells per day using 120, 60, and 30 min LC gradients, resp. As a demonstration for single-cell proteomics, the autosampler was first applied to profiling protein expression in single MCF10A cells using a label-free approach. At a throughput of 24 single cells per day, an av. of 256 proteins was identified from each cell and the no. was increased to 731 when the Match Between Runs algorithm of MaxQuant was used. Using a multiplexed isobaric labeling approach (TMT-11plex), ~ 77 single cells could be analyzed per day. The authors analyzed 152 cells from three acute myeloid leukemia cell lines, resulting in a total of 2558 identified proteins with 1465 proteins quantifiable (70% valid values) across the 152 cells. These data showed quant. single-cell proteomics can cluster cells to distinct groups and reveal functionally distinct differences.
- 32DEP: Differential Enrichment analysis of Proteomics data version 1.10.0 from Bioconductor, https://rdrr.io/bioc/DEP/ (accessed Jul 4, 2020).Google ScholarThere is no corresponding record for this reference.
- 33Zhang, X.; Smits, A. H.; van Tilburg, G. B.; Ovaa, H.; Huber, W.; Vermeulen, M. Proteome-Wide Identification of Ubiquitin Interactions Using UbIA-MS. Nat. Protoc. 2018, 13 (3), 530– 550, DOI: 10.1038/nprot.2017.147Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisl2ku74%253D&md5=88719f04dffb8fffe078df606dd6f5d7Proteome-wide identification of ubiquitin interactions using UbIA-MSZhang, Xiaofei; Smits, Arne H.; van Tilburg, Gabrielle B. A.; Ovaa, Huib; Huber, Wolfgang; Vermeulen, MichielNature Protocols (2018), 13 (3), 530-550CODEN: NPARDW; ISSN:1750-2799. (Nature Research)Ubiquitin-binding proteins play an important role in eukaryotes by translating differently linked polyubiquitin chains into proper cellular responses. Current knowledge about ubiquitin-binding proteins and ubiquitin linkage-selective interactions is mostly based on case-by-case studies. We have recently reported a method called ubiquitin interactor affinity enrichment-mass spectrometry (UbIA-MS), which enables comprehensive identification of ubiquitin interactors for all ubiquitin linkages from crude cell lysates. One major strength of UbIA-MS is the fact that ubiquitin interactors are enriched from crude cell lysates, in which proteins are present at endogenous levels, contain biol. relevant post-translational modifications (PTMs) and are assembled in native protein complexes. In addn., UbIA-MS uses chem. synthesized nonhydrolyzable diubiquitin, which mimics native diubiquitin and is inert to cleavage by endogenous deubiquitinases (DUBs). Here, we present a detailed protocol for UbIA-MS that proceeds in five stages: (i) chem. synthesis of ubiquitin precursors and click chem. for the generation of biotinylated nonhydrolyzable diubiquitin baits, (ii) in vitro affinity purifn. of ubiquitin interactors, (iii) on-bead interactor digestion, (iv) liq. chromatog. (LC)-MS/MS anal. and (v) data anal. to identify differentially enriched proteins. The computational anal. tools are freely available as an open-source R software package, including a graphical interface. Typically, UbIA-MS allows the identification of dozens to hundreds of ubiquitin interactors from any type of cell lysate, and can be used to study cell type or stimulus-dependent ubiquitin interactions. The nonhydrolyzable diubiquitin synthesis can be completed in 3 wk, followed by ubiquitin interactor enrichment and identification, which can be completed within another 2 wk.
- 34test_diff: Differential enrichment test in DEP: Differential Enrichment analysis of Proteomics data, https://rdrr.io/bioc/DEP/man/test_diff.html (accessed Jul 5, 2020).Google ScholarThere is no corresponding record for this reference.
- 35Ritchie, M. E.; Phipson, B.; Wu, D.; Hu, Y.; Law, C. W.; Shi, W.; Smyth, G. K. Limma Powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res. 2015, 43 (7), e47– e47, DOI: 10.1093/nar/gkv007Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFaiu7%252FN&md5=e8a5f44160c650ad89adc48a9912bd96limma powers differential expression analyses for RNA-sequencing and microarray studiesRitchie, Matthew E.; Phipson, Belinda; Wu, Di; Hu, Yifang; Law, Charity W.; Shi, Wei; Smyth, Gordon K.Nucleic Acids Research (2015), 43 (7), e47/1-e47/13CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Limma is an R/Bioconductor software package that provides an integrated soln. for analyzing data from gene expression expts. It contains rich features for handling complex exptl. designs and for information borrowing to overcome the problem of small sample sizes. Over the past decade, limma has been a popular choice for gene discovery through differential expression analyses of microarray and high-throughput PCR data. The package contains particularly strong facilities for reading, normalizing and exploring such data. Recently, the capabilities of limma have been significantly expanded in two important directions. First, the package can now perform both differential expression and differential splicing analyses of RNA sequencing (RNA-seq) data. All the downstream anal. tools previously restricted to microarray data are now available for RNA-seq as well. These capabilities allow users to analyze both RNA-seq and microarray data with very similar pipelines. Second, the package is now able to go past the traditional gene-wise expression analyses in a variety of ways, analyzing expression profiles in terms of co-regulated sets of genes or in terms of higher-order expression signatures. This provides enhanced possibilities for biol. interpretation of gene expression differences. This article reviews the philosophy and design of the limma package, summarizing both new and historical features, with an emphasis on recent enhancements and features that have not been previously described.
- 36P’ng, C.; Green, J.; Chong, L. C.; Waggott, D.; Prokopec, S. D.; Shamsi, M.; Nguyen, F.; Mak, D. Y. F.; Lam, F.; Albuquerque, M. A.; Wu, Y.; Jung, E. H.; Starmans, M. H. W.; Chan-Seng-Yue, M. A.; Yao, C. Q.; Liang, B.; Lalonde, E.; Haider, S.; Simone, N. A.; Sendorek, D.; Chu, K. C.; Moon, N. C.; Fox, N. S.; Grzadkowski, M. R.; Harding, N. J.; Fung, C.; Murdoch, A. R.; Houlahan, K. E.; Wang, J.; Garcia, D. R.; de Borja, R.; Sun, R. X.; Lin, X.; Chen, G. M.; Lu, A.; Shiah, Y.-J.; Zia, A.; Kearns, R.; Boutros, P. C. BPG: Seamless, Automated and Interactive Visualization of Scientific Data. BMC Bioinf. 2019, 20 (1), 42, DOI: 10.1186/s12859-019-2610-2Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cjjs1altA%253D%253D&md5=61ec34f7005b4dfc1819b7e76300d80fBPG: Seamless, automated and interactive visualization of scientific dataP'ng Christine; Green Jeffrey; Chong Lauren C; Waggott Daryl; Prokopec Stephenie D; Shamsi Mehrdad; Nguyen Francis; Mak Denise Y F; Lam Felix; Albuquerque Marco A; Wu Ying; Jung Esther H; Starmans Maud H W; Chan-Seng-Yue Michelle A; Yao Cindy Q; Liang Bianca; Lalonde Emilie; Haider Syed; Simone Nicole A; Sendorek Dorota; Chu Kenneth C; Moon Nathalie C; Fox Natalie S; Grzadkowski Michal R; Harding Nicholas J; Fung Clement; Murdoch Amanda R; Houlahan Kathleen E; Wang Jianxin; Garcia David R; de Borja Richard; Sun Ren X; Lin Xihui; Chen Gregory M; Lu Aileen; Shiah Yu-Jia; Zia Amin; Kearns Ryan; Boutros Paul C; Yao Cindy Q; Lalonde Emilie; Fox Natalie S; Houlahan Kathleen E; Shiah Yu-Jia; Boutros Paul C; Wang Jianxin; Sun Ren X; Lu Aileen; Boutros Paul C; Boutros Paul C; Boutros Paul C; Boutros Paul C; Boutros Paul CBMC bioinformatics (2019), 20 (1), 42 ISSN:.BACKGROUND: We introduce BPG, a framework for generating publication-quality, highly-customizable plots in the R statistical environment. RESULTS: This open-source package includes multiple methods of displaying high-dimensional datasets and facilitates generation of complex multi-panel figures, making it suitable for complex datasets. A web-based interactive tool allows online figure customization, from which R code can be downloaded for integration with computational pipelines. CONCLUSION: BPG provides a new approach for linking interactive and scripted data visualization and is available at http://labs.oicr.on.ca/boutros-lab/software/bpg or via CRAN at https://cran.r-project.org/web/packages/BoutrosLab.plotting.general.
- 37Hulsen, T.; de Vlieg, J.; Alkema, W. BioVenn – a Web Application for the Comparison and Visualization of Biological Lists Using Area-Proportional Venn Diagrams. BMC Genomics 2008, 9 (1), 488, DOI: 10.1186/1471-2164-9-488Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1cjksF2ltQ%253D%253D&md5=1c79e2fa6accc5ff3c4cd77bab9aca8cBioVenn - a web application for the comparison and visualization of biological lists using area-proportional Venn diagramsHulsen Tim; de Vlieg Jacob; Alkema WynandBMC genomics (2008), 9 (), 488 ISSN:.BACKGROUND: In many genomics projects, numerous lists containing biological identifiers are produced. Often it is useful to see the overlap between different lists, enabling researchers to quickly observe similarities and differences between the data sets they are analyzing. One of the most popular methods to visualize the overlap and differences between data sets is the Venn diagram: a diagram consisting of two or more circles in which each circle corresponds to a data set, and the overlap between the circles corresponds to the overlap between the data sets. Venn diagrams are especially useful when they are 'area-proportional' i.e. the sizes of the circles and the overlaps correspond to the sizes of the data sets. Currently there are no programs available that can create area-proportional Venn diagrams connected to a wide range of biological databases. RESULTS: We designed a web application named BioVenn to summarize the overlap between two or three lists of identifiers, using area-proportional Venn diagrams. The user only needs to input these lists of identifiers in the textboxes and push the submit button. Parameters like colors and text size can be adjusted easily through the web interface. The position of the text can be adjusted by 'drag-and-drop' principle. The output Venn diagram can be shown as an SVG or PNG image embedded in the web application, or as a standalone SVG or PNG image. The latter option is useful for batch queries. Besides the Venn diagram, BioVenn outputs lists of identifiers for each of the resulting subsets. If an identifier is recognized as belonging to one of the supported biological databases, the output is linked to that database. Finally, BioVenn can map Affymetrix and EntrezGene identifiers to Ensembl genes. CONCLUSION: BioVenn is an easy-to-use web application to generate area-proportional Venn diagrams from lists of biological identifiers. It supports a wide range of identifiers from the most used biological databases currently available. Its implementation on the World Wide Web makes it available for use on any computer with internet connection, independent of operating system and without the need to install programs locally. BioVenn is freely accessible at http://www.cmbi.ru.nl/cdd/biovenn/.
- 38Perez-Riverol, Y.; Csordas, A.; Bai, J.; Bernal-Llinares, M.; Hewapathirana, S.; Kundu, D. J.; Inuganti, A.; Griss, J.; Mayer, G.; Eisenacher, M.; Pérez, E.; Uszkoreit, J.; Pfeuffer, J.; Sachsenberg, T.; Yılmaz, Ş.; Tiwary, S.; Cox, J.; Audain, E.; Walzer, M.; Jarnuczak, A. F.; Ternent, T.; Brazma, A.; Vizcaíno, J. A. The PRIDE Database and Related Tools and Resources in 2019: Improving Support for Quantification Data. Nucleic Acids Res. 2019, 47 (D1), D442– D450, DOI: 10.1093/nar/gky1106Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1GqtrzK&md5=4cd0f929c2df840a5cc0631d0d708d6cThe PRIDE database and related tools and resources in 2019: improving support for quantification dataPerez-Riverol, Yasset; Csordas, Attila; Bai, Jingwen; Bernal-Llinare, Manuel; Hewapathirana, Suresh; Kundu, Deepti J.; Iuganti, Avinash; Griss, Johannes; Mayer, Gerhard; Eisenacher, Martin; Perez, Enrique; Uszkoreit, Julian; Pfeuffer, Julianus; Sachsenberg, Timo; Yilmaz, Sule; Tiwary, Shivani; Cox, Jurgen; Audain, Enrique; Walzer, Mathias; Jarnuczak, Andrew F.; Ternent, Tobias; Brazma, Alvis; Vizcaino, Juan AntonioNucleic Acids Research (2019), 47 (D1), D442-D450CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. The PRoteomics IDEntifications (PRIDE) database is the world's largest data repository of mass spectrometry-based proteomics data, and is one of the founding members of the global ProteomeXchange (PX) consortium. In this manuscript, we summarize the developments in PRIDE resources and related tools since the previous update manuscript was published in Nucleic Acids Research in 2016. In the last 3 years, public data sharing through PRIDE (as part of PX) has definitely become the norm in the field. In parallel, data re-use of public proteomics data has increased enormously, with multiple applications. We first describe the new architecture of PRIDE Archive, the archival component of PRIDE. PRIDE Archive and the related data submission framework have been further developed to support the increase in submitted data vols. and addnl. data types. A new scalable and fault tolerant storage backed, Application Programming Interface and web interface have been implemented, as a part of an ongoing process. Addnl., we emphasize the improved support for quant. proteomics data through the mzTab format. At last, we outline key statistics on the current data contents and vol. of downloads, and how PRIDE data are starting to be disseminated to added-value resources including Ensembl, UniProt and Expression Atlas.
- 39UniProt, https://www.uniprot.org/ (accessed Jun 25, 2020).Google ScholarThere is no corresponding record for this reference.
- 40Protein GRAVY, https://www.bioinformatics.org/sms2/protein_gravy.html (accessed Jun 25, 2020).Google ScholarThere is no corresponding record for this reference.
- 41Budnik, B.; Levy, E.; Harmange, G.; Slavov, N. SCoPE-MS: Mass Spectrometry of Single Mammalian Cells Quantifies Proteome Heterogeneity during Cell Differentiation. Genome Biol. 2018, 19 (1), 161, DOI: 10.1186/s13059-018-1547-5Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFKls7nM&md5=bfdeda4d526356689afaa4c6d97a20d5SCoPE-MS: mass spectrometry of single mammalian cells quantifies proteome heterogeneity during cell differentiationBudnik, Bogdan; Levy, Ezra; Harmange, Guillaume; Slavov, NikolaiGenome Biology (2018), 19 (), 161/1-161/12CODEN: GNBLFW; ISSN:1474-760X. (BioMed Central Ltd.)Some exciting biol. questions require quantifying thousands of proteins in single cells. To achieve this goal, we develop Single Cell ProtEomics by Mass Spectrometry (SCoPE-MS) and validate its ability to identify distinct human cancer cell types based on their proteomes. We use SCoPE-MS to quantify over a thousand proteins in differentiating mouse embryonic stem cells. The single-cell proteomes enable us to deconstruct cell populations and infer protein abundance relationships. Comparison between single-cell proteomes and transcriptomes indicates coordinated mRNA and protein covariation, yet many genes exhibit functionally concerted and distinct regulatory patterns at the mRNA and the protein level.
- 42Dou, M.; Clair, G.; Tsai, C.-F.; Xu, K.; Chrisler, W. B.; Sontag, R. L.; Zhao, R.; Moore, R. J.; Liu, T.; Pasa-Tolic, L.; Smith, R. D.; Shi, T.; Adkins, J. N.; Qian, W.-J.; Kelly, R. T.; Ansong, C.; Zhu, Y. High-Throughput Single Cell Proteomics Enabled by Multiplex Isobaric Labelling in a Nanodroplet Sample Preparation Platform. Anal. Chem. 2019, 91 (20), 13119– 13127, DOI: 10.1021/acs.analchem.9b03349Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslKisbbJ&md5=800d1bf514934f3fe80ae23fa44729d7High-Throughput Single Cell Proteomics Enabled by Multiplex Isobaric Labeling in a Nanodroplet Sample Preparation PlatformDou, Maowei; Clair, Geremy; Tsai, Chia-Feng; Xu, Kerui; Chrisler, William B.; Sontag, Ryan L.; Zhao, Rui; Moore, Ronald J.; Liu, Tao; Pasa-Tolic, Ljiljana; Smith, Richard D.; Shi, Tujin; Adkins, Joshua N.; Qian, Wei-Jun; Kelly, Ryan T.; Ansong, Charles; Zhu, YingAnalytical Chemistry (Washington, DC, United States) (2019), 91 (20), 13119-13127CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Effective extension of mass spectrometry-based proteomics to single cells remains challenging. Herein the authors combined microfluidic nanodroplet technol. with tandem mass tag (TMT) isobaric labeling to significantly improve anal. throughput and proteome coverage for single mammalian cells. Isobaric labeling facilitated multiplex anal. of single cell-sized protein quantities to a depth of ∼1600 proteins with median CV of 10.9% and correlation coeff. of 0.98. To demonstrate in-depth high throughput single cell anal., the platform was applied to measure protein expression in 72 single cells from three murine cell populations (epithelial, immune, and endothelial cells) in <2 days instrument time with over 2300 proteins identified. Principal component anal. grouped the single cells into three distinct populations based on protein expression with each population characterized by well-known cell-type specific markers. The platform enables high throughput and unbiased characterization of single cell heterogeneity at the proteome level.
- 43Weng, S. S. H.; Demir, F.; Ergin, E. K.; Dirnberger, S.; Uzozie, A.; Tuscher, D.; Nierves, L.; Tsui, J.; Huesgen, P. F.; Lange, P. F. Sensitive Determination of Proteolytic Proteoforms in Limited Microscale Proteome Samples. Mol. Cell. Proteomics 2019, 18 (11), 2335– 2347, DOI: 10.1074/mcp.TIR119.001560Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisVSjsrjN&md5=5f4d95ae406fe34978aa8860add4fa9cSensitive determination of proteolytic proteoforms in limited microscale proteome samplesWeng, Samuel S. H.; Demir, Fatih; Ergin, Enes K.; Dirnberger, Sabrina; Uzozie, Anuli; Tuscher, Domenic; Nierves, Lorenz; Tsui, Janice; Huesgen, Pitter F.; Lange, Philipp F.Molecular & Cellular Proteomics (2019), 18 (11), 2335-2347CODEN: MCPOBS; ISSN:1535-9484. (American Society for Biochemistry and Molecular Biology)Protein N termini unambiguously identify truncated, alternatively translated or modified proteoforms with distinct functions and reveal perturbations in disease. Selective enrichment of N-terminal peptides is necessary to achieve proteome-wide coverage for unbiased identification of site-specific regulatory proteolytic processing and protease substrates. However, many proteolytic processes are strictly confined in time and space and therefore can only be analyzed in minute samples that provide insufficient starting material for current enrichment protocols. Here we present High-efficiency Undecanal-based N Termini EnRichment (HUNTER), a robust, sensitive and scalable method for the anal. of previously inaccessible microscale samples. HUNTER achieved identification of >1000 N termini from as little as 2 mug raw HeLa cell lysate. Broad applicability is demonstrated by the first N-terminome anal. of sorted human primary immune cells and enriched mitochondrial fractions from pediatric cancer patients, as well as protease substrate identification from individual Arabidopsis thaliana wild type and Vacuolar Processing Enzyme-deficient mutant seedlings. We further implemented the workflow on a liq. handling system and demonstrate the feasibility of clin. degradomics by automated processing of liq. biopsies from pediatric cancer patients.
- 44Kasuga, K.; Katoh, Y.; Nagase, K.; Igarashi, K. Microproteomics with Microfluidic-Based Cell Sorting: Application to 1000 and 100 Immune Cells. Proteomics 2017, 17 (13–14), 1600420, DOI: 10.1002/pmic.201600420Google ScholarThere is no corresponding record for this reference.
- 45Sun, J.; Zhang, G. L.; Li, S.; Ivanov, A. R.; Fenyo, D.; Lisacek, F.; Murthy, S. K.; Karger, B. L.; Brusic, V. Pathway Analysis and Transcriptomics Improve Protein Identification by Shotgun Proteomics from Samples Comprising Small Number of Cells - a Benchmarking Study. BMC Genomics 2014, 15 (Suppl 9), S1, DOI: 10.1186/1471-2164-15-S9-S1Google ScholarThere is no corresponding record for this reference.
- 46Tanca, A.; Abbondio, M.; Pisanu, S.; Pagnozzi, D.; Uzzau, S.; Addis, M. F. Critical Comparison of Sample Preparation Strategies for Shotgun Proteomic Analysis of Formalin-Fixed, Paraffin-Embedded Samples: Insights from Liver Tissue. Clin. Proteomics 2014, 11 (1), 28, DOI: 10.1186/1559-0275-11-28Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhslShsLjJ&md5=407da191800181ff40883e9c36fef0c3Critical comparison of sample preparation strategies for shotgun proteomic analysis of formalin-fixed, paraffin-embedded samples: insights from liver tissueTanca, Alessandro; Abbondio, Marcello; Pisanu, Salvatore; Pagnozzi, Daniela; Uzzau, Sergio; Addis, Maria FilippaClinical Proteomics (2014), 11 (), 28/1-28/12, 12 pp.CODEN: CPLRCX; ISSN:1542-6416. (BioMed Central Ltd.)Background: The growing field of formalin-fixed paraffin-embedded (FFPE) tissue proteomics holds promise for improving translational research. Direct tissue trypsinization (DT) and protein extn. followed by in soln. digestion (ISD) or filter-aided sample prepn. (FASP) are the most common workflows for shotgun anal. of FFPE samples, but a crit. comparison of the different methods is currently lacking. Exptl. Design: DT, FASP and ISD workflows were compared by subjecting to the same label-free quant. approach three independent tech. replicates of each method applied to FFPE liver tissue. Data were evaluated in terms of method reproducibility and protein/peptide distribution according to localization, MW, pI and hydrophobicity. Results: DT showed lower reproducibility, good preservation of high-MW proteins, a general bias towards hydrophilic and acidic proteins, much lower keratin contamination, as well as higher abundance of non-tryptic peptides. Conversely, FASP and ISD proteomes were depleted in high-MW proteins and enriched in hydrophobic and membrane proteins; FASP provided higher identification yields, while ISD exhibited higher reproducibility. Conclusions: These results highlight that diverse sample prepn. strategies provide significantly different proteomic information, and present typical biases that should be taken into account when dealing with FFPE samples. When a sufficient amt. of tissue is available, the complementary use of different methods is suggested to increase proteome coverage and depth.
- 47Föll, M. C.; Fahrner, M.; Oria, V. O.; Kühs, M.; Biniossek, M. L.; Werner, M.; Bronsert, P.; Schilling, O. Reproducible Proteomics Sample Preparation for Single FFPE Tissue Slices Using Acid-Labile Surfactant and Direct Trypsinization. Clin. Proteomics 2018, 15 (1), 11, DOI: 10.1186/s12014-018-9188-yGoogle Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MrpvFarsg%253D%253D&md5=48eb2283b6a607be9332be0d1ddf983bReproducible proteomics sample preparation for single FFPE tissue slices using acid-labile surfactant and direct trypsinizationFoll Melanie Christine; Fahrner Matthias; Oria Victor Oginga; Biniossek Martin Lothar; Schilling Oliver; Foll Melanie Christine; Fahrner Matthias; Oria Victor Oginga; Fahrner Matthias; Oria Victor Oginga; Kuhs Markus; Werner Martin; Bronsert Peter; Kuhs Markus; Werner Martin; Bronsert Peter; Kuhs Markus; Werner Martin; Bronsert Peter; Werner Martin; Bronsert Peter; Schilling Oliver; Schilling OliverClinical proteomics (2018), 15 (), 11 ISSN:1542-6416.BACKGROUND: Proteomic analyses of clinical specimens often rely on human tissues preserved through formalin-fixation and paraffin embedding (FFPE). Minimal sample consumption is the key to preserve the integrity of pathological archives but also to deal with minimal invasive core biopsies. This has been achieved by using the acid-labile surfactant RapiGest in combination with a direct trypsinization (DTR) strategy. A critical comparison of the DTR protocol with the most commonly used filter aided sample preparation (FASP) protocol is lacking. Furthermore, it is unknown how common histological stainings influence the outcome of the DTR protocol. METHODS: Four single consecutive murine kidney tissue specimens were prepared with the DTR approach or with the FASP protocol using both 10 and 30 k filter devices and analyzed by label-free, quantitative liquid chromatography-tandem mass spectrometry (LC-MS/MS). We compared the different protocols in terms of proteome coverage, relative label-free quantitation, missed cleavages, physicochemical properties and gene ontology term annotations of the proteins. Additionally, we probed compatibility of the DTR protocol for the analysis of common used histological stainings, namely hematoxylin & eosin (H&E), hematoxylin and hemalaun. These were proteomically compared to an unstained control by analyzing four human tonsil FFPE tissue specimens per condition. RESULTS: On average, the DTR protocol identified 1841 ± 22 proteins in a single, non-fractionated LC-MS/MS analysis, whereas these numbers were 1857 ± 120 and 1970 ± 28 proteins for the FASP 10 and 30 k protocol. The DTR protocol showed 15% more missed cleavages, which did not adversely affect quantitation and intersample comparability. Hematoxylin or hemalaun staining did not adversely impact the performance of the DTR protocol. A minor perturbation was observed for H&E staining, decreasing overall protein identification by 13%. CONCLUSIONS: In essence, the DTR protocol can keep up with the FASP protocol in terms of qualitative and quantitative reproducibility and performed almost as well in terms of proteome coverage and missed cleavages. We highlight the suitability of the DTR protocol as a viable and straightforward alternative to the FASP protocol for proteomics-based clinical research.
- 48Broeckx, V.; Boonen, K.; Pringels, L.; Sagaert, X.; Prenen, H.; Landuyt, B.; Schoofs, L.; Maes, E. Comparison of Multiple Protein Extraction Buffers for GeLC-MS/MS Proteomic Analysis of Liver and Colon Formalin-Fixed, Paraffin-Embedded Tissues. Mol. BioSyst. 2016, 12 (2), 553– 565, DOI: 10.1039/C5MB00670HGoogle Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvF2hsrvK&md5=1e245f198339d7405f0c8709bbc4e44aComparison of multiple protein extraction buffers for GeLC-MS/MS proteomic analysis of liver and colon formalin-fixed, paraffin-embedded tissuesBroeckx, Valerie; Boonen, Kurt; Pringels, Lentel; Sagaert, Xavier; Prenen, Hans; Landuyt, Bart; Schoofs, Liliane; Maes, EvelyneMolecular BioSystems (2016), 12 (2), 553-565CODEN: MBOIBW; ISSN:1742-2051. (Royal Society of Chemistry)Formalin-fixed paraffin-embedded (FFPE) tissue specimens represent a potential valuable source of samples for clin. research. Since these specimens are banked in hospital archives, large cohorts of samples can be collected in short periods of time which can all be linked with a patients' clin. history. Therefore, the use of FFPE tissue in protein biomarker discovery studies gains interest. However, despite the growing no. of FFPE proteome studies in the literature, there is a lack of a FFPE proteomics std. operating procedure (SOP). One of the challenging steps in the development of such a SOP is the ability to obtain an efficient and repeatable extn. of full length FFPE proteins. In this study, the protein extn. efficiency of eight protein extn. buffers is critically compared with GeLC-MS/MS (1D gel electrophoresis followed by in-gel digestion and LC-MS/MS). The data variation caused by using these extn. buffers was investigated since the variation is a very important aspect when using FFPE tissue as a source for biomarker detection. In addn., a qual. comparison was made between the protein extn. efficiency and repeatability for FFPE tissue and fresh frozen tissue.
- 49Tabb, D. L.; Vega-Montoto, L.; Rudnick, P. A.; Variyath, A. M.; Ham, A.-J. L.; Bunk, D. M.; Kilpatrick, L. E.; Billheimer, D. D.; Blackman, R. K.; Cardasis, H. L.; Carr, S. A.; Clauser, K. R.; Jaffe, J. D.; Kowalski, K. A.; Neubert, T. A.; Regnier, F. E.; Schilling, B.; Tegeler, T. J.; Wang, M.; Wang, P.; Whiteaker, J. R.; Zimmerman, L. J.; Fisher, S. J.; Gibson, B. W.; Kinsinger, C. R.; Mesri, M.; Rodriguez, H.; Stein, S. E.; Tempst, P.; Paulovich, A. G.; Liebler, D. C.; Spiegelman, C. Repeatability and Reproducibility in Proteomic Identifications by Liquid Chromatography–Tandem Mass Spectrometry. J. Proteome Res. 2010, 9 (2), 761– 776, DOI: 10.1021/pr9006365Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFGnt7bP&md5=77c1052918728af58b3b5c4a5bc250a9Repeatability and Reproducibility in Proteomic Identifications by Liquid Chromatography-Tandem Mass SpectrometryTabb, David L.; Vega-Montoto, Lorenzo; Rudnick, Paul A.; Variyath, Asokan Mulayath; Ham, Amy-Joan L.; Bunk, David M.; Kilpatrick, Lisa E.; Billheimer, Dean D.; Blackman, Ronald K.; Cardasis, Helene L.; Carr, Steven A.; Clauser, Karl R.; Jaffe, Jacob D.; Kowalski, Kevin A.; Neubert, Thomas A.; Regnier, Fred E.; Schilling, Birgit; Tegeler, Tony J.; Wang, Mu; Wang, Pei; Whiteaker, Jeffrey R.; Zimmerman, Lisa J.; Fisher, Susan J.; Gibson, Bradford W.; Kinsinger, Christopher R.; Mesri, Mehdi; Rodriguez, Henry; Stein, Stephen E.; Tempst, Paul; Paulovich, Amanda G.; Liebler, Daniel C.; Spiegelman, CliffJournal of Proteome Research (2010), 9 (2), 761-776CODEN: JPROBS; ISSN:1535-3893. (American Chemical Society)The complexity of proteomic instrumentation for LC-MS/MS introduces many possible sources of variability. Data-dependent sampling of peptides constitutes a stochastic element at the heart of discovery proteomics. Although this variation impacts the identification of peptides, proteomic identifications are far from completely random. In this study, the authors analyzed interlab. data sets from the NCI Clin. Proteomic Technol. Assessment for Cancer to examine repeatability and reproducibility in peptide and protein identifications. Included data spanned 144 LC-MS/MS expts. on four Thermo LTQ and four Orbitrap instruments. Samples included yeast lysate, the NCI-20 defined dynamic range protein mix, and the Sigma UPS 1 defined equimolar protein mix. Some of the authors' findings reinforced conventional wisdom, such as repeatability and reproducibility being higher for proteins than for peptides. Most lessons from the data, however, were more subtle. Orbitrap proved capable of higher repeatability and reproducibility, but aberrant performance occasionally erased these gains. Even the simplest protein digestions yielded more peptide ions than LC-MS/MS could identify during a single expt. The authors obsd. that peptide lists from pairs of tech. replicates overlapped by 35-60%, giving a range for peptide-level repeatability in these expts. Sample complexity did not appear to affect peptide identification repeatability, even as nos. of identified spectra changed by an order of magnitude. Statistical anal. of protein spectral counts revealed greater stability across tech. replicates for Orbitrap, making them superior to LTQ instruments for biomarker candidate discovery. The most repeatable peptides were those corresponding to conventional tryptic cleavage sites, those that produced intense MS signals, and those that resulted from proteins generating many distinct peptides. Reproducibility among different instruments of the same type lagged behind repeatability of tech. replicates on a single instrument by several percent. These findings reinforce the importance of evaluating repeatability as a fundamental characteristic of anal. technologies.
- 50Delmotte, N.; Lasaosa, M.; Tholey, A.; Heinzle, E.; van Dorsselaer, A.; Huber, C. G. Repeatability of Peptide Identifications in Shotgun Proteome Analysis Employing Off-Line Two-Dimensional Chromatographic Separations and Ion-Trap MS. J. Sep. Sci. 2009, 32 (8), 1156– 1164, DOI: 10.1002/jssc.200800615Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlvVCltr8%253D&md5=81ccb5ce2f00a4a8fc0eec13bf94fd05Repeatability of peptide identifications in shotgun proteome analysis employing off-line two-dimensional chromatographic separations and ion-trap MSDelmotte, Nathanael; Lasaosa, Maria; Tholey, Andreas; Heinzle, Elmar; van Dorsselaer, Alain; Huber, Christian G.Journal of Separation Science (2009), 32 (8), 1156-1164CODEN: JSSCCJ; ISSN:1615-9306. (Wiley-VCH Verlag GmbH & Co. KGaA)The repeatability of peptide identifications in shotgun proteome analyses employing strong cation-exchange- × ion-pair RP HPLC hyphenated to ESI MS/MS was compared to an alternative scheme, comprising high-pH RP chromatog. combined with low-pH ion-pair RP chromatog. Equivalent results were obtained with both methods in proteome anal. of Corynebacterium glutamicum. From a total no. of 1350 to 1850 peptides identified in triplicate analyses of five consecutive fractions chosen from the first-dimension sepn., 41-45% of the peptides were identified three times, whereas 16-22 and 37-39% of the peptides were identified only twice or once, resp. A comparison of the repeatability of peptide identifications from complex samples upon 1- or 2-D chromatog. sepn. revealed that an addnl. sepn. dimension decreases the repeatability by approx. 25%.
- 51Resing, K. A.; Meyer-Arendt, K.; Mendoza, A. M.; Aveline-Wolf, L. D.; Jonscher, K. R.; Pierce, K. G.; Old, W. M.; Cheung, H. T.; Russell, S.; Wattawa, J. L.; Goehle, G. R.; Knight, R. D.; Ahn, N. G. Improving Reproducibility and Sensitivity in Identifying Human Proteins by Shotgun Proteomics. Anal. Chem. 2004, 76 (13), 3556– 3568, DOI: 10.1021/ac035229mGoogle Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXktlWmsbs%253D&md5=056551e9a48b0bdb1d0f5668bb9625a9Improving Reproducibility and Sensitivity in Identifying Human Proteins by Shotgun ProteomicsResing, Katheryn A.; Meyer-Arendt, Karen; Mendoza, Alex M.; Aveline-Wolf, Lauren D.; Jonscher, Karen R.; Pierce, Kevin G.; Old, William M.; Cheung, Hiu T.; Russell, Steven; Wattawa, Joy L.; Goehle, Geoff R.; Knight, Robin D.; Ahn, Natalie G.Analytical Chemistry (2004), 76 (13), 3556-3568CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Identifying proteins in cell exts. by shotgun proteomics involves digesting the proteins, sequencing the resulting peptides by data-dependent mass spectrometry (MS/MS), and searching protein databases to identify the proteins from which the peptides are derived. Manual anal. and direct spectral comparison reveal that scores from two commonly used search programs (Sequest and Mascot) validate less than half of potentially identifiable MS/MS spectra (class pos.) from shotgun analyses of the human erythroleukemia K562 cell line. Here we demonstrate increased sensitivity and accuracy using a focused search strategy along with a peptide sequence validation script that does not rely exclusively on XCorr or Mowse scores generated by Sequest or Mascot, but uses consensus between the search programs, along with chem. properties and scores describing the nature of the fragmentation spectrum (ion score and RSP). The approach yielded 4.2% false pos. and 8% false neg. frequencies in peptide assignments. The protein profile is then assembled from peptide assignments using a novel peptide-centric protein nomenclature that more accurately reports protein variants that contain identical peptide sequences. An Isoform Resolver algorithm ensures that the protein count is not inflated by variants in the protein database, eliminating ∼25% of redundant proteins. Anal. of sol. proteins from a human K562 cells identified 5130 unique proteins, with ∼100 false pos. protein assignments.
- 52Washburn, M. P.; Ulaszek, R. R.; Yates, J. R. Reproducibility of Quantitative Proteomic Analyses of Complex Biological Mixtures by Multidimensional Protein Identification Technology. Anal. Chem. 2003, 75 (19), 5054– 5061, DOI: 10.1021/ac034120bGoogle Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXmsVegsLs%253D&md5=93f083bea4d279e1cf92ea0f6248a8c3Reproducibility of quantitative proteomic analyses of complex biological mixtures by multidimensional protein identification technologyWashburn, Michael P.; Ulaszek, Ryan R.; Yates, John R., IIIAnalytical Chemistry (2003), 75 (19), 5054-5061CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)If quant. proteomic technologies are to be of widespread use to the biol. community, the reproducibility of each method must be investigated and detd. We have analyzed the reproducibility of complex quant. proteomic analyses of metabolically labeled S. cerevisiae analyzed via multidimensional protein identification technol. (MudPIT). Three independent cell growths of S. cerevisiae grown in rich and minimal media and independent MudPIT analyses of each were compared and contrasted. Quant. MudPIT was found to be intra- and interexperimentally reproducible at both the peptide and protein levels. Proteins of potential low abundance were detected, identified, and quantified by identical peptides from three independent samples. In addn., when multiple peptides were matched to a protein, the relative abundance of each peptide was in agreement across the three samples. Despite the reproducibility, errors in the exptl. detn. of protein expression levels occurred, but the impact of the variation was minimized by replicate expts. Last, quant. MudPIT analyses will likely be improved by increasing the no. of peptide hits per protein in a given anal., which will provide for greater intraexperimental reproducibility.
- 53Magdeldin, S.; Yamamoto, T. Toward Deciphering Proteomes of Formalin-Fixed Paraffin-Embedded (FFPE) Tissues. Proteomics 2012, 12 (7), 1045– 1058, DOI: 10.1002/pmic.201100550Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XlvVKms78%253D&md5=7cd5165b42a902e467226a114103f3c1Toward deciphering proteomes of formalin-fixed paraffin-embedded (FFPE) tissuesMagdeldin, Sameh; Yamamoto, TadashiProteomics (2012), 12 (7), 1045-1058CODEN: PROTC7; ISSN:1615-9853. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Formalin-fixed paraffin-embedded (FFPE) tissue specimens comprise a potentially valuable resource for both prospective and retrospective biomarker discovery. Unlocking the proteomic profile of clinicopathol. FFPE tissues is a critically essential step for annotating clin. findings and predicting biomarkers for ultimate disease prognosis and therapeutic follow-up.
- 54Maes, E.; Broeckx, V.; Mertens, I.; Sagaert, X.; Prenen, H.; Landuyt, B.; Schoofs, L. Analysis of the Formalin-Fixed Paraffin-Embedded Tissue Proteome: Pitfalls, Challenges, and Future Prospectives. Amino Acids 2013, 45 (2), 205– 218, DOI: 10.1007/s00726-013-1494-0Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFemtbvN&md5=cfb25a4dc12c7a6a9b76472d8c4da015Analysis of the formalin-fixed paraffin-embedded tissue proteome: pitfalls, challenges, and future prospectivesMaes, Evelyne; Broeckx, Valerie; Mertens, Inge; Sagaert, Xavier; Prenen, Hans; Landuyt, Bart; Schoofs, LilianeAmino Acids (2013), 45 (2), 205-218CODEN: AACIE6; ISSN:0939-4451. (Springer-Verlag GmbH)A review. Formalin-fixed paraffin-embedded (FFPE) tissues are a real treasure for retrospective anal. considering the amt. of samples present in hospital archives, combined with pathol., clin., and outcome information available for every sample. Although unlocking the proteome of these tissues is still a challenge, new approaches are being developed. In this review, we summarize the different mass spectrometry platforms that are used in human clin. studies to unravel the FFPE proteome. The different ways of extg. crosslinked proteins and the anal. strategies are pointed out. Also, the pitfalls and challenges concerning the quality of FFPE proteomic approaches are depicted. We also evaluated the potential of these anal. methods for future clin. FFPE proteomics applications.
- 55Proungvitaya, S.; Klinthong, W.; Proungvitaya, T.; Limpaiboon, T.; Jearanaikoon, P.; Roytrakul, S.; Wongkham, C.; Nimboriboonporn, A.; Wongkham, S. High Expression of CCDC25 in Cholangiocarcinoma Tissue Samples. Oncol. Lett. 2017, 14 (2), 2566– 2572, DOI: 10.3892/ol.2017.6446Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cflslWhsg%253D%253D&md5=dbbf13668447ea01c723e4bf965dca50High expression of CCDC25 in cholangiocarcinoma tissue samplesProungvitaya Siriporn; Klinthong Wachiraya; Proungvitaya Tanakorn; Limpaiboon Temduang; Jearanaikoon Patcharee; Proungvitaya Siriporn; Proungvitaya Tanakorn; Limpaiboon Temduang; Jearanaikoon Patcharee; Wongkham Chaisiri; Nimboriboonporn Anongporn; Wongkham Sopit; Roytrakul Sittiruk; Nimboriboonporn AnongpornOncology letters (2017), 14 (2), 2566-2572 ISSN:1792-1074.Cholangiocarcinoma (CCA) is a malignant transformation of biliary epithelial cells. It is a slow growing tumor, but is also highly metastatic with a poor prognosis. Bile acids are known to transactivate the epidermal growth factor receptor (EGFR) in cholangiocytes and induce cyclooxygenase-2 expression. The protein expression profiles of bile acid-treated CCA cells were studied using a proteomic approach. To elucidate the possible mechanisms involved in the bile acid-mediated enhancement of CCA cell migration, the effects of six bile acids, including cholic, deoxycholic, taurocholic, taurodeoxycholic, glycocholic and glycodeoxycholic acid, on the migration of CCA cells were examined in vitro using wound healing assays. Subsequently, the possible proteins involved in enhanced CCA cell migration were investigated using a proteomic approach. Changes to the protein expression profiles of CCA cells following bile acid treatment was examined using two-dimensional electrophoresis and mass spectrometry. The results demonstrated that cholic and deoxycholic acid significantly enhanced the migration of CCA cells, compared with the treated MMNK-1 control cells. CCA cells had 77 overexpressed protein spots following cholic acid treatment, and 50 protein spots following deoxycholic acid treatment, compared with the treated MMNK-1 control cells. Liquid chromatography tandem-mass spectrometry analysis revealed that coiled-coil domain containing 25 (CCDC25) was significantly overexpressed in cholic acid-treated CCA cells compared with in cholic acid-treated control cells. When the expression levels of CCDC25 were investigated using western blot analysis, CCDC25 was demonstrated to be highly expressed in CCA tissues, but not in the adjacent non-cancerous tissue samples. The identified proteins were further analyzed for protein-chemical interactions using STITCH version 3.1 software. CCDC25 protein was identified to be associated with Son of sevenless homolog 1 and growth factor receptor-bound protein 2, which are involved in EGFR signaling. The results of the present study demonstrated that following cholic acid treatment, CCDC25 is overexpressed in CCA cells, which is associated with significantly enhanced cell migration. This suggests that CCDC25 is a potential therapeutic target for the treatment of patients with CCA.
- 56Brychtova, V.; Vojtesek, B.; Hrstka, R. Anterior Gradient 2: A Novel Player in Tumor Cell Biology. Cancer Lett. 2011, 304 (1), 1– 7, DOI: 10.1016/j.canlet.2010.12.023Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjsFWru7g%253D&md5=6baccba91e15dfacd80ed874ae409574Anterior gradient 2: A novel player in tumor cell biologyBrychtova, Veronika; Vojtesek, Borivoj; Hrstka, RomanCancer Letters (New York, NY, United States) (2011), 304 (1), 1-7CODEN: CALEDQ; ISSN:0304-3835. (Elsevier)A review. AGR2 has evolutionarily conserved roles in development and tissue regeneration and is linked with several human cancers. The exact functions and regulation of AGR2 are poorly understood, but current data identify AGR2 as a clin. relevant factor that modulates the behavior and response of hormone-dependent cancers (breast, prostate) and hormone-independent cancers (colorectal, pancreatic, esophageal and other common cancers). AGR2 protein expression induces metastasis, acts as a p53 tumor suppressor inhibitor and survival factor, participates directly in neoplastic transformation and is involved in drug resistance. Thus, AGR2 is an important tumor biomarker and neg. prognostic factor potentially exploitable in clin. practice.
- 57Suwanmanee, G.; Yosudjai, J.; Phimsen, S.; Wongkham, S.; Jirawatnotai, S.; Kaewkong, W. Upregulation of AGR2vH Facilitates Cholangiocarcinoma Cell Survival under Endoplasmic Reticulum Stress via the Activation of the Unfolded Protein Response Pathway. Int. J. Mol. Med. 2019, 45 (2), 669– 677, DOI: 10.3892/ijmm.2019.4432Google ScholarThere is no corresponding record for this reference.
Cited By
This article is cited by 11 publications.
- Dashuang Jia, Peter Nemes. Development and Validation of RoboCap, a Robotic Capillary Platform to Automate Capillary Electrophoresis Mass Spectrometry En Route to High-Throughput Single-Cell Proteomics. Analytical Chemistry 2024, 96
(42)
, 16985-16993. https://doi.org/10.1021/acs.analchem.4c04353
- Marija Veličković, Thomas L. Fillmore, Isaac Kwame Attah, Camilo Posso, James C. Pino, Rui Zhao, Sarah M. Williams, Dušan Veličković, Jon M. Jacobs, Kristin E. Burnum-Johnson, Ying Zhu, Paul D. Piehowski. Coupling Microdroplet-Based Sample Preparation, Multiplexed Isobaric Labeling, and Nanoflow Peptide Fractionation for Deep Proteome Profiling of the Tissue Microenvironment. Analytical Chemistry 2024, 96
(32)
, 12973-12982. https://doi.org/10.1021/acs.analchem.4c00523
- Leena R. Pade, Camille Lombard-Banek, Jie Li, Peter Nemes. Dilute to Enrich for Deeper Proteomics: A Yolk-Depleted Carrier for Limited Populations of Embryonic (Frog) Cells. Journal of Proteome Research 2024, 23
(2)
, 692-703. https://doi.org/10.1021/acs.jproteome.3c00541
- Narae Park, Hankyul Lee, Yumi Kwon, Shinyeong Ju, Seonjeong Lee, Seongjin Yoo, Kang-Sik Park, Cheolju Lee. One-STAGE Tip Method for TMT-Based Proteomic Analysis of a Minimal Amount of Cells. ACS Omega 2023, 8
(22)
, 19741-19751. https://doi.org/10.1021/acsomega.3c01392
- Azad Eshghi, Xiaofeng Xie, Darryl Hardie, Michael X. Chen, Fabiana Izaguirre, Rachael Newman, Ying Zhu, Ryan T. Kelly, David R. Goodlett. Sample Preparation Methods for Targeted Single-Cell Proteomics. Journal of Proteome Research 2023, 22
(6)
, 1589-1602. https://doi.org/10.1021/acs.jproteome.2c00429
- J. Robert O'Neill, Marcos Yébenes Mayordomo, Goran Mitulović, Sofian Al Shboul, Georges Bedran, Jakub Faktor, Lenka Hernychova, Lukas Uhrik, Maria Gómez-Herranz, Mikołaj Kocikowski, Vicki Save, Bořivoj Vojtěšek, Mark J. Arends, Rebecca C. Fitzgerald, Paul A.W. Edwards, Nicola Grehan, Barbara Nutzinger, Christine Loreno, Aisling M. Redmond, Sujath Abbas, Adam Freeman, Elizabeth C. Smyth, Maria O'Donovan, Ahmad Miremadi, Shalini Malhotra, Monika Tripathi, Calvin Cheah, Hannah Coles, Curtis Millington, Ginny Devonshire, Matthew Eldridge, Maria Secrier, Sriganesh Jammula, Jim Davies, Charles Crichton, Nick Carroll, Richard H. Hardwick, Peter Safranek, Andrew Hindmarsh, Vijayendran Sujendran, Stephen J. Hayes, Yeng Ang, Andrew Sharrocks, Shaun R. Preston, Izhar Bagwan, Vicki Save, Richard J.E. Skipworth, Ted R. Hupp, J Robert O'Neill, Olga Tucker, Andrew Beggs, Philippe Taniere, Sonia Puig, Gianmarco Contino, Timothy J. Underwood, Robert C. Walker, Ben L. Grace, Jesper Lagergren, James Gossage, Andrew Davies, Fuju Chang, Ula Mahadeva, Vicky Goh, Francesca D. Ciccarelli, Grant Sanders, Richard Berrisford, David Chan, Ed Cheong, Bhaskar Kumar, L. Sreedharan, Simon L. Parsons, Irshad Soomro, Philip Kaye, John Saunders, Laurence Lovat, Rehan Haidry, Michael Scott, Sharmila Sothi, Suzy Lishman, George B. Hanna, Christopher J. Peters, Krishna Moorthy, Anna Grabowska, Richard Turkington, Damian McManus, Helen Coleman, Russell D. Petty, Freddie Bartlet, Ted Hupp, Javier Antonio Alfaro. Multi-Omic Analysis of Esophageal Adenocarcinoma Uncovers Candidate Therapeutic Targets and Cancer-Selective Posttranscriptional Regulation. Molecular & Cellular Proteomics 2024, 23
(6)
, 100764. https://doi.org/10.1016/j.mcpro.2024.100764
- Thy Truong, Ximena Sanchez-Avila, Kei G. I. Webber, S. Madisyn Johnston, Ryan T. Kelly. Efficient and Sensitive Sample Preparation, Separations, and Data Acquisition for Label-Free Single-Cell Proteomics. 2024, 67-84. https://doi.org/10.1007/978-1-0716-3934-4_7
- Rushdy Ahmad, Bogdan Budnik. A review of the current state of single-cell proteomics and future perspective. Analytical and Bioanalytical Chemistry 2023, 415
(28)
, 6889-6899. https://doi.org/10.1007/s00216-023-04759-8
- Sara J.C. Gosline, Marija Veličković, James C. Pino, Le Z. Day, Isaac K. Attah, Adam C. Swensen, Vincent Danna, Camilo Posso, Karin D. Rodland, Jing Chen, Clayton E. Matthews, Martha Campbell-Thompson, Julia Laskin, Kristin Burnum-Johnson, Ying Zhu, Paul D. Piehowski. Proteome Mapping of the Human Pancreatic Islet Microenvironment Reveals Endocrine–Exocrine Signaling Sphere of Influence. Molecular & Cellular Proteomics 2023, 22
(8)
, 100592. https://doi.org/10.1016/j.mcpro.2023.100592
- Marija Veličković, Thomas L. Fillmore, Kwame Attah, Camilo Posso, James C. Pino, Rui Zhao, Sarah M. Williams, Dušan Veličković, Jon M. Jacobs, Kristin E. Burnum-Johnson, Ying Zhu, Paul D. Piehowski. Coupling microdroplet-based sample preparation, multiplexed isobaric labeling, and nanoflow peptide fractionation for deep proteome profiling of tissue microenvironment. 2023https://doi.org/10.1101/2023.03.13.531822
- Hongxian Zhao, Yongle Chen, Hang Li, Yangjun Zhang, Wanjun Zhang, Weijie Qin. An angled-shape tip-based strategy for highly sensitive proteomic profiling of a low number of cells. Analytical Methods 2023, 15
(9)
, 1215-1222. https://doi.org/10.1039/D2AY01884E
- J Robert O’Neill, Marcos Yébenes Mayordomo, Goran Mitulović, Sofian Al Shboul, Georges Bedran, Jakub Faktor, Lenka Hernychova, Lukas Uhrik, Maria Gomez-Herranz, Mikołaj Kocikowski, Vicki Save, Bořivoj Vojtěšek, Mark Arends, , Ted Hupp, Javier Alfaro. Multi-omics integration in Esophageal Adenocarcinoma reveals therapeutic targets and EAC-specific regulation of protein abundances. 2022https://doi.org/10.1101/2022.11.24.22281691
- Sara JC Gosline, Marija Velickovic, James Pino, Le Z. Day, Isaac K. Attah, Adam C. Swensen, Vincent Danna, Karin D. Rodland, Jing Chen, Clayton E. Matthews, Martha Campbell-Thompson, Julia Laskin, Kristin Burnum-Johnson, Ying Zhu, Paul D. Piehowski. Proteome mapping of the human pancreatic islet microenvironment reveals endocrine-exocrine signaling sphere of influence. 2022https://doi.org/10.1101/2022.11.21.517388
- James M Fulcher. Laser Capture Microdissection of Tissue Functional Units for microPOTS Top-Down Proteomics v1. 2022https://doi.org/10.17504/protocols.io.ceu5tey6
- James M Fulcher. Laser Capture Microdissection of Tissue Functional Units for microPOTS Top-Down Proteomics v1. 2022https://doi.org/10.17504/protocols.io.ewov1nrz7gr2/v1
- Kenneth Weke, Sachin Kote, Jakub Faktor, Sofian Al Shboul, Naomi Uwugiaren, Paul M. Brennan, David R. Goodlett, Ted R. Hupp, Irena Dapic. DIA-MS proteome analysis of formalin-fixed paraffin-embedded glioblastoma tissues. Analytica Chimica Acta 2022, 1204 , 339695. https://doi.org/10.1016/j.aca.2022.339695
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 57 other publications.
- 1Mann, M.; Hendrickson, R. C.; Pandey, A. Analysis of Proteins and Proteomes by Mass Spectrometry. Annu. Rev. Biochem. 2001, 70, 437– 473, DOI: 10.1146/annurev.biochem.70.1.4371https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXlsVehtL4%253D&md5=1bd749a47bfc1ec30226dce9a820aef6Analysis of proteins and proteomes by mass spectrometryMann, Matthias; Hendrickson, Ronald C.; Pandey, AkhileshAnnual Review of Biochemistry (2001), 70 (), 437-473CODEN: ARBOAW; ISSN:0066-4154. (Annual Reviews Inc.)A review with 91 refs. A decade after the discovery of electrospray and matrix-assisted laser desorption ionization (MALDI), methods that finally allowed gentle ionization of large biomols., mass spectrometry has become a powerful tool in protein anal. and the key technol. in the emerging field of proteomics. The success of mass spectrometry is driven both by innovative instrumentation designs, esp. those operating on the time-of-flight or ion-trapping principles, and by large-scale biochem. strategies, which use mass spectrometry to detect the isolated proteins. Any human protein can now be identified directly from genome databases on the basis of minimal data derived by mass spectrometry. As has already happened in genomics, increased automation of sample handling, anal., and the interpretation of results will generate an avalanche of qual. and quant. proteomic data. Protein-protein interactions can be analyzed directly by pptn. of a tagged bait followed by mass spectrometric identification of its binding partners. By these and similar strategies, entire protein complexes, signaling pathways, and whole organelles are being characterized. Posttranslational modifications remain difficult to analyze but are starting to yield to generic strategies.
- 2Wasinger, V. C.; Cordwell, S. J.; Cerpa-Poljak, A.; Yan, J. X.; Gooley, A. A.; Wilkins, M. R.; Duncan, M. W.; Harris, R.; Williams, K. L.; Humphery-Smith, I. Progress with Gene-Product Mapping of the Mollicutes: Mycoplasma Genitalium. Electrophoresis 1995, 16 (7), 1090– 1094, DOI: 10.1002/elps.115016011852https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXntF2lsLg%253D&md5=11e7acd04dc0eaf17ec7f99996a0c654Progress with gene-product mapping of the Mollicutes: Mycoplasma genitaliumWasinger, Valerie C.; Cordwell, Stuart J.; Cerpa-Poljak, Anne; Yan, Jun X.; Gooley, Andrew A.; Wilkins, Marc R.; Duncan, Mark W.; Harris, Ray; Williams, Keith L.; Humphery-Smith, IanElectrophoresis (1995), 16 (7), 1090-4CODEN: ELCTDN; ISSN:0173-0835. (VCH)A protein map of the smallest known self-replicating organism Mycoplasma genitalium (Class: Mollicutes) revealed a high proportion of acidic proteins. Amino acid compn. was used to putatively identify or provide unique parameters for 50 gene products sepd. by two-dimensional gel electrophoresis. A further 19 proteins were subjected to peptide-mass fingerprinting using matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry and 4 were subjected to N-terminal Edman degrdn. The majority of M. genitalium proteins remain uncharacterized. However, the combined approach of amino acid anal. and peptide-mass fingerprinting allowed gene products to be linked to homologous genes in a variety of organisms. This allowed proteins to be identified prior to detection of their resp. genes via the M. genitalium sequencing initiative. The principle of 'hierarchical' anal. for the mass screening of proteins and the anal. of microbial genomes via their protein complement or 'proteome' is detailed. Here, characterization of gene products depends upon the quickest and most economical technols. being employed initially to det. if a large no. of proteins are already present in both homologous and heterologous species databases. Initial screening, which lends itself to automation and robotics, can then be followed by more time and cost intensive procedures, when necessary.
- 3Zhang, Y.; Fonslow, B. R.; Shan, B.; Baek, M.-C.; Yates, J. R. Protein Analysis by Shotgun/Bottom-up Proteomics. Chem. Rev. 2013, 113 (4), 2343– 2394, DOI: 10.1021/cr30035333https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXjtVClu7g%253D&md5=a27c72f72bc6fa2f827725d2e71df241Protein Analysis by Shotgun/Bottom-up ProteomicsZhang, Yaoyang; Fonslow, Bryan R.; Shan, Bing; Baek, Moon-Chang; Yates, John R.Chemical Reviews (Washington, DC, United States) (2013), 113 (4), 2343-2394CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review.
- 4Mayne, J.; Ning, Z.; Zhang, X.; Starr, A. E.; Chen, R.; Deeke, S.; Chiang, C.-K.; Xu, B.; Wen, M.; Cheng, K.; Seebun, D.; Star, A.; Moore, J. I.; Figeys, D. Bottom-Up Proteomics (2013–2015): Keeping up in the Era of Systems Biology. Anal. Chem. 2016, 88 (1), 95– 121, DOI: 10.1021/acs.analchem.5b042304https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvVSlsLvJ&md5=291844fae9e91fc1b1d219948ef7d3c4Bottom-Up Proteomics (2013-2015): Keeping up in the Era of Systems BiologyMayne, Janice; Ning, Zhibin; Zhang, Xu; Starr, Amanda E.; Chen, Rui; Deeke, Shelley; Chiang, Cheng-Kang; Xu, Bo; Wen, Ming; Cheng, Kai; Seebun, Deeptee; Star, Alexandra; Moore, Jasmine I.; Figeys, DanielAnalytical Chemistry (Washington, DC, United States) (2016), 88 (1), 95-121CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A review. Over 17,000 papers related to proteomics have been published since the authors' 2011-2013 review. A subset are true gems reporting amazing applications of proteomics to better understand protein interactions, pathways, dynamic signaling and the human proteome, to name a few. While the authors have clearly seen more attention paid to data quality, and the proper exptl. design in manuscripts reporting biol. applications of proteomics, more improvements are needed. Technol. development still remains a strong driving force of proteomics, making up a large portion of these papers. As in any maturing field, some areas of technol. development in proteomics are satg. However, many new areas in proteomics are burgeoning. Examples include the development of enrichment materials to study different protein modifications, approaches for protein quantification, data independent acquisition (DIA) methods, bioinformatics and the rapid developments in metaproteomics. The acceptance of proteomics in the biol. community really depends on its applications. For instance, protein interaction mapping, post-translational modification (PTM) analyses and protein Atlas are routinely used by the biol. community. However, other aspects of proteomics are still under appreciated including biomarker discovery and studies of diseases from tissues. In part, this is due to previous and, to some extent, still ongoing overstatements from underpowered studies that reported unsubstantiated biol. conclusions. Very large-scale, good quality, quant. proteomic studies are now tech. feasible and ongoing in a few labs. around the world. However, few bioinformatics tools are capable of handling these large studies. Moreover, proteomics is increasingly integrated in expts. involving multiple -omics datasets obtained across large nos. of biol. replicates. "Big data" challenges are clearly appearing in proteomics. This biannual review is focused on technol. trends, which the authors believe are important in proteomics. The authors continue to foresee that technol. and bioinformatics development will remain important areas of research in proteomics.
- 5Fagerberg, L.; Strömberg, S.; El-Obeid, A.; Gry, M.; Nilsson, K.; Uhlen, M.; Ponten, F.; Asplund, A. Large-Scale Protein Profiling in Human Cell Lines Using Antibody-Based Proteomics. J. Proteome Res. 2011, 10 (9), 4066– 4075, DOI: 10.1021/pr200259v5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXptFKlsr4%253D&md5=38f0615cccc16390ae4f6be6fd806c85Large-Scale Protein Profiling in Human Cell Lines Using Antibody-Based ProteomicsFagerberg, Linn; Stromberg, Sara; El-Obeid, Adila; Gry, Marcus; Nilsson, Kenneth; Uhlen, Mathias; Ponten, Fredrik; Asplund, AnnaJournal of Proteome Research (2011), 10 (9), 4066-4075CODEN: JPROBS; ISSN:1535-3893. (American Chemical Society)Human cancer cell lines grown in vitro are frequently used to decipher basic cell biol. phenomena and to also specifically study different forms of cancer. Here the authors present the first large-scale study of protein expression patterns in cell lines using an antibody-based proteomics approach. The authors analyzed the expression pattern of 5436 proteins in 45 different cell lines using hierarchical clustering, principal component anal., and two-group comparisons for the identification of differentially expressed proteins. The authors' results show that immunohistochem. detd. protein profiles can categorize cell lines into groups that overall reflect the tumor tissue of origin and that hematol. cell lines appear to retain their protein profiles to a higher degree than cell lines established from solid tumors. The two-group comparisons reveal well-characterized proteins as well as previously unstudied proteins that could be of potential interest for further investigations. Moreover, multiple myeloma cells and cells of myeloid origin were found to share a protein profile, relative to the protein profile of lymphoid leukemia and lymphoma cells, possibly reflecting their common dependency of bone marrow microenvironment. This work also provides an extensive list of antibodies, for which high-resoln. images as well as validation data are available on the Human Protein Atlas (www.proteinatlas.org), that are of potential use in cell line studies.
- 6Branca, R. M. M.; Orre, L. M.; Johansson, H. J.; Granholm, V.; Huss, M.; Pérez-Bercoff, Å.; Forshed, J.; Käll, L.; Lehtiö, J. HiRIEF LC-MS Enables Deep Proteome Coverage and Unbiased Proteogenomics. Nat. Methods 2014, 11 (1), 59– 62, DOI: 10.1038/nmeth.27326https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslygsLnE&md5=acdad94163f52313a336a41ccfed44c4HiRIEF LC-MS enables deep proteome coverage and unbiased proteogenomicsBranca, Rui M. M.; Orre, Lukas M.; Johansson, Henrik J.; Granholm, Viktor; Huss, Mikael; Perez-Bercoff, Aasa; Forshed, Jenny; Kaell, Lukas; Lehtioe, JanneNature Methods (2014), 11 (1), 59-62CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)We present a liq. chromatog.-mass spectrometry (LC-MS)-based method permitting unbiased (gene prediction-independent) genome-wide discovery of protein-coding loci in higher eukaryotes. Using high-resoln. isoelec. focusing (HiRIEF) at the peptide level in the 3.7-5.0 pH range and accurate peptide isoelec. point (pI) prediction, we probed the six-reading-frame translation of the human and mouse genomes and identified 98 and 52 previously undiscovered protein-coding loci, resp. The method also enabled deep proteome coverage, identifying 13,078 human and 10,637 mouse proteins.
- 7Wang, G.; Brennan, C.; Rook, M.; Wolfe, J. L.; Leo, C.; Chin, L.; Pan, H.; Liu, W.-H.; Price, B.; Makrigiorgos, G. M. Balanced-PCR Amplification Allows Unbiased Identification of Genomic Copy Changes in Minute Cell and Tissue Samples. Nucleic Acids Res. 2004, 32 (9), e76, DOI: 10.1093/nar/gnh0707https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXksVahsLk%253D&md5=e87e9779bb0d6186644d4f7b394d1448Balanced-PCR amplification allows unbiased identification of genomic copy changes in minute cell and tissue samplesWang, Gang; Brennan, Cameron; Rook, Martha; Wolfe, Jia Liu; Leo, Christopher; Chin, Lynda; Pan, Hongjie; Liu, Wei-Hua; Price, Brendan; Makrigiorgos, G. MikeNucleic Acids Research (2004), 32 (9), e76/1-e76/10CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Anal. of genomic DNA derived from cells and fresh or fixed tissues often requires whole genome amplification prior to microarray screening. Tech. hurdles to this process are the introduction of amplification bias and/or the inhibitory effects of formalin fixation on DNA amplification. Here we demonstrate a balanced-PCR procedure that allows unbiased amplification of genomic DNA from fresh or modestly degraded paraffin-embedded DNA samples. Following digestion and ligation of a target and a control genome with distinct linkers, the two are mixed and amplified in a single PCR, thereby avoiding biases assocd. with PCR satn. and impurities. We demonstrate genome-wide retention of allelic differences following balanced-PCR amplification of DNA from breast cancer and normal human cells and genomic profiling by array-CGH (cDNA arrays, 100 kb resoln.) and by real-time PCR (single gene resoln.). Comparison of balanced-PCR with multiple displacement amplification (MDA) demonstrates equiv. performance between the two when intact genomic DNA is used. When DNA from paraffin-embedded samples is used, balanced PCR overcomes problems assocd. with modest DNA degrdn. and produces unbiased amplification whereas MDA does not. Balanced-PCR allows amplification and recovery of modestly degraded genomic DNA for subsequent retrospective anal. of human tumors with known outcomes.
- 8Wang, N.; Xu, M.; Wang, P.; Li, L. Development of Mass Spectrometry-Based Shotgun Method for Proteome Analysis of 500 to 5000 Cancer Cells. Anal. Chem. 2010, 82 (6), 2262– 2271, DOI: 10.1021/ac90230228https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXptlGrtA%253D%253D&md5=89ac83bbe62a4e164961925b870a13e4Development of Mass Spectrometry-Based Shotgun Method for Proteome Analysis of 500 to 5000 Cancer CellsWang, Nan; Xu, Mingguo; Wang, Peng; Li, LiangAnalytical Chemistry (Washington, DC, United States) (2010), 82 (6), 2262-2271CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)A shotgun proteome anal. method and its performance for protein identification from 500 to 5000 cells are described. Sample prepn., which was done in one tube, involved the use of a surfactant (NP-40) for cell lysis, followed by acetone pptn. of the proteins. The resulting protein pellet was washed with cold acetone to remove remaining surfactant, and the pellet was then solubilized in NH4HCO3. After trypsin digestion of the proteins, the digest was analyzed by the use of nanoflow liq. chromatog. (LC) quadrupole time-of-flight mass spectrometry (QTOF MS). Sample injection and gradient speed in running LC QTOF MS were optimized. It was shown that this method could identify an av. (n = 3) of 167±21, 237±30, 491±63, and 619±59 proteins from 500, 1000, 2500, and 5000 MCF-7 breast cancer cells, resp. To demonstrate the potential use of this method for generating proteome profile from circulating tumor cells (CTCs) isolated from human blood, a healthy human blood sample was spiked with MCF-7 cells, and this mixt. was processed and then subjected to antibody tagging of the MCF-7 cells. The tagged cells were sorted and collected using flow cytometry. The proteome profiles of small nos. of cells isolated in this way were similar to those of the original MCF-7 cells, suggesting the possibility of the use of this method for cell typing of CTCs.
- 9Parapatics, K.; Huber, M. L.; Lehmann, D.; Knoll, C.; Superti-Furga, G.; Bennett, K. L.; Rudashevskaya, E. L. Proteomic Analysis of Low Quantities of Cellular Material in the Range Obtainable from Scarce Patient Samples. J. Integr. OMICS 2015, 5 (1), 30– 43–43, DOI: 10.5584/jiomics.v5i1.172There is no corresponding record for this reference.
- 10Clair, G.; Piehowski, P. D.; Nicola, T.; Kitzmiller, J. A.; Huang, E. L.; Zink, E. M.; Sontag, R. L.; Orton, D. J.; Moore, R. J.; Carson, J. P.; Smith, R. D.; Whitsett, J. A.; Corley, R. A.; Ambalavanan, N.; Ansong, C. Spatially-Resolved Proteomics: Rapid Quantitative Analysis of Laser Capture Microdissected Alveolar Tissue Samples. Sci. Rep. 2016, 6, 6, DOI: 10.1038/srep39223There is no corresponding record for this reference.
- 11Coscia, F.; Doll, S.; Bech, J. M.; Mund, A.; Lengyel, E.; Lindebjerg, J.; Madsen, G. I.; Moreira, J. M. A.; Mann, M. A Streamlined Mass Spectrometry-Based Proteomics Workflow for Large Scale FFPE Tissue Analysis. bioRxiv , Sep 23, 2019, 779009. DOI: 10.1101/779009 .There is no corresponding record for this reference.
- 12Martin, J. G.; Rejtar, T.; Martin, S. A. Integrated Microscale Analysis System for Targeted Liquid Chromatography Mass Spectrometry Proteomics on Limited Amounts of Enriched Cell Populations. Anal. Chem. 2013, 85 (22), 10680– 10685, DOI: 10.1021/ac401937c12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsFGkurbN&md5=7fd20d18e145d970a56b1aadfe651f89Integrated Microscale Analysis System for Targeted Liquid Chromatography Mass Spectrometry Proteomics on Limited Amounts of Enriched Cell PopulationsMartin, Jeffrey G.; Rejtar, Tomas; Martin, Stephen A.Analytical Chemistry (Washington, DC, United States) (2013), 85 (22), 10680-10685CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Limited samples, such as those that are in vivo sourced via biopsy, are closely representative of biol. systems and contain valuable information for drug discovery. However, these precious samples are often heterogeneous and require cellular prefractionation prior to proteomic anal. to isolate specific subpopulations of interest. Enriched cells from in vivo samples are often very limited (<104 cells) and pose a significant challenge to proteomic nanoliquid chromatog. mass spectrometry (nanoLCMS) sample prepn. To enable the streamlined anal. of these limited samples, the authors have developed an online cell enrichment, microscale sample prepn., nanoLCMS proteomics workflow by integrating fluorescence activated cell sorting (FACS), focused ultrasonication, microfluidics, immobilized trypsin digestion, and nanoLCMS. To assess the performance of the online FACS-Chip-LCMS workflow, 5000 fluorescent labeled cells were enriched from a 5% heterogeneous cell population and processed for LCMS proteomics in <2 h. Within these 5000 enriched cells, 30 peptides corresponding to 17 proteins spanning >4 orders of magnitude of cellular abundance were quantified using a QExactive MS. The results from the online FACS-Chip-LCMS workflow starting from 5000 enriched cells were directly compared to results from a traditional macroscale sample prepn. workflow starting from 2.0 × 106 cells. The microscale FACS-Chip-LCMS workflow demonstrated high cellular enrichment efficiency and high peptide recovery across the wide dynamic range of targeted peptides. Overall the microscale FACS-Chip-LCMS workflow showed effectiveness in efficiently prepg. limited amts. of FACS enriched cells in an online manner for proteomic LCMS.
- 13Yamaguchi, H.; Miyazaki, M.; Honda, T.; Briones-Nagata, M. P.; Arima, K.; Maeda, H. Rapid and Efficient Proteolysis for Proteomic Analysis by Protease-Immobilized Microreactor. Electrophoresis 2009, 30 (18), 3257– 3264, DOI: 10.1002/elps.20090013413https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFGqsLjK&md5=9870da1a12d77e6e302a53e76019fd79Rapid and efficient proteolysis for proteomic analysis by protease-immobilized microreactorYamaguchi, Hiroshi; Miyazaki, Masaya; Honda, Takeshi; Briones-Nagata, Maria Portia; Arima, Kazunari; Maeda, HideakiElectrophoresis (2009), 30 (18), 3257-3264CODEN: ELCTDN; ISSN:0173-0835. (Wiley-VCH Verlag GmbH & Co. KGaA)Proteolysis is an important part of protein identification in proteomics anal. The conventional method of in-soln. digestion of proteins is time-consuming and has limited sensitivity. In this study, trypsin- or α-chymotrypsin-immobilized microreactors prepd. by a microfluidics-based enzyme-immobilization technique were studied for rapid sample prepn. in proteomic anal. The kinetic studies for hydrolysis of substrate by microreactors revealed that immobilized proteases had higher hydrolytic efficiency than those performed by in-soln. digestion. The performance of the microreactors was evaluated by digesting cytochrome c and bovine serum albumin (BSA). Protein digestion was achieved within a short period of time (∼5 min) at 30° without any complicated redn. and alkylation procedures. The efficiency of digestion by trypsin-immobilized reactor was evaluated by analyzing the sequence coverage, which was 47 and 12% for cytochrome c and BSA, resp. These values were higher than those performed by in-soln. digestion. Besides, because of higher stability against high concn. of denaturant, the microreactors can be useful for immediate digestion of the denatured protein. In the present study, the authors propose a protease-immobilized microreactor digestion method, which can utilize as a proteome technique for biol. and clin. research.
- 14Myers, S. A.; Rhoads, A.; Cocco, A. R.; Peckner, R.; Haber, A. L.; Schweitzer, L. D.; Krug, K.; Mani, D. R.; Clauser, K. R.; Rozenblatt-Rosen, O.; Hacohen, N.; Regev, A.; Carr, S. A. Streamlined Protocol for Deep Proteomic Profiling of FAC-Sorted Cells and Its Application to Freshly Isolated Murine Immune Cells. Mol. Cell. Proteomics 2019, 18 (5), 995– 1009, DOI: 10.1074/mcp.RA118.00125914https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXht1OhsbbP&md5=c4a2707f8e65bcc5fd9462e4952b5e06Streamlined protocol for deep proteomic profiling of FAC-sorted cells and its application to freshly isolated murine immune cellsMyers, Samuel A.; Rhoads, Andrew; Cocco, Alexandra R.; Peckner, Ryan; Haber, Adam L.; Schweitzer, Lawrence D.; Krug, Karsten; Mani, D. R.; Clauser, Karl R.; Rozenblatt-Rosen, Orit; Hacohen, Nir; Regev, Aviv; Carr, Steven A.Molecular & Cellular Proteomics (2019), 18 (5), 995-1009CODEN: MCPOBS; ISSN:1535-9484. (American Society for Biochemistry and Molecular Biology)Proteomic profiling describes the mol. landscape of proteins in cells immediately available to sense, transduce, and enact the appropriate responses to extracellular queues. Transcriptional profiling has proven invaluable to our understanding of cellular responses; however, insights may be lost as mounting evidence suggests transcript levels only moderately correlate with protein levels in steady state cells. Mass spectrometry-based quant. proteomics is a well-suited and widely used anal. tool for studying global protein abundances. Typical proteomic workflows are often limited by the amt. of sample input that is required for deep and quant. proteome profiling. This is esp. true if the cells of interest need to be purified by fluorescence-activated cell sorting (FACS) and one wants to avoid ex vivo culturing. To address this need, we developed an easy to implement, streamlined workflow that enables quant. proteome profiling from roughly 2μg of protein input per exptl. condition. Utilizing a combination of facile cell collection from cell sorting, solid-state isobaric labeling and multiplexing of peptides, and small-scale fractionation, we profiled the proteomes of 12 freshly isolated, primary murine immune cell types. Analyzing half of the 3e5 cells collected per cell type, we quantified over 7000 proteins across 12 key immune cell populations directly from their resident tissues. We show that low input proteomics is precise, and the data generated accurately reflects many aspects of known immunol., while expanding the list of cell-type specific proteins across the cell types profiled. The low input proteomics methods we developed are readily adaptable and broadly applicable to any cell or sample types and should enable proteome profiling in systems previously unattainable.
- 15Gao, W.; Li, H.; Liu, L.; Huang, P.; Wang, Z.; Chen, W.; Ye, M.; Yu, X.; Tian, R. An Integrated Strategy for High-Sensitive and Multi-Level Glycoproteome Analysis from Low Micrograms of Protein Samples. J. Chromatogr. A 2019, 1600, 46– 54, DOI: 10.1016/j.chroma.2019.04.04115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXosVyjsrg%253D&md5=d483a4c2b76420673a2b34063171c9b4An integrated strategy for high-sensitive and multi-level glycoproteome analysis from low micrograms of protein samplesGao, Weina; Li, Hongjie; Liu, Liping; Huang, Peiwu; Wang, Zhikun; Chen, Wendong; Ye, Mingliang; Yu, Xiaofang; Tian, RuijunJournal of Chromatography A (2019), 1600 (), 46-54CODEN: JCRAEY; ISSN:0021-9673. (Elsevier B.V.)Glycosylation, as a biol. important protein post-translational modification, often alters on both glycosites and glycans, simultaneously. However, most of current approaches focused on biased profiling of either glycosites or glycans, and limited by time-consuming process and milligrams of starting protein material. We describe here a simple and integrated spintip-based glycoproteomics technol. (termed Glyco-SISPROT) for achieving a comprehensive view of glycoproteome with shorter sample processing time and low microgram starting material. By carefully integrating and optimizing SCX, C18 and Con A (Con A) packing material and their combination in spintip format, both predigested peptides and protein lysates could be processed by Glyco-SISPROT with high efficiency. More importantly, deglycopeptide, intact glycopeptide and glycans released by multiple glycosidases could be readily collected from the same Glyco-SISPROT workflow for LC-MS anal. In total, above 1850 glycosites in ∼1770 unique deglycopeptides were characterized from mouse liver by using either 100 μg of predigested peptides or directly using 100 μg of protein lysates, in which about 30% of glycosites were released by both PNGase F and Endos. To the best of our knowledge, this approach should be one of the most comprehensive glycoproteomic approaches by using limited protein starting material. One significant benefit of Glyco-SISPROT is that whole processing time is dramatically reduced from a few days to less than 6 h with good reproducibility when protein lysates were directly processed by Glyco-SISPROT. We expect that this method will be suitable for multi-level glycoproteome anal. of rare biol. samples with high sensitivity.
- 16Chen, W.; Wang, S.; Adhikari, S.; Deng, Z.; Wang, L.; Chen, L.; Ke, M.; Yang, P.; Tian, R. Simple and Integrated Spintip-Based Technology Applied for Deep Proteome Profiling. Anal. Chem. 2016, 88 (9), 4864– 4871, DOI: 10.1021/acs.analchem.6b0063116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xls1Kis7w%253D&md5=8d60eba33022626272064cf060d9fa1eSimple and Integrated Spintip-Based Technology Applied for Deep Proteome ProfilingChen, Wendong; Wang, Shuai; Adhikari, Subash; Deng, Zuhui; Wang, Lingjue; Chen, Lan; Ke, Mi; Yang, Pengyuan; Tian, RuijunAnalytical Chemistry (Washington, DC, United States) (2016), 88 (9), 4864-4871CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Great efforts have been taken for developing high-sensitive mass spectrometry (MS)-based proteomic technologies, among which sample prepn. is one of the major focus. Here, a simple and integrated spintip-based proteomics technol. (SISPROT) consisting of strong cation exchange beads and C18 disk in one pipet tip was developed. Both proteomics sample prepn. steps, including protein preconcn., redn., alkylation, and digestion, and reversed phase (RP)-based desalting and high-pH RP-based peptide fractionation can be achieved in a fully integrated manner for the first time. This easy-to-use technol. achieved high sensitivity with negligible sample loss. Proteomic anal. of 2000 HEK 293 cells readily identified 1270 proteins within 1.4 h of MS time, while 7826 proteins were identified when 100000 cells were processed and analyzed within only 22 h of MS time. More importantly, the SISPROT can be easily multiplexed on a std. centrifuge with good reproducibility (Pearson correlation coeff. > 0.98) for both single-shot anal. and deep proteome profiling with five-step high-pH RP fractionation. The SISPROT was exemplified by the triplicate anal. of 100000 stem cells from human exfoliated deciduous teeth (SHED). This led to the identification of 9078 proteins contg. 3771 annotated membrane proteins, which was the largest proteome data set for dental stem cells reported to date. We expect that the SISPROT will be well suited for deep proteome profiling for fewer than 100000 cells and applied for translational studies where multiplexed technol. with good label-free quantification precision is required.
- 17Dou, M.; Zhu, Y.; Liyu, A.; Liang, Y.; Chen, J.; Piehowski, P. D.; Xu, K.; Zhao, R.; Moore, R. J.; Atkinson, M. A.; Mathews, C. E.; Qian, W.-J.; Kelly, R. T. Nanowell-Mediated Two-Dimensional Liquid Chromatography Enables Deep Proteome Profiling of < 1000 Mammalian Cells. Chem. Sci. 2018, 9 (34), 6944– 6951, DOI: 10.1039/C8SC02680G17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlCmtbbF&md5=1ec9556a474d130acc404d70f7a6a838Nanowell-mediated two-dimensional liquid chromatography enables deep proteome profiling of <1000 mammalian cellsDou, Maowei; Zhu, Ying; Liyu, Andrey; Liang, Yiran; Chen, Jing; Piehowski, Paul D.; Xu, Kerui; Zhao, Rui; Moore, Ronald J.; Atkinson, Mark A.; Mathews, Clayton E.; Qian, Wei-Jun; Kelly, Ryan T.Chemical Science (2018), 9 (34), 6944-6951CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Multidimensional peptide sepns. can greatly increase the depth of coverage in proteome profiling. However, a major challenge for multidimensional sepns. is the requirement of large biol. samples, often contg. milligram amts. of protein. We have developed nanowell-mediated two-dimensional (2D) reversed-phase nanoflow liq. chromatog. (LC) sepns. for in-depth proteome profiling of low-nanogram samples. Peptides are first sepd. using high-pH LC and the effluent is concatenated into 4 or 12 nanowells. The contents of each nanowell are reconstituted in LC buffer and collected for subsequent sepn. and anal. by low-pH nanoLC-MS/MS. The nanowell platform minimizes peptide losses to surfaces in offline 2D LC fractionation, enabling >5800 proteins to be confidently identified from just 50 ng of HeLa digest. Furthermore, in combination with a recently developed nanowell-based sample prepn. workflow, we demonstrated deep proteome profiling of >6000 protein groups from small populations of cells, including ∼650 HeLa cells and 10 single human pancreatic islet thin sections (∼1000 cells) from a pre-symptomatic type 1 diabetic donor.
- 18Zhu, Y.; Scheibinger, M.; Ellwanger, D. C.; Krey, J. F.; Choi, D.; Kelly, R. T.; Heller, S.; Barr-Gillespie, P. G. Single-Cell Proteomics Reveals Changes in Expression during Hair-Cell Development. eLife 2019, 8, e50777, DOI: 10.7554/eLife.5077718https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlamurbN&md5=66e9a7e7876b6b48af6f2fb378ccd1d1Single-cell proteomics reveals changes in expression during hair-cell developmentZhu, Ying; Scheibinger, Mirko; Ellwanger, Daniel Christian; Krey, Jocelyn F.; Choi, Dongseok; Kelly, Ryan T.; Heller, Stefan; Barr-Gillespie, Peter G.eLife (2019), 8 (), e50777CODEN: ELIFA8; ISSN:2050-084X. (eLife Sciences Publications Ltd.)Hearing and balance rely on small sensory hair cells that reside in the inner ear. To explore dynamic changes in the abundant proteins present in differentiating hair cells, we used nanoliter-scale shotgun mass spectrometry of single cells, each ~ 1 pL, from utricles of embryonic day 15 chickens. We identified unique constellations of proteins or protein groups from presumptive hair cells and from progenitor cells. The single-cell proteomes enabled the de novo reconstruction of a developmental trajectory using protein expression levels, revealing proteins that greatly increased in expression during differentiation of hair cells (e.g., OCM, CRABP1, GPX2, AK1, GSTO1) and those that decreased during differentiation (e.g., TMSB4X, AGR3). Complementary single-cell transcriptome profiling showed corresponding changes in mRNA during maturation of hair cells. Single-cell proteomics data thus can be mined to reveal features of cellular development that may be missed with transcriptomics.
- 19Shao, X.; Zhang, X. Design of Five-Layer Gold Nanoparticles Self-Assembled in a Liquid Open Tubular Column for Ultrasensitive Nano-LC-MS/MS Proteomic Analysis of 80 Living Cells. Proteomics 2017, 17 (8), 1600463, DOI: 10.1002/pmic.201600463There is no corresponding record for this reference.
- 20Chen, Q.; Yan, G.; Gao, M.; Zhang, X. Ultrasensitive Proteome Profiling for 100 Living Cells by Direct Cell Injection, Online Digestion and Nano-LC-MS/MS Analysis. Anal. Chem. 2015, 87 (13), 6674– 6680, DOI: 10.1021/acs.analchem.5b0080820https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVaku77P&md5=1b3f40134e4afd0f68bbec6483dd7455Ultrasensitive Proteome Profiling for 100 Living Cells by Direct Cell Injection, Online Digestion and Nano-LC-MS/MS AnalysisChen, Qi; Yan, Guoquan; Gao, Mingxia; Zhang, XiangminAnalytical Chemistry (Washington, DC, United States) (2015), 87 (13), 6674-6680CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Single-cell proteome anal. has always been an exciting goal because it provides crucial information about cellular heterogeneity and dynamic change. Here the authors presented an integrated proteome anal. device (iPAD) for 100 living cells (iPAD-100) that might be suitable for single-cell anal. Once cells were cultured, the iPAD-100 could be applied to inject 100 living cells, to transform the living cells into peptides, and to produce protein identification results with total automation. Due to the major obstacle for detection limit of mass spectrometry, the authors applied the iPAD-100 to analyze the proteome of 100 cells. In total, 813 proteins were identified in a DLD-cell proteome by three duplicate runs. Gene Ontol. anal. revealed that proteins from different cellular compartments were well-represented, including membrane proteins. The iPAD-100 greatly simplified the sampling process, reduced sample loss, and prevented contamination. As a result, proteins whose copy nos. were <1000 were identified from 100-cell samples with the iPAD-100, showing that a detection limit of 200 zmol was achieved. With increased sensitivity of mass spectrometry, the iPAD-100 may be able to reveal bountiful proteome information from a single cell in the near future.
- 21Zhu, Y.; Clair, G.; Chrisler, W. B.; Shen, Y.; Zhao, R.; Shukla, A. K.; Moore, R. J.; Misra, R. S.; Pryhuber, G. S.; Smith, R. D.; Ansong, C.; Kelly, R. T. Proteomic Analysis of Single Mammalian Cells Enabled by Microfluidic Nanodroplet Sample Preparation and Ultrasensitive NanoLC-MS. Angew. Chem., Int. Ed. 2018, 57 (38), 12370– 12374, DOI: 10.1002/anie.20180284321https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtFemur7N&md5=1c6276ba46c3866bfaeb163aee051663Proteomic Analysis of Single Mammalian Cells Enabled by Microfluidic Nanodroplet Sample Preparation and Ultrasensitive NanoLC-MSZhu, Ying; Clair, Geremy; Chrisler, William B.; Shen, Yufeng; Zhao, Rui; Shukla, Anil K.; Moore, Ronald J.; Misra, Ravi S.; Pryhuber, Gloria S.; Smith, Richard D.; Ansong, Charles; Kelly, Ryan T.Angewandte Chemie, International Edition (2018), 57 (38), 12370-12374CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)The authors report on the quant. proteomic anal. of single mammalian cells. Fluorescence-activated cell sorting was employed to deposit cells into a newly developed nanodroplet sample processing chip, after which samples were analyzed by ultrasensitive nanoLC-MS. An av. of circa 670 protein groups were confidently identified from single HeLa cells, which is a far greater level of proteome coverage for single cells than has been previously reported. The single-cell proteomics platform can be used to differentiate cell types from enzyme-dissocd. human lung primary cells and identify specific protein markers for epithelial and mesenchymal cells.
- 22Shao, X.; Wang, X.; Guan, S.; Lin, H.; Yan, G.; Gao, M.; Deng, C.; Zhang, X. Integrated Proteome Analysis Device for Fast Single-Cell Protein Profiling. Anal. Chem. 2018, 90 (23), 14003– 14010, DOI: 10.1021/acs.analchem.8b0369222https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitVart7%252FO&md5=f7f84b752d5638b2765d395b56eeecf6Integrated Proteome Analysis Device for Fast Single-Cell Protein ProfilingShao, Xi; Wang, Xuantang; Guan, Sheng; Lin, Haizhu; Yan, Guoquan; Gao, Mingxia; Deng, Chunhui; Zhang, XiangminAnalytical Chemistry (Washington, DC, United States) (2018), 90 (23), 14003-14010CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)In the authors' previous work, the authors demonstrated an integrated proteome anal. device (iPAD-100) to analyze proteomes from 100 cells. For the first time, a novel integrated device for single-cell anal. (iPAD-1) was developed to profile proteins in a single cell within 1 h. In the iPAD-1, a selected single cell was directly sucked into a 22 μm internal diam. capillary. Then the cell lysis and protein digestion were simultaneously accomplished in the capillary in a 2 nL vol., which could prevent protein loss and excessive diln. Digestion was accelerated by using elevated temp. with ultrasonication. The whole time of cell treatment was 30 min. After that, single-cell digest peptides were transferred into an LC column directly through a true zero dead vol. union, to minimize protein transfer loss. A homemade 22 μm internal diam. nano-LC packing column with 3 μm internal diam. ESI tip was used in the device to achieve ultrasensitive detection. A 30 min elution program was applied to anal. of the single-cell proteome. Therefore, the total time needed for a single-cell anal. was only 1 h. In an anal. of 10 single HeLa cells, a max. of 328 proteins were identified in one cell by using an Orbitrap Fusion Tribrid MS instrument, and the detection limit was estd. at ∼1.7-170 zmol. Such a sensitivity of the iPAD-1 was ∼120-fold higher than that of the authors' previously developed iPAD-100 system. Prominent cellular heterogeneity in protein expressive profiling was obsd. Furthermore, the authors roughly estd. the phases of the cell cycle of tested HeLa cells by the amt. of core histone proteins.
- 23Zhu, Y.; Piehowski, P. D.; Zhao, R.; Chen, J.; Shen, Y.; Moore, R. J.; Shukla, A. K.; Petyuk, V. A.; Campbell-Thompson, M.; Mathews, C. E.; Smith, R. D.; Qian, W.-J.; Kelly, R. T. Nanodroplet Processing Platform for Deep and Quantitative Proteome Profiling of 10–100 Mammalian Cells. Nat. Commun. 2018, 9 (1), 882, DOI: 10.1038/s41467-018-03367-w23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MrmtlyntQ%253D%253D&md5=ed52404cccbcdc4f8b9081ede1d092cbNanodroplet processing platform for deep and quantitative proteome profiling of 10-100 mammalian cellsZhu Ying; Zhao Rui; Kelly Ryan T; Piehowski Paul D; Shen Yufeng; Moore Ronald J; Shukla Anil K; Petyuk Vladislav A; Smith Richard D; Qian Wei-Jun; Chen Jing; Campbell-Thompson Martha; Mathews Clayton ENature communications (2018), 9 (1), 882 ISSN:.Nanoscale or single-cell technologies are critical for biomedical applications. However, current mass spectrometry (MS)-based proteomic approaches require samples comprising a minimum of thousands of cells to provide in-depth profiling. Here, we report the development of a nanoPOTS (nanodroplet processing in one pot for trace samples) platform for small cell population proteomics analysis. NanoPOTS enhances the efficiency and recovery of sample processing by downscaling processing volumes to <200 nL to minimize surface losses. When combined with ultrasensitive liquid chromatography-MS, nanoPOTS allows identification of ~1500 to ~3000 proteins from ~10 to ~140 cells, respectively. By incorporating the Match Between Runs algorithm of MaxQuant, >3000 proteins are consistently identified from as few as 10 cells. Furthermore, we demonstrate quantification of ~2400 proteins from single human pancreatic islet thin sections from type 1 diabetic and control donors, illustrating the application of nanoPOTS for spatially resolved proteome measurements from clinical tissues.
- 24Zhou, M.; Uwugiaren, N.; Williams, S. M.; Moore, R. J.; Zhao, R.; Goodlett, D.; Dapic, I.; Paša-Tolić, L.; Zhu, Y. Sensitive Top-Down Proteomics Analysis of a Low Number of Mammalian Cells Using a Nanodroplet Sample Processing Platform. Anal. Chem. 2020, 92 (10), 7087– 7095, DOI: 10.1021/acs.analchem.0c0046724https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXosFWnsrg%253D&md5=2472ebd1f191b4f3419913f0d5999fcaSensitive Top-Down Proteomics Analysis of a Low Number of Mammalian Cells Using a Nanodroplet Sample Processing PlatformZhou, Mowei; Uwugiaren, Naomi; Williams, Sarah M.; Moore, Ronald J.; Zhao, Rui; Goodlett, David; Dapic, Irena; Pasa-Tolic, Ljiljana; Zhu, YingAnalytical Chemistry (Washington, DC, United States) (2020), 92 (10), 7087-7095CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Top-down proteomics is a powerful tool for characterizing genetic variations and post-translational modifications at intact protein level. However, one significant tech. gap of top-down proteomics is the inability to analyze a low amt. of biol. samples, which limits its access to isolated rare cells, fine needle aspiration biopsies, and tissue substructures. Herein, we developed an ultrasensitive top-down platform by incorporating a microfluidic sample prepn. system, termed nanoPOTS (nanodroplet processing in one pot for trace samples), into a top-down proteomic workflow. A unique combination of a nonionic detergent dodecyl-β-D-maltopyranoside (DDM) with urea as protein extn. buffer significantly improved both protein extn. efficiency and sample recovery. We hypothesize that the DDM detergent improves protein recovery by efficiently reducing nonspecific adsorption of intact proteins on container surfaces, while urea serves as a strong denaturant to disrupt noncovalent complexes and release intact proteins for downstream anal. The nanoPOTS-based top-down platform reproducibly and quant. identified ~ 170 to ~ 620 proteoforms from ~ 70 to ~ 770 HeLa cells contg. ~ 10 to ~ 115 ng of total protein. A variety of post-translational modifications including acetylation, myristoylation, and iron binding were identified using only less than 800 cells. We anticipate the nanoPOTS top-down proteomics platform will be broadly applicable in biomedical research, particularly where clin. specimens are not available in amts. amenable to std. workflows.
- 25Xu, K.; Liang, Y.; Piehowski, P. D.; Dou, M.; Schwarz, K. C.; Zhao, R.; Sontag, R. L.; Moore, R. J.; Zhu, Y.; Kelly, R. T. Benchtop-Compatible Sample Processing Workflow for Proteome Profiling of < 100 Mammalian Cells. Anal. Bioanal. Chem. 2019, 411 (19), 4587– 4596, DOI: 10.1007/s00216-018-1493-925https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXit12ls7nJ&md5=5f57393c6315af49f67303e080dfe016Benchtop-compatible sample processing workflow for proteome profiling of < 100 mammalian cellsXu, Kerui; Liang, Yiran; Piehowski, Paul D.; Dou, Maowei; Schwarz, Kaitlynn C.; Zhao, Rui; Sontag, Ryan L.; Moore, Ronald J.; Zhu, Ying; Kelly, Ryan T.Analytical and Bioanalytical Chemistry (2019), 411 (19), 4587-4596CODEN: ABCNBP; ISSN:1618-2642. (Springer)Extending proteomics to smaller samples can enable the mapping of protein expression across tissues with high spatial resoln. and can reveal sub-group heterogeneity. However, despite the continually improving sensitivity of LC-MS instrumentation, in-depth profiling of samples contg. low-nanogram amts. of protein has remained challenging due to analyte losses incurred during prepn. and anal. To address this, the authors recently developed nanodroplet processing in one pot for trace samples (nanoPOTS), a robotic/microfluidic platform that generates ready-to-analyze peptides from cellular material in ∼200 nL droplets with greatly reduced sample losses. In combination with ultrasensitive LC-MS, nanoPOTS has enabled >3000 proteins to be confidently identified from as few as 10 cultured human cells and ∼700 proteins from single cells. However, the nanoPOTS platform requires a highly skilled operator and a costly inhouse-built robotic nanopipetting instrument. The authors sought to evaluate the extent to which the benefits of nanodroplet processing could be preserved when upscaling reagent dispensing vols. by a factor of 10 to those addressable by com. micropipette. The authors characterized the resulting platform, termed microdroplet processing in one pot for trace samples (μPOTS), for the anal. of as few as ∼25 cultured HeLa cells (4 ng total protein) or 50 μm square mouse liver tissue thin sections and found that ∼1800 and ∼1200 unique proteins were resp. identified with high reproducibility. The reduced equipment requirements should facilitate broad dissemination of nanoproteomics workflows by obviating the need for a capital-intensive custom liq. handling system.
- 26Ross-Innes, C. S.; Becq, J.; Warren, A.; Cheetham, R. K.; Northen, H.; O’Donovan, M.; Malhotra, S.; di Pietro, M.; Ivakhno, S.; He, M.; Weaver, J. M. J.; Lynch, A. G.; Kingsbury, Z.; Ross, M.; Humphray, S.; Bentley, D.; Fitzgerald, R. C. Whole-Genome Sequencing Provides New Insights into the Clonal Architecture of Barrett’s Esophagus and Esophageal Adenocarcinoma. Nat. Genet. 2015, 47 (9), 1038– 1046, DOI: 10.1038/ng.335726https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXht1WltLjN&md5=a60d7a9bfd4221c3838bdfdffc50ee2aWhole-genome sequencing provides new insights into the clonal architecture of Barrett's esophagus and esophageal adenocarcinomaRoss-Innes, Caryn S.; Becq, Jennifer; Warren, Andrew; Cheetham, R. Keira; Northen, Helen; O'Donovan, Maria; Malhotra, Shalini; di Pietro, Massimiliano; Ivakhno, Sergii; He, Miao; Weaver, Jamie M. J.; Lynch, Andy G.; Kingsbury, Zoya; Ross, Mark; Humphray, Sean; Bentley, David; Fitzgerald, Rebecca C.; Hayes, Stephen J.; Ang, Yeng; Welch, Ian; Preston, Shaun; Oakes, Sarah; Save, Vicki; Skipworth, Richard; Tucker, Olga; Davies, Jim; Crichton, Charles; Schusterreiter, Christian; Underwood, Tim; Noble, Fergus; Stacey, Bernard; Kelly, Jamie; Byrne, James; Haydon, Annette; Sharland, Donna; Owsley, Jack; Barr, Hugh; Lagergren, Jesper; Gossage, James; Davies, Andrew; Mason, Robert; Chang, Fuju; Zylstra, Janine; Sanders, Grant; Wheatley, Tim; Berrisford, Richard; Bracey, Tim; Harden, Catherine; Bunting, David; Roques, Tom; Nobes, Jenny; Loo, Suat; Lewis, Mike; Cheong, Ed; Priest, Oliver; Parsons, Simon L.; Soomro, Irshad; Kaye, Philip; Saunders, John; Pang, Vincent; Welch, Neil T.; Catton, James A.; Duffy, John P.; Ragunath, Krish; Lovat, Laurence; Haidry, Rehan; Miah, Haroon; Kerr, Sarah; Eneh, Victor; Butawan, Rommel; Roques, Tom; Lewis, Michael; Cheong, Edward; Kumar, Bhasker; Igali, Laszlo; Walton, Sharon; Dann, Adela; Safranek, Peter; Hindmarsh, Andy; Sudjendran, Vijayendran; Scott, Michael; Cluroe, Alison; Miremadi, Ahmad; Mahler-Araujo, Betania; Nutzinger, Barbara; Peters, Chris; Abdullahi, Zarah; Crawte, Jason; MacRae, Shona; Noorani, Ayesha; Elliott, Rachael Fels; Bower, Lawrence; Edwards, Paul; Tavare, Simon; Eldridge, Matthew; Bornschein, Jan; Secrier, Maria; Yang, Tsun-Po; O'Neill, J. Robert; Adamczuk, Kasia; Lao-Sirieix, Pierre; Grehan, Nicola; Smith, Laura; Lishman, Suzy; Beardsmore, Duncan; Dawson, SarahNature Genetics (2015), 47 (9), 1038-1046CODEN: NGENEC; ISSN:1061-4036. (Nature Publishing Group)The mol. genetic relationship between esophageal adenocarcinoma (EAC) and its precursor lesion, Barrett's esophagus, is poorly understood. Using whole-genome sequencing on 23 paired Barrett's esophagus and EAC samples, together with one in-depth Barrett's esophagus case study sampled over time and space, we have provided the following new insights: (i) Barrett's esophagus is polyclonal and highly mutated even in the absence of dysplasia; (ii) when cancer develops, copy no. increases and heterogeneity persists such that the spectrum of mutations often shows surprisingly little overlap between EAC and adjacent Barrett's esophagus; and (iii) despite differences in specific coding mutations, the mutational context suggests a common causative insult underlying these two conditions. From a clin. perspective, the histopathol. assessment of dysplasia appears to be a poor reflection of the mol. disarray within the Barrett's epithelium, and a mol. Cytosponge technique overcomes sampling bias and has the capacity to reflect the entire clonal architecture.
- 27Peters, Y.; Al-Kaabi, A.; Shaheen, N. J.; Chak, A.; Blum, A.; Souza, R. F.; Di Pietro, M.; Iyer, P. G.; Pech, O.; Fitzgerald, R. C.; Siersema, P. D. Barrett Oesophagus. Nat. Rev. Dis. Primer 2019, 5 (1), 35, DOI: 10.1038/s41572-019-0086-zThere is no corresponding record for this reference.
- 28Hofmann, A. F. The Continuing Importance of Bile Acids in Liver and Intestinal Disease. Arch. Intern. Med. 1999, 159 (22), 2647– 2658, DOI: 10.1001/archinte.159.22.264728https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3c%252FmsVKhtw%253D%253D&md5=4f017c841da85f0723c292fb31e70037The continuing importance of bile acids in liver and intestinal diseaseHofmann A FArchives of internal medicine (1999), 159 (22), 2647-58 ISSN:0003-9926.Bile acids, the water-soluble, amphipathic end products of cholesterol metabolism, are involved in liver, biliary, and intestinal disease. Formed in the liver, bile acids are absorbed actively from the small intestine, with each molecule undergoing multiple enterohepatic circulations before being excreted. After their synthesis from cholesterol, bile acids are conjugated with glycine or taurine, a process that makes them impermeable to cell membranes and permits high concentrations to persist in bile and intestinal content. The relation between the chemical structure and the multiple physiological functions of bile acids is reviewed. Bile acids induce biliary lipid secretion and solubilize cholesterol in bile, promoting its elimination. In the small intestine, bile acids solubilize dietary lipids promoting their absorption. Bile acids are cytotoxic when present in abnormally high concentrations. This may occur intracellularly, as occurs in the hepatocyte in cholestasis, or extracellularly, as occurs in the colon in patients with bile acid malabsorption. Disturbances in bile acid metabolism can be caused by (1) defective biosynthesis from cholesterol or defective conjugation, (2) defective membrane transport in the hepatocyte or ileal enterocyte, (3) defective transport between organs or biliary diversion, and (4) increased bacterial degradation during enterohepatic cycling. Bile acid therapy involves bile acid replacement in deficiency states or bile acid displacement by ursodeoxycholic acid, a noncytotoxic bile acid. In cholestatic liver disease, administration of ursodeoxycholic acid decreases hepatocyte injury by retained bile acids, improving liver tests, and slowing disease progression. Bile acid malabsorption may lead to high concentrations of bile acids in the colon and impaired colonic mucosal function; bile acid sequestrants provide symptomatic benefit for diarrhea. A knowledge of bile acid physiology and the perturbations of bile acid metabolism in liver and digestive disease should be useful for the internist.
- 29Fitzgerald, R. C.; di Pietro, M.; O’Donovan, M.; Maroni, R.; Muldrew, B.; Debiram-Beecham, I.; Gehrung, M.; Offman, J.; Tripathi, M.; Smith, S. G.; Aigret, B.; Walter, F. M.; Rubin, G.; Bagewadi, A.; Patrick, A.; Shenoy, A.; Redmond, A.; Muddu, A.; Northrop, A.; Groves, A.; Shiner, A.; Heer, A.; Takhar, A.; Bowles, A.; Jarman, A.; Wong, A.; Lucas, A.; Gibbons, A.; Dhar, A.; Curry, A.; Lalonde, A.; Swinburn, A.; Turner, A.; Lydon, A.-M.; Gunstone, A.; Lee, A.; Nambi, A.; Ariyarathenam, A.; Elden, A.; Wilson, A.; Donepudi, B.; Campbell, B.; Uszycka, B.; Bowers, B.; Coghill, B.; de Quadros, B.; Cheah, C.; Bratten, C.; Brown, C.; Moorbey, C.; Clisby, C.; Gordon, C.; Schramm, C.; Castle, C.; Newark, C.; Norris, C.; A’Court, C.; Graham, C.; Fletcher, C.; Grocott, C.; Rees, C.; Bakker, C.; Paschalides, C.; Vickery, C.; Schembri, D.; Morris, D.; Hagan, D.; Cronk, D.; Goddard, D.; Graham, D.; Phillips, D.; Prabhu, D.; Kejariwal, D.; Garg, D.; Lonsdale, D.; Butterworth, D.; Clements, D.; Bradman, D.; Blake, D.; Mather, E.; O’Farrell, E.; Markowetz, F.; Adams, F.; Pesola, F.; Forbes, G.; Taylor, G.; Collins, G.; Irvine, G.; Fourie, G.; Doyle, H.; Barnes, H.; Bowyer, H.; Whiting, H.; Beales, I.; Binnian, I.; Bremner, I.; Jennings, I.; Troiceanu, I.; Modelell, I.; Emmerson, I.; Ortiz, J.; Lilley, J.; Harvey, J.; Vicars, J.; Takhar, J.; Larcombe, J.; Bornschein, J.; Aldegather, J.; Johnson, J.; Ducker, J.; Skinner, J.; Dash, J.; Walsh, J.; Miralles, J.; Ridgway, J.; Ince, J.; Kennedy, J.; Hampson, K.; Milne, K.; Ellerby, K.; Priddis, K.; Rainsbury, K.; Powell, K.; Gunner, K.; Ragunath, K.; Knox, K.; Baseley, L.; White, L.; Lovat, L.; Berney, L.; Crockett, L.; Murray, L.; Westwood, L.; Chalkley, L.; Leggett, L.; Dale, L.; Scovell, L.; Brooks, L.; Saunders, L.; Owen, L.; Dilwershah, M.; Baldry, M.; Corcoran, M.; Roy, M.; Macedo, M.; Attah, M.; Anson, M.-J.; Rutter, M.; Wallard, M.; Gaw, M.; Hunt, M.; Lea-Hagerty, M.; Penacerrada, M.; Bianchi, M.; Baker-Moffatt, M.; Czajkowski, M.; Sleeth, M.; Brewer, N.; Wooding, N.; Todd, N.; Millen, N.; Zolle, O.; Whitehead, O.; Ojechi, P.; Moore, P.; Banim, P.; Spellar, P.; Bhandari, P.; Kant, P.; Nixon, R.; Russell, R.; Roberts, R.; Skule, R.; West, R.; Fox, R.; Beesley, R.; Gibbins, R.; Osborne, R.; Thiagarajan, S.; Bastiman, S.; Warburton, S.; Pai, S.; Leith-Russell, S.; Utting, S.; Watson, S.; Wytrykowski, S.; Singh, S.; Malhotra, S.; Woods, S.; Conway, S.; Mateer, S.; Macrae, S.; Singh, S.; Fourie, S.; Campbell, S.; Parslow-Williams, S.; Goel, S.; Dellar, S.; Jones, S.; Knight, S.; Mackay-Thomas, S.; Mukherjee, S.; Allen, S.; Henry, S.; Evans, T.; Leighton, T.; Bray, T.; Shackleton, T.; Santosh, V.; Glover, V.; Chandraraj, V.; Elson, W.; Briggs, W.; Barron, Z.; Khan, Z.; Sasieni, P. Cytosponge-Trefoil Factor 3 versus Usual Care to Identify Barrett’s Oesophagus in a Primary Care Setting: A Multicentre, Pragmatic, Randomised Controlled Trial. Lancet 2020, 396 (10247), 333– 344, DOI: 10.1016/S0140-6736(20)31099-029https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38jpvVGrtQ%253D%253D&md5=b0b2f86fdf8f90356abf375b51307abeCytosponge-trefoil factor 3 versus usual care to identify Barrett's oesophagus in a primary care setting: a multicentre, pragmatic, randomised controlled trialFitzgerald Rebecca C; di Pietro Massimiliano; O'Donovan Maria; Tripathi Monika; Maroni Roberta; Muldrew Beth; Aigret Benoit; Sasieni Peter; Debiram-Beecham Irene; Gehrung Marcel; Offman Judith; Smith Samuel G; Walter Fiona M; Rubin GregLancet (London, England) (2020), 396 (10247), 333-344 ISSN:.BACKGROUND: Treatment of dysplastic Barrett's oesophagus prevents progression to adenocarcinoma; however, the optimal diagnostic strategy for Barrett's oesophagus is unclear. The Cytosponge-trefoil factor 3 (TFF3) is a non-endoscopic test for Barrett's oesophagus. The aim of this study was to investigate whether offering this test to patients on medication for gastro-oesophageal reflux would increase the detection of Barrett's oesophagus compared with standard management. METHODS: This multicentre, pragmatic, randomised controlled trial was done in 109 socio-demographically diverse general practice clinics in England. Randomisation was done both at the general practice clinic level (cluster randomisation) and at the individual patient level, and the results for each type of randomisation were analysed separately before being combined. Patients were eligible if they were aged 50 years or older, had been taking acid-suppressants for symptoms of gastro-oesophageal reflux for more than 6 months, and had not undergone an endoscopy procedure within the past 5 years. General practice clinics were selected by the local clinical research network and invited to participate in the trial. For cluster randomisation, clinics were randomly assigned (1:1) by the trial statistician using a computer-generated randomisation sequence; for individual patient-level randomisation, patients were randomly assigned (1:1) by the general practice clinics using a centrally prepared computer-generated randomisation sequence. After randomisation, participants received either standard management of gastro-oesophageal reflux (usual care group), in which participants only received an endoscopy if required by their general practitioner, or usual care plus an offer of the Cytosponge-TFF3 procedure, with a subsequent endoscopy if the procedure identified TFF3-positive cells (intervention group). The primary outcome was the diagnosis of Barrett's oesophagus at 12 months after enrolment, expressed as a rate per 1000 person-years, in all participants in the intervention group (regardless of whether they had accepted the offer of the Cytosponge-TFF3 procedure) compared with all participants in the usual care group. Analyses were intention-to-treat. The trial is registered with the ISRCTN registry, ISRCTN68382401, and is completed. FINDINGS: Between March 20, 2017, and March 21, 2019, 113 general practice clinics were enrolled, but four clinics dropped out shortly after randomisation. Using an automated search of the electronic prescribing records of the remaining 109 clinics, we identified 13 657 eligible patients who were sent an introductory letter with 14 days to opt out. 13 514 of these patients were randomly assigned (per practice or at the individual patient level) to the usual care group (n=6531) or the intervention group (n=6983). Following randomisation, 149 (2%) of 6983 participants in the intervention group and 143 (2%) of 6531 participants in the usual care group, on further scrutiny, did not meet all eligibility criteria or withdrew from the study. Of the remaining 6834 participants in the intervention group, 2679 (39%) expressed an interest in undergoing the Cytosponge-TFF3 procedure. Of these, 1750 (65%) met all of the eligibility criteria on telephone screening and underwent the procedure. Most of these participants (1654 [95%]; median age 69 years) swallowed the Cytosponge successfully and produced a sample. 231 (3%) of 6834 participants had a positive Cytosponge-TFF3 result and were referred for an endoscopy. Patients who declined the offer of the Cytosponge-TFF3 procedure and all participants in the usual care group only had an endoscopy if deemed necessary by their general practitioner. During an average of 12 months of follow-up, 140 (2%) of 6834 participants in the intervention group and 13 (<1%) of 6388 participants in the usual care group were diagnosed with Barrett's oesophagus (absolute difference 18·3 per 1000 person-years [95% CI 14·8-21·8]; rate ratio adjusted for cluster randomisation 10·6 [95% CI 6·0-18·8], p<0·0001). Nine (<1%) of 6834 participants were diagnosed with dysplastic Barrett's oesophagus (n=4) or stage I oesophago-gastric cancer (n=5) in the intervention group, whereas no participants were diagnosed with dysplastic Barrett's oesophagus or stage I gastro-oesophageal junction cancer in the usual care group. Among 1654 participants in the intervention group who swallowed the Cytosponge device successfully, 221 (13%) underwent endoscopy after testing positive for TFF3 and 131 (8%, corresponding to 59% of those having an endoscopy) were diagnosed with Barrett's oesophagus or cancer. One patient had a detachment of the Cytosponge from the thread requiring endoscopic removal, and the most common side-effect was a sore throat in 63 (4%) of 1654 participants. INTERPRETATION: In patients with gastro-oesophageal reflux, the offer of Cytosponge-TFF3 testing results in improved detection of Barrett's oesophagus. Cytosponge-TFF3 testing could also lead to the diagnosis of treatable dysplasia and early cancer. This strategy will lead to additional endoscopies with some false positive results. FUNDING: Cancer Research UK, National Institute for Health Research, the UK National Health Service, Medtronic, and the Medical Research Council.
- 30Ross-Innes, C. S.; Chettouh, H.; Achilleos, A.; Galeano-Dalmau, N.; Debiram-Beecham, I.; MacRae, S.; Fessas, P.; Walker, E.; Varghese, S.; Evan, T.; Lao-Sirieix, P. S.; O’Donovan, M.; Malhotra, S.; Novelli, M.; Disep, B.; Kaye, P. V.; Lovat, L. B.; Haidry, R.; Griffin, M.; Ragunath, K.; Bhandari, P.; Haycock, A.; Morris, D.; Attwood, S.; Dhar, A.; Rees, C.; Rutter, M. D.; Ostler, R.; Aigret, B.; Sasieni, P. D.; Fitzgerald, R. C. Risk Stratification of Barrett’s Oesophagus Using a Non-Endoscopic Sampling Method Coupled with a Biomarker Panel: A Cohort Study. Lancet Gastroenterol. Hepatol. 2017, 2 (1), 23– 31, DOI: 10.1016/S2468-1253(16)30118-230https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cvnt1OmtQ%253D%253D&md5=e51955fc3160fbf1f2d64146831713ddRisk stratification of Barrett's oesophagus using a non-endoscopic sampling method coupled with a biomarker panel: a cohort studyRoss-Innes Caryn S; Chettouh Hamza; Achilleos Achilleas; Galeano-Dalmau Nuria; Debiram-Beecham Irene; MacRae Shona; Fessas Petros; Walker Elaine; Varghese Sibu; Evan Theodore; Lao-Sirieix Pierre S; O'Donovan Maria; Malhotra Shalini; Novelli Marco; Lovat Laurence B; Haidry Rehan; Disep Babett; Griffin Michael; Kaye Phillip V; Ragunath Krish; Bhandari Pradeep; Haycock Adam; Morris Danielle; Attwood Stephen; Dhar Anjan; Rees Colin; Rutter Matt D; Ostler Richard; Aigret Benoit; Sasieni Peter D; Fitzgerald Rebecca CThe lancet. Gastroenterology & hepatology (2017), 2 (1), 23-31 ISSN:.BACKGROUND: Barrett's oesophagus predisposes to adenocarcinoma. However, most patients with Barrett's oesophagus will not progress and endoscopic surveillance is invasive, expensive, and fraught by issues of sampling bias and the subjective assessment of dysplasia. We investigated whether a non-endoscopic device, the Cytosponge, could be coupled with clinical and molecular biomarkers to identify a group of patients with low risk of progression suitable for non-endoscopic follow-up. METHODS: In this multicentre cohort study (BEST2), patients with Barrett's oesophagus underwent the Cytosponge test before their surveillance endoscopy. We collected clinical and demographic data and tested Cytosponge samples for a molecular biomarker panel including three protein biomarkers (P53, c-Myc, and Aurora kinase A), two methylation markers (MYOD1 and RUNX3), glandular atypia, and TP53 mutation status. We used a multivariable logistic regression model to compute the conditional probability of dysplasia status. We selected a simple model with high classification accuracy and applied it to an independent validation cohort. The BEST2 study is registered with ISRCTN, number 12730505. FINDINGS: The discovery cohort consisted of 468 patients with Barrett's oesophagus and intestinal metaplasia. Of these, 376 had no dysplasia and 22 had high-grade dysplasia or intramucosal adenocarcinoma. In the discovery cohort, a model with high classification accuracy consisted of glandular atypia, P53 abnormality, and Aurora kinase A positivity, and the interaction of age, waist-to-hip ratio, and length of the Barrett's oesophagus segment. 162 (35%) of 468 of patients fell into the low-risk category and the probability of being a true non-dysplastic patient was 100% (99% CI 96-100) and the probability of having high-grade dysplasia or intramucosal adenocarcinoma was 0% (0-4). 238 (51%) of participants were classified as of moderate risk; the probability of having high-grade dysplasia was 14% (9-21). 58 (12%) of participants were classified as high-risk; the probability of having non-dysplastic endoscopic biopsies was 13% (5-27), whereas the probability of having high-grade dysplasia or intramucosal adenocarcinoma was 87% (73-95). In the validation cohort (65 patients), 51 were non-dysplastic and 14 had high-grade dysplasia. In this cohort, 25 (38%) of 65 patients were classified as being low-risk, and the probability of being non-dysplastic was 96·0% (99% CI 73·80-99·99). The moderate-risk group comprised 27 non-dysplastic and eight high-grade dysplasia cases, whereas the high-risk group (8% of the cohort) had no non-dysplastic cases and five patients with high-grade dysplasia. INTERPRETATION: A combination of biomarker assays from a single Cytosponge sample can be used to determine a group of patients at low risk of progression, for whom endoscopy could be avoided. This strategy could help to avoid overdiagnosis and overtreatment in patients with Barrett's oesophagus. FUNDING: Cancer Research UK.
- 31Williams, S. M.; Liyu, A. V.; Tsai, C.-F.; Moore, R. J.; Orton, D. J.; Chrisler, W. B.; Gaffrey, M. J.; Liu, T.; Smith, R. D.; Kelly, R. T.; Pasa-Tolic, L.; Zhu, Y. Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography–Mass Spectrometry for High-Throughput Single-Cell Proteomics. Anal. Chem. 2020, 92 (15), 10588– 10596, DOI: 10.1021/acs.analchem.0c0155131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtlCmtL3O&md5=11a723a01a532b3fd5dbb8f43fb00134Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography-Mass Spectrometry for High-Throughput Single-Cell ProteomicsWilliams, Sarah M.; Liyu, Andrey V.; Tsai, Chia-Feng; Moore, Ronald J.; Orton, Daniel J.; Chrisler, William B.; Gaffrey, Matthew J.; Liu, Tao; Smith, Richard D.; Kelly, Ryan T.; Pasa-Tolic, Ljiljana; Zhu, YingAnalytical Chemistry (Washington, DC, United States) (2020), 92 (15), 10588-10596CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Single-cell proteomics can provide crit. biol. insight into the cellular heterogeneity that is masked by bulk-scale anal. The authors have developed a nanoPOTS (nanodroplet processing in one pot for trace samples) platform and demonstrated its broad applicability for single-cell proteomics. However, because of nanoliter-scale sample vols., the nanoPOTS platform is not compatible with automated LC-MS systems, which significantly limits sample throughput and robustness. To address this challenge, the authors have developed a nanoPOTS autosampler allowing fully automated sample injection from nanowells to LC-MS systems. The authors also developed a sample drying, extn., and loading workflow to enable reproducible and reliable sample injection. The sequential anal. of 20 samples contg. 10 ng tryptic peptides demonstrated high reproducibility with correlation coeffs. of >0.995 between any two samples. The nanoPOTS autosampler can provide anal. throughput of 9.6, 16, and 24 single cells per day using 120, 60, and 30 min LC gradients, resp. As a demonstration for single-cell proteomics, the autosampler was first applied to profiling protein expression in single MCF10A cells using a label-free approach. At a throughput of 24 single cells per day, an av. of 256 proteins was identified from each cell and the no. was increased to 731 when the Match Between Runs algorithm of MaxQuant was used. Using a multiplexed isobaric labeling approach (TMT-11plex), ~ 77 single cells could be analyzed per day. The authors analyzed 152 cells from three acute myeloid leukemia cell lines, resulting in a total of 2558 identified proteins with 1465 proteins quantifiable (70% valid values) across the 152 cells. These data showed quant. single-cell proteomics can cluster cells to distinct groups and reveal functionally distinct differences.
- 32DEP: Differential Enrichment analysis of Proteomics data version 1.10.0 from Bioconductor, https://rdrr.io/bioc/DEP/ (accessed Jul 4, 2020).There is no corresponding record for this reference.
- 33Zhang, X.; Smits, A. H.; van Tilburg, G. B.; Ovaa, H.; Huber, W.; Vermeulen, M. Proteome-Wide Identification of Ubiquitin Interactions Using UbIA-MS. Nat. Protoc. 2018, 13 (3), 530– 550, DOI: 10.1038/nprot.2017.14733https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisl2ku74%253D&md5=88719f04dffb8fffe078df606dd6f5d7Proteome-wide identification of ubiquitin interactions using UbIA-MSZhang, Xiaofei; Smits, Arne H.; van Tilburg, Gabrielle B. A.; Ovaa, Huib; Huber, Wolfgang; Vermeulen, MichielNature Protocols (2018), 13 (3), 530-550CODEN: NPARDW; ISSN:1750-2799. (Nature Research)Ubiquitin-binding proteins play an important role in eukaryotes by translating differently linked polyubiquitin chains into proper cellular responses. Current knowledge about ubiquitin-binding proteins and ubiquitin linkage-selective interactions is mostly based on case-by-case studies. We have recently reported a method called ubiquitin interactor affinity enrichment-mass spectrometry (UbIA-MS), which enables comprehensive identification of ubiquitin interactors for all ubiquitin linkages from crude cell lysates. One major strength of UbIA-MS is the fact that ubiquitin interactors are enriched from crude cell lysates, in which proteins are present at endogenous levels, contain biol. relevant post-translational modifications (PTMs) and are assembled in native protein complexes. In addn., UbIA-MS uses chem. synthesized nonhydrolyzable diubiquitin, which mimics native diubiquitin and is inert to cleavage by endogenous deubiquitinases (DUBs). Here, we present a detailed protocol for UbIA-MS that proceeds in five stages: (i) chem. synthesis of ubiquitin precursors and click chem. for the generation of biotinylated nonhydrolyzable diubiquitin baits, (ii) in vitro affinity purifn. of ubiquitin interactors, (iii) on-bead interactor digestion, (iv) liq. chromatog. (LC)-MS/MS anal. and (v) data anal. to identify differentially enriched proteins. The computational anal. tools are freely available as an open-source R software package, including a graphical interface. Typically, UbIA-MS allows the identification of dozens to hundreds of ubiquitin interactors from any type of cell lysate, and can be used to study cell type or stimulus-dependent ubiquitin interactions. The nonhydrolyzable diubiquitin synthesis can be completed in 3 wk, followed by ubiquitin interactor enrichment and identification, which can be completed within another 2 wk.
- 34test_diff: Differential enrichment test in DEP: Differential Enrichment analysis of Proteomics data, https://rdrr.io/bioc/DEP/man/test_diff.html (accessed Jul 5, 2020).There is no corresponding record for this reference.
- 35Ritchie, M. E.; Phipson, B.; Wu, D.; Hu, Y.; Law, C. W.; Shi, W.; Smyth, G. K. Limma Powers Differential Expression Analyses for RNA-Sequencing and Microarray Studies. Nucleic Acids Res. 2015, 43 (7), e47– e47, DOI: 10.1093/nar/gkv00735https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFaiu7%252FN&md5=e8a5f44160c650ad89adc48a9912bd96limma powers differential expression analyses for RNA-sequencing and microarray studiesRitchie, Matthew E.; Phipson, Belinda; Wu, Di; Hu, Yifang; Law, Charity W.; Shi, Wei; Smyth, Gordon K.Nucleic Acids Research (2015), 43 (7), e47/1-e47/13CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Limma is an R/Bioconductor software package that provides an integrated soln. for analyzing data from gene expression expts. It contains rich features for handling complex exptl. designs and for information borrowing to overcome the problem of small sample sizes. Over the past decade, limma has been a popular choice for gene discovery through differential expression analyses of microarray and high-throughput PCR data. The package contains particularly strong facilities for reading, normalizing and exploring such data. Recently, the capabilities of limma have been significantly expanded in two important directions. First, the package can now perform both differential expression and differential splicing analyses of RNA sequencing (RNA-seq) data. All the downstream anal. tools previously restricted to microarray data are now available for RNA-seq as well. These capabilities allow users to analyze both RNA-seq and microarray data with very similar pipelines. Second, the package is now able to go past the traditional gene-wise expression analyses in a variety of ways, analyzing expression profiles in terms of co-regulated sets of genes or in terms of higher-order expression signatures. This provides enhanced possibilities for biol. interpretation of gene expression differences. This article reviews the philosophy and design of the limma package, summarizing both new and historical features, with an emphasis on recent enhancements and features that have not been previously described.
- 36P’ng, C.; Green, J.; Chong, L. C.; Waggott, D.; Prokopec, S. D.; Shamsi, M.; Nguyen, F.; Mak, D. Y. F.; Lam, F.; Albuquerque, M. A.; Wu, Y.; Jung, E. H.; Starmans, M. H. W.; Chan-Seng-Yue, M. A.; Yao, C. Q.; Liang, B.; Lalonde, E.; Haider, S.; Simone, N. A.; Sendorek, D.; Chu, K. C.; Moon, N. C.; Fox, N. S.; Grzadkowski, M. R.; Harding, N. J.; Fung, C.; Murdoch, A. R.; Houlahan, K. E.; Wang, J.; Garcia, D. R.; de Borja, R.; Sun, R. X.; Lin, X.; Chen, G. M.; Lu, A.; Shiah, Y.-J.; Zia, A.; Kearns, R.; Boutros, P. C. BPG: Seamless, Automated and Interactive Visualization of Scientific Data. BMC Bioinf. 2019, 20 (1), 42, DOI: 10.1186/s12859-019-2610-236https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cjjs1altA%253D%253D&md5=61ec34f7005b4dfc1819b7e76300d80fBPG: Seamless, automated and interactive visualization of scientific dataP'ng Christine; Green Jeffrey; Chong Lauren C; Waggott Daryl; Prokopec Stephenie D; Shamsi Mehrdad; Nguyen Francis; Mak Denise Y F; Lam Felix; Albuquerque Marco A; Wu Ying; Jung Esther H; Starmans Maud H W; Chan-Seng-Yue Michelle A; Yao Cindy Q; Liang Bianca; Lalonde Emilie; Haider Syed; Simone Nicole A; Sendorek Dorota; Chu Kenneth C; Moon Nathalie C; Fox Natalie S; Grzadkowski Michal R; Harding Nicholas J; Fung Clement; Murdoch Amanda R; Houlahan Kathleen E; Wang Jianxin; Garcia David R; de Borja Richard; Sun Ren X; Lin Xihui; Chen Gregory M; Lu Aileen; Shiah Yu-Jia; Zia Amin; Kearns Ryan; Boutros Paul C; Yao Cindy Q; Lalonde Emilie; Fox Natalie S; Houlahan Kathleen E; Shiah Yu-Jia; Boutros Paul C; Wang Jianxin; Sun Ren X; Lu Aileen; Boutros Paul C; Boutros Paul C; Boutros Paul C; Boutros Paul C; Boutros Paul CBMC bioinformatics (2019), 20 (1), 42 ISSN:.BACKGROUND: We introduce BPG, a framework for generating publication-quality, highly-customizable plots in the R statistical environment. RESULTS: This open-source package includes multiple methods of displaying high-dimensional datasets and facilitates generation of complex multi-panel figures, making it suitable for complex datasets. A web-based interactive tool allows online figure customization, from which R code can be downloaded for integration with computational pipelines. CONCLUSION: BPG provides a new approach for linking interactive and scripted data visualization and is available at http://labs.oicr.on.ca/boutros-lab/software/bpg or via CRAN at https://cran.r-project.org/web/packages/BoutrosLab.plotting.general.
- 37Hulsen, T.; de Vlieg, J.; Alkema, W. BioVenn – a Web Application for the Comparison and Visualization of Biological Lists Using Area-Proportional Venn Diagrams. BMC Genomics 2008, 9 (1), 488, DOI: 10.1186/1471-2164-9-48837https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1cjksF2ltQ%253D%253D&md5=1c79e2fa6accc5ff3c4cd77bab9aca8cBioVenn - a web application for the comparison and visualization of biological lists using area-proportional Venn diagramsHulsen Tim; de Vlieg Jacob; Alkema WynandBMC genomics (2008), 9 (), 488 ISSN:.BACKGROUND: In many genomics projects, numerous lists containing biological identifiers are produced. Often it is useful to see the overlap between different lists, enabling researchers to quickly observe similarities and differences between the data sets they are analyzing. One of the most popular methods to visualize the overlap and differences between data sets is the Venn diagram: a diagram consisting of two or more circles in which each circle corresponds to a data set, and the overlap between the circles corresponds to the overlap between the data sets. Venn diagrams are especially useful when they are 'area-proportional' i.e. the sizes of the circles and the overlaps correspond to the sizes of the data sets. Currently there are no programs available that can create area-proportional Venn diagrams connected to a wide range of biological databases. RESULTS: We designed a web application named BioVenn to summarize the overlap between two or three lists of identifiers, using area-proportional Venn diagrams. The user only needs to input these lists of identifiers in the textboxes and push the submit button. Parameters like colors and text size can be adjusted easily through the web interface. The position of the text can be adjusted by 'drag-and-drop' principle. The output Venn diagram can be shown as an SVG or PNG image embedded in the web application, or as a standalone SVG or PNG image. The latter option is useful for batch queries. Besides the Venn diagram, BioVenn outputs lists of identifiers for each of the resulting subsets. If an identifier is recognized as belonging to one of the supported biological databases, the output is linked to that database. Finally, BioVenn can map Affymetrix and EntrezGene identifiers to Ensembl genes. CONCLUSION: BioVenn is an easy-to-use web application to generate area-proportional Venn diagrams from lists of biological identifiers. It supports a wide range of identifiers from the most used biological databases currently available. Its implementation on the World Wide Web makes it available for use on any computer with internet connection, independent of operating system and without the need to install programs locally. BioVenn is freely accessible at http://www.cmbi.ru.nl/cdd/biovenn/.
- 38Perez-Riverol, Y.; Csordas, A.; Bai, J.; Bernal-Llinares, M.; Hewapathirana, S.; Kundu, D. J.; Inuganti, A.; Griss, J.; Mayer, G.; Eisenacher, M.; Pérez, E.; Uszkoreit, J.; Pfeuffer, J.; Sachsenberg, T.; Yılmaz, Ş.; Tiwary, S.; Cox, J.; Audain, E.; Walzer, M.; Jarnuczak, A. F.; Ternent, T.; Brazma, A.; Vizcaíno, J. A. The PRIDE Database and Related Tools and Resources in 2019: Improving Support for Quantification Data. Nucleic Acids Res. 2019, 47 (D1), D442– D450, DOI: 10.1093/nar/gky110638https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs1GqtrzK&md5=4cd0f929c2df840a5cc0631d0d708d6cThe PRIDE database and related tools and resources in 2019: improving support for quantification dataPerez-Riverol, Yasset; Csordas, Attila; Bai, Jingwen; Bernal-Llinare, Manuel; Hewapathirana, Suresh; Kundu, Deepti J.; Iuganti, Avinash; Griss, Johannes; Mayer, Gerhard; Eisenacher, Martin; Perez, Enrique; Uszkoreit, Julian; Pfeuffer, Julianus; Sachsenberg, Timo; Yilmaz, Sule; Tiwary, Shivani; Cox, Jurgen; Audain, Enrique; Walzer, Mathias; Jarnuczak, Andrew F.; Ternent, Tobias; Brazma, Alvis; Vizcaino, Juan AntonioNucleic Acids Research (2019), 47 (D1), D442-D450CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. The PRoteomics IDEntifications (PRIDE) database is the world's largest data repository of mass spectrometry-based proteomics data, and is one of the founding members of the global ProteomeXchange (PX) consortium. In this manuscript, we summarize the developments in PRIDE resources and related tools since the previous update manuscript was published in Nucleic Acids Research in 2016. In the last 3 years, public data sharing through PRIDE (as part of PX) has definitely become the norm in the field. In parallel, data re-use of public proteomics data has increased enormously, with multiple applications. We first describe the new architecture of PRIDE Archive, the archival component of PRIDE. PRIDE Archive and the related data submission framework have been further developed to support the increase in submitted data vols. and addnl. data types. A new scalable and fault tolerant storage backed, Application Programming Interface and web interface have been implemented, as a part of an ongoing process. Addnl., we emphasize the improved support for quant. proteomics data through the mzTab format. At last, we outline key statistics on the current data contents and vol. of downloads, and how PRIDE data are starting to be disseminated to added-value resources including Ensembl, UniProt and Expression Atlas.
- 39UniProt, https://www.uniprot.org/ (accessed Jun 25, 2020).There is no corresponding record for this reference.
- 40Protein GRAVY, https://www.bioinformatics.org/sms2/protein_gravy.html (accessed Jun 25, 2020).There is no corresponding record for this reference.
- 41Budnik, B.; Levy, E.; Harmange, G.; Slavov, N. SCoPE-MS: Mass Spectrometry of Single Mammalian Cells Quantifies Proteome Heterogeneity during Cell Differentiation. Genome Biol. 2018, 19 (1), 161, DOI: 10.1186/s13059-018-1547-541https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXisFKls7nM&md5=bfdeda4d526356689afaa4c6d97a20d5SCoPE-MS: mass spectrometry of single mammalian cells quantifies proteome heterogeneity during cell differentiationBudnik, Bogdan; Levy, Ezra; Harmange, Guillaume; Slavov, NikolaiGenome Biology (2018), 19 (), 161/1-161/12CODEN: GNBLFW; ISSN:1474-760X. (BioMed Central Ltd.)Some exciting biol. questions require quantifying thousands of proteins in single cells. To achieve this goal, we develop Single Cell ProtEomics by Mass Spectrometry (SCoPE-MS) and validate its ability to identify distinct human cancer cell types based on their proteomes. We use SCoPE-MS to quantify over a thousand proteins in differentiating mouse embryonic stem cells. The single-cell proteomes enable us to deconstruct cell populations and infer protein abundance relationships. Comparison between single-cell proteomes and transcriptomes indicates coordinated mRNA and protein covariation, yet many genes exhibit functionally concerted and distinct regulatory patterns at the mRNA and the protein level.
- 42Dou, M.; Clair, G.; Tsai, C.-F.; Xu, K.; Chrisler, W. B.; Sontag, R. L.; Zhao, R.; Moore, R. J.; Liu, T.; Pasa-Tolic, L.; Smith, R. D.; Shi, T.; Adkins, J. N.; Qian, W.-J.; Kelly, R. T.; Ansong, C.; Zhu, Y. High-Throughput Single Cell Proteomics Enabled by Multiplex Isobaric Labelling in a Nanodroplet Sample Preparation Platform. Anal. Chem. 2019, 91 (20), 13119– 13127, DOI: 10.1021/acs.analchem.9b0334942https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslKisbbJ&md5=800d1bf514934f3fe80ae23fa44729d7High-Throughput Single Cell Proteomics Enabled by Multiplex Isobaric Labeling in a Nanodroplet Sample Preparation PlatformDou, Maowei; Clair, Geremy; Tsai, Chia-Feng; Xu, Kerui; Chrisler, William B.; Sontag, Ryan L.; Zhao, Rui; Moore, Ronald J.; Liu, Tao; Pasa-Tolic, Ljiljana; Smith, Richard D.; Shi, Tujin; Adkins, Joshua N.; Qian, Wei-Jun; Kelly, Ryan T.; Ansong, Charles; Zhu, YingAnalytical Chemistry (Washington, DC, United States) (2019), 91 (20), 13119-13127CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Effective extension of mass spectrometry-based proteomics to single cells remains challenging. Herein the authors combined microfluidic nanodroplet technol. with tandem mass tag (TMT) isobaric labeling to significantly improve anal. throughput and proteome coverage for single mammalian cells. Isobaric labeling facilitated multiplex anal. of single cell-sized protein quantities to a depth of ∼1600 proteins with median CV of 10.9% and correlation coeff. of 0.98. To demonstrate in-depth high throughput single cell anal., the platform was applied to measure protein expression in 72 single cells from three murine cell populations (epithelial, immune, and endothelial cells) in <2 days instrument time with over 2300 proteins identified. Principal component anal. grouped the single cells into three distinct populations based on protein expression with each population characterized by well-known cell-type specific markers. The platform enables high throughput and unbiased characterization of single cell heterogeneity at the proteome level.
- 43Weng, S. S. H.; Demir, F.; Ergin, E. K.; Dirnberger, S.; Uzozie, A.; Tuscher, D.; Nierves, L.; Tsui, J.; Huesgen, P. F.; Lange, P. F. Sensitive Determination of Proteolytic Proteoforms in Limited Microscale Proteome Samples. Mol. Cell. Proteomics 2019, 18 (11), 2335– 2347, DOI: 10.1074/mcp.TIR119.00156043https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXisVSjsrjN&md5=5f4d95ae406fe34978aa8860add4fa9cSensitive determination of proteolytic proteoforms in limited microscale proteome samplesWeng, Samuel S. H.; Demir, Fatih; Ergin, Enes K.; Dirnberger, Sabrina; Uzozie, Anuli; Tuscher, Domenic; Nierves, Lorenz; Tsui, Janice; Huesgen, Pitter F.; Lange, Philipp F.Molecular & Cellular Proteomics (2019), 18 (11), 2335-2347CODEN: MCPOBS; ISSN:1535-9484. (American Society for Biochemistry and Molecular Biology)Protein N termini unambiguously identify truncated, alternatively translated or modified proteoforms with distinct functions and reveal perturbations in disease. Selective enrichment of N-terminal peptides is necessary to achieve proteome-wide coverage for unbiased identification of site-specific regulatory proteolytic processing and protease substrates. However, many proteolytic processes are strictly confined in time and space and therefore can only be analyzed in minute samples that provide insufficient starting material for current enrichment protocols. Here we present High-efficiency Undecanal-based N Termini EnRichment (HUNTER), a robust, sensitive and scalable method for the anal. of previously inaccessible microscale samples. HUNTER achieved identification of >1000 N termini from as little as 2 mug raw HeLa cell lysate. Broad applicability is demonstrated by the first N-terminome anal. of sorted human primary immune cells and enriched mitochondrial fractions from pediatric cancer patients, as well as protease substrate identification from individual Arabidopsis thaliana wild type and Vacuolar Processing Enzyme-deficient mutant seedlings. We further implemented the workflow on a liq. handling system and demonstrate the feasibility of clin. degradomics by automated processing of liq. biopsies from pediatric cancer patients.
- 44Kasuga, K.; Katoh, Y.; Nagase, K.; Igarashi, K. Microproteomics with Microfluidic-Based Cell Sorting: Application to 1000 and 100 Immune Cells. Proteomics 2017, 17 (13–14), 1600420, DOI: 10.1002/pmic.201600420There is no corresponding record for this reference.
- 45Sun, J.; Zhang, G. L.; Li, S.; Ivanov, A. R.; Fenyo, D.; Lisacek, F.; Murthy, S. K.; Karger, B. L.; Brusic, V. Pathway Analysis and Transcriptomics Improve Protein Identification by Shotgun Proteomics from Samples Comprising Small Number of Cells - a Benchmarking Study. BMC Genomics 2014, 15 (Suppl 9), S1, DOI: 10.1186/1471-2164-15-S9-S1There is no corresponding record for this reference.
- 46Tanca, A.; Abbondio, M.; Pisanu, S.; Pagnozzi, D.; Uzzau, S.; Addis, M. F. Critical Comparison of Sample Preparation Strategies for Shotgun Proteomic Analysis of Formalin-Fixed, Paraffin-Embedded Samples: Insights from Liver Tissue. Clin. Proteomics 2014, 11 (1), 28, DOI: 10.1186/1559-0275-11-2846https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhslShsLjJ&md5=407da191800181ff40883e9c36fef0c3Critical comparison of sample preparation strategies for shotgun proteomic analysis of formalin-fixed, paraffin-embedded samples: insights from liver tissueTanca, Alessandro; Abbondio, Marcello; Pisanu, Salvatore; Pagnozzi, Daniela; Uzzau, Sergio; Addis, Maria FilippaClinical Proteomics (2014), 11 (), 28/1-28/12, 12 pp.CODEN: CPLRCX; ISSN:1542-6416. (BioMed Central Ltd.)Background: The growing field of formalin-fixed paraffin-embedded (FFPE) tissue proteomics holds promise for improving translational research. Direct tissue trypsinization (DT) and protein extn. followed by in soln. digestion (ISD) or filter-aided sample prepn. (FASP) are the most common workflows for shotgun anal. of FFPE samples, but a crit. comparison of the different methods is currently lacking. Exptl. Design: DT, FASP and ISD workflows were compared by subjecting to the same label-free quant. approach three independent tech. replicates of each method applied to FFPE liver tissue. Data were evaluated in terms of method reproducibility and protein/peptide distribution according to localization, MW, pI and hydrophobicity. Results: DT showed lower reproducibility, good preservation of high-MW proteins, a general bias towards hydrophilic and acidic proteins, much lower keratin contamination, as well as higher abundance of non-tryptic peptides. Conversely, FASP and ISD proteomes were depleted in high-MW proteins and enriched in hydrophobic and membrane proteins; FASP provided higher identification yields, while ISD exhibited higher reproducibility. Conclusions: These results highlight that diverse sample prepn. strategies provide significantly different proteomic information, and present typical biases that should be taken into account when dealing with FFPE samples. When a sufficient amt. of tissue is available, the complementary use of different methods is suggested to increase proteome coverage and depth.
- 47Föll, M. C.; Fahrner, M.; Oria, V. O.; Kühs, M.; Biniossek, M. L.; Werner, M.; Bronsert, P.; Schilling, O. Reproducible Proteomics Sample Preparation for Single FFPE Tissue Slices Using Acid-Labile Surfactant and Direct Trypsinization. Clin. Proteomics 2018, 15 (1), 11, DOI: 10.1186/s12014-018-9188-y47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MrpvFarsg%253D%253D&md5=48eb2283b6a607be9332be0d1ddf983bReproducible proteomics sample preparation for single FFPE tissue slices using acid-labile surfactant and direct trypsinizationFoll Melanie Christine; Fahrner Matthias; Oria Victor Oginga; Biniossek Martin Lothar; Schilling Oliver; Foll Melanie Christine; Fahrner Matthias; Oria Victor Oginga; Fahrner Matthias; Oria Victor Oginga; Kuhs Markus; Werner Martin; Bronsert Peter; Kuhs Markus; Werner Martin; Bronsert Peter; Kuhs Markus; Werner Martin; Bronsert Peter; Werner Martin; Bronsert Peter; Schilling Oliver; Schilling OliverClinical proteomics (2018), 15 (), 11 ISSN:1542-6416.BACKGROUND: Proteomic analyses of clinical specimens often rely on human tissues preserved through formalin-fixation and paraffin embedding (FFPE). Minimal sample consumption is the key to preserve the integrity of pathological archives but also to deal with minimal invasive core biopsies. This has been achieved by using the acid-labile surfactant RapiGest in combination with a direct trypsinization (DTR) strategy. A critical comparison of the DTR protocol with the most commonly used filter aided sample preparation (FASP) protocol is lacking. Furthermore, it is unknown how common histological stainings influence the outcome of the DTR protocol. METHODS: Four single consecutive murine kidney tissue specimens were prepared with the DTR approach or with the FASP protocol using both 10 and 30 k filter devices and analyzed by label-free, quantitative liquid chromatography-tandem mass spectrometry (LC-MS/MS). We compared the different protocols in terms of proteome coverage, relative label-free quantitation, missed cleavages, physicochemical properties and gene ontology term annotations of the proteins. Additionally, we probed compatibility of the DTR protocol for the analysis of common used histological stainings, namely hematoxylin & eosin (H&E), hematoxylin and hemalaun. These were proteomically compared to an unstained control by analyzing four human tonsil FFPE tissue specimens per condition. RESULTS: On average, the DTR protocol identified 1841 ± 22 proteins in a single, non-fractionated LC-MS/MS analysis, whereas these numbers were 1857 ± 120 and 1970 ± 28 proteins for the FASP 10 and 30 k protocol. The DTR protocol showed 15% more missed cleavages, which did not adversely affect quantitation and intersample comparability. Hematoxylin or hemalaun staining did not adversely impact the performance of the DTR protocol. A minor perturbation was observed for H&E staining, decreasing overall protein identification by 13%. CONCLUSIONS: In essence, the DTR protocol can keep up with the FASP protocol in terms of qualitative and quantitative reproducibility and performed almost as well in terms of proteome coverage and missed cleavages. We highlight the suitability of the DTR protocol as a viable and straightforward alternative to the FASP protocol for proteomics-based clinical research.
- 48Broeckx, V.; Boonen, K.; Pringels, L.; Sagaert, X.; Prenen, H.; Landuyt, B.; Schoofs, L.; Maes, E. Comparison of Multiple Protein Extraction Buffers for GeLC-MS/MS Proteomic Analysis of Liver and Colon Formalin-Fixed, Paraffin-Embedded Tissues. Mol. BioSyst. 2016, 12 (2), 553– 565, DOI: 10.1039/C5MB00670H48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvF2hsrvK&md5=1e245f198339d7405f0c8709bbc4e44aComparison of multiple protein extraction buffers for GeLC-MS/MS proteomic analysis of liver and colon formalin-fixed, paraffin-embedded tissuesBroeckx, Valerie; Boonen, Kurt; Pringels, Lentel; Sagaert, Xavier; Prenen, Hans; Landuyt, Bart; Schoofs, Liliane; Maes, EvelyneMolecular BioSystems (2016), 12 (2), 553-565CODEN: MBOIBW; ISSN:1742-2051. (Royal Society of Chemistry)Formalin-fixed paraffin-embedded (FFPE) tissue specimens represent a potential valuable source of samples for clin. research. Since these specimens are banked in hospital archives, large cohorts of samples can be collected in short periods of time which can all be linked with a patients' clin. history. Therefore, the use of FFPE tissue in protein biomarker discovery studies gains interest. However, despite the growing no. of FFPE proteome studies in the literature, there is a lack of a FFPE proteomics std. operating procedure (SOP). One of the challenging steps in the development of such a SOP is the ability to obtain an efficient and repeatable extn. of full length FFPE proteins. In this study, the protein extn. efficiency of eight protein extn. buffers is critically compared with GeLC-MS/MS (1D gel electrophoresis followed by in-gel digestion and LC-MS/MS). The data variation caused by using these extn. buffers was investigated since the variation is a very important aspect when using FFPE tissue as a source for biomarker detection. In addn., a qual. comparison was made between the protein extn. efficiency and repeatability for FFPE tissue and fresh frozen tissue.
- 49Tabb, D. L.; Vega-Montoto, L.; Rudnick, P. A.; Variyath, A. M.; Ham, A.-J. L.; Bunk, D. M.; Kilpatrick, L. E.; Billheimer, D. D.; Blackman, R. K.; Cardasis, H. L.; Carr, S. A.; Clauser, K. R.; Jaffe, J. D.; Kowalski, K. A.; Neubert, T. A.; Regnier, F. E.; Schilling, B.; Tegeler, T. J.; Wang, M.; Wang, P.; Whiteaker, J. R.; Zimmerman, L. J.; Fisher, S. J.; Gibson, B. W.; Kinsinger, C. R.; Mesri, M.; Rodriguez, H.; Stein, S. E.; Tempst, P.; Paulovich, A. G.; Liebler, D. C.; Spiegelman, C. Repeatability and Reproducibility in Proteomic Identifications by Liquid Chromatography–Tandem Mass Spectrometry. J. Proteome Res. 2010, 9 (2), 761– 776, DOI: 10.1021/pr900636549https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsFGnt7bP&md5=77c1052918728af58b3b5c4a5bc250a9Repeatability and Reproducibility in Proteomic Identifications by Liquid Chromatography-Tandem Mass SpectrometryTabb, David L.; Vega-Montoto, Lorenzo; Rudnick, Paul A.; Variyath, Asokan Mulayath; Ham, Amy-Joan L.; Bunk, David M.; Kilpatrick, Lisa E.; Billheimer, Dean D.; Blackman, Ronald K.; Cardasis, Helene L.; Carr, Steven A.; Clauser, Karl R.; Jaffe, Jacob D.; Kowalski, Kevin A.; Neubert, Thomas A.; Regnier, Fred E.; Schilling, Birgit; Tegeler, Tony J.; Wang, Mu; Wang, Pei; Whiteaker, Jeffrey R.; Zimmerman, Lisa J.; Fisher, Susan J.; Gibson, Bradford W.; Kinsinger, Christopher R.; Mesri, Mehdi; Rodriguez, Henry; Stein, Stephen E.; Tempst, Paul; Paulovich, Amanda G.; Liebler, Daniel C.; Spiegelman, CliffJournal of Proteome Research (2010), 9 (2), 761-776CODEN: JPROBS; ISSN:1535-3893. (American Chemical Society)The complexity of proteomic instrumentation for LC-MS/MS introduces many possible sources of variability. Data-dependent sampling of peptides constitutes a stochastic element at the heart of discovery proteomics. Although this variation impacts the identification of peptides, proteomic identifications are far from completely random. In this study, the authors analyzed interlab. data sets from the NCI Clin. Proteomic Technol. Assessment for Cancer to examine repeatability and reproducibility in peptide and protein identifications. Included data spanned 144 LC-MS/MS expts. on four Thermo LTQ and four Orbitrap instruments. Samples included yeast lysate, the NCI-20 defined dynamic range protein mix, and the Sigma UPS 1 defined equimolar protein mix. Some of the authors' findings reinforced conventional wisdom, such as repeatability and reproducibility being higher for proteins than for peptides. Most lessons from the data, however, were more subtle. Orbitrap proved capable of higher repeatability and reproducibility, but aberrant performance occasionally erased these gains. Even the simplest protein digestions yielded more peptide ions than LC-MS/MS could identify during a single expt. The authors obsd. that peptide lists from pairs of tech. replicates overlapped by 35-60%, giving a range for peptide-level repeatability in these expts. Sample complexity did not appear to affect peptide identification repeatability, even as nos. of identified spectra changed by an order of magnitude. Statistical anal. of protein spectral counts revealed greater stability across tech. replicates for Orbitrap, making them superior to LTQ instruments for biomarker candidate discovery. The most repeatable peptides were those corresponding to conventional tryptic cleavage sites, those that produced intense MS signals, and those that resulted from proteins generating many distinct peptides. Reproducibility among different instruments of the same type lagged behind repeatability of tech. replicates on a single instrument by several percent. These findings reinforce the importance of evaluating repeatability as a fundamental characteristic of anal. technologies.
- 50Delmotte, N.; Lasaosa, M.; Tholey, A.; Heinzle, E.; van Dorsselaer, A.; Huber, C. G. Repeatability of Peptide Identifications in Shotgun Proteome Analysis Employing Off-Line Two-Dimensional Chromatographic Separations and Ion-Trap MS. J. Sep. Sci. 2009, 32 (8), 1156– 1164, DOI: 10.1002/jssc.20080061550https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXlvVCltr8%253D&md5=81ccb5ce2f00a4a8fc0eec13bf94fd05Repeatability of peptide identifications in shotgun proteome analysis employing off-line two-dimensional chromatographic separations and ion-trap MSDelmotte, Nathanael; Lasaosa, Maria; Tholey, Andreas; Heinzle, Elmar; van Dorsselaer, Alain; Huber, Christian G.Journal of Separation Science (2009), 32 (8), 1156-1164CODEN: JSSCCJ; ISSN:1615-9306. (Wiley-VCH Verlag GmbH & Co. KGaA)The repeatability of peptide identifications in shotgun proteome analyses employing strong cation-exchange- × ion-pair RP HPLC hyphenated to ESI MS/MS was compared to an alternative scheme, comprising high-pH RP chromatog. combined with low-pH ion-pair RP chromatog. Equivalent results were obtained with both methods in proteome anal. of Corynebacterium glutamicum. From a total no. of 1350 to 1850 peptides identified in triplicate analyses of five consecutive fractions chosen from the first-dimension sepn., 41-45% of the peptides were identified three times, whereas 16-22 and 37-39% of the peptides were identified only twice or once, resp. A comparison of the repeatability of peptide identifications from complex samples upon 1- or 2-D chromatog. sepn. revealed that an addnl. sepn. dimension decreases the repeatability by approx. 25%.
- 51Resing, K. A.; Meyer-Arendt, K.; Mendoza, A. M.; Aveline-Wolf, L. D.; Jonscher, K. R.; Pierce, K. G.; Old, W. M.; Cheung, H. T.; Russell, S.; Wattawa, J. L.; Goehle, G. R.; Knight, R. D.; Ahn, N. G. Improving Reproducibility and Sensitivity in Identifying Human Proteins by Shotgun Proteomics. Anal. Chem. 2004, 76 (13), 3556– 3568, DOI: 10.1021/ac035229m51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXktlWmsbs%253D&md5=056551e9a48b0bdb1d0f5668bb9625a9Improving Reproducibility and Sensitivity in Identifying Human Proteins by Shotgun ProteomicsResing, Katheryn A.; Meyer-Arendt, Karen; Mendoza, Alex M.; Aveline-Wolf, Lauren D.; Jonscher, Karen R.; Pierce, Kevin G.; Old, William M.; Cheung, Hiu T.; Russell, Steven; Wattawa, Joy L.; Goehle, Geoff R.; Knight, Robin D.; Ahn, Natalie G.Analytical Chemistry (2004), 76 (13), 3556-3568CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)Identifying proteins in cell exts. by shotgun proteomics involves digesting the proteins, sequencing the resulting peptides by data-dependent mass spectrometry (MS/MS), and searching protein databases to identify the proteins from which the peptides are derived. Manual anal. and direct spectral comparison reveal that scores from two commonly used search programs (Sequest and Mascot) validate less than half of potentially identifiable MS/MS spectra (class pos.) from shotgun analyses of the human erythroleukemia K562 cell line. Here we demonstrate increased sensitivity and accuracy using a focused search strategy along with a peptide sequence validation script that does not rely exclusively on XCorr or Mowse scores generated by Sequest or Mascot, but uses consensus between the search programs, along with chem. properties and scores describing the nature of the fragmentation spectrum (ion score and RSP). The approach yielded 4.2% false pos. and 8% false neg. frequencies in peptide assignments. The protein profile is then assembled from peptide assignments using a novel peptide-centric protein nomenclature that more accurately reports protein variants that contain identical peptide sequences. An Isoform Resolver algorithm ensures that the protein count is not inflated by variants in the protein database, eliminating ∼25% of redundant proteins. Anal. of sol. proteins from a human K562 cells identified 5130 unique proteins, with ∼100 false pos. protein assignments.
- 52Washburn, M. P.; Ulaszek, R. R.; Yates, J. R. Reproducibility of Quantitative Proteomic Analyses of Complex Biological Mixtures by Multidimensional Protein Identification Technology. Anal. Chem. 2003, 75 (19), 5054– 5061, DOI: 10.1021/ac034120b52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXmsVegsLs%253D&md5=93f083bea4d279e1cf92ea0f6248a8c3Reproducibility of quantitative proteomic analyses of complex biological mixtures by multidimensional protein identification technologyWashburn, Michael P.; Ulaszek, Ryan R.; Yates, John R., IIIAnalytical Chemistry (2003), 75 (19), 5054-5061CODEN: ANCHAM; ISSN:0003-2700. (American Chemical Society)If quant. proteomic technologies are to be of widespread use to the biol. community, the reproducibility of each method must be investigated and detd. We have analyzed the reproducibility of complex quant. proteomic analyses of metabolically labeled S. cerevisiae analyzed via multidimensional protein identification technol. (MudPIT). Three independent cell growths of S. cerevisiae grown in rich and minimal media and independent MudPIT analyses of each were compared and contrasted. Quant. MudPIT was found to be intra- and interexperimentally reproducible at both the peptide and protein levels. Proteins of potential low abundance were detected, identified, and quantified by identical peptides from three independent samples. In addn., when multiple peptides were matched to a protein, the relative abundance of each peptide was in agreement across the three samples. Despite the reproducibility, errors in the exptl. detn. of protein expression levels occurred, but the impact of the variation was minimized by replicate expts. Last, quant. MudPIT analyses will likely be improved by increasing the no. of peptide hits per protein in a given anal., which will provide for greater intraexperimental reproducibility.
- 53Magdeldin, S.; Yamamoto, T. Toward Deciphering Proteomes of Formalin-Fixed Paraffin-Embedded (FFPE) Tissues. Proteomics 2012, 12 (7), 1045– 1058, DOI: 10.1002/pmic.20110055053https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XlvVKms78%253D&md5=7cd5165b42a902e467226a114103f3c1Toward deciphering proteomes of formalin-fixed paraffin-embedded (FFPE) tissuesMagdeldin, Sameh; Yamamoto, TadashiProteomics (2012), 12 (7), 1045-1058CODEN: PROTC7; ISSN:1615-9853. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Formalin-fixed paraffin-embedded (FFPE) tissue specimens comprise a potentially valuable resource for both prospective and retrospective biomarker discovery. Unlocking the proteomic profile of clinicopathol. FFPE tissues is a critically essential step for annotating clin. findings and predicting biomarkers for ultimate disease prognosis and therapeutic follow-up.
- 54Maes, E.; Broeckx, V.; Mertens, I.; Sagaert, X.; Prenen, H.; Landuyt, B.; Schoofs, L. Analysis of the Formalin-Fixed Paraffin-Embedded Tissue Proteome: Pitfalls, Challenges, and Future Prospectives. Amino Acids 2013, 45 (2), 205– 218, DOI: 10.1007/s00726-013-1494-054https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFemtbvN&md5=cfb25a4dc12c7a6a9b76472d8c4da015Analysis of the formalin-fixed paraffin-embedded tissue proteome: pitfalls, challenges, and future prospectivesMaes, Evelyne; Broeckx, Valerie; Mertens, Inge; Sagaert, Xavier; Prenen, Hans; Landuyt, Bart; Schoofs, LilianeAmino Acids (2013), 45 (2), 205-218CODEN: AACIE6; ISSN:0939-4451. (Springer-Verlag GmbH)A review. Formalin-fixed paraffin-embedded (FFPE) tissues are a real treasure for retrospective anal. considering the amt. of samples present in hospital archives, combined with pathol., clin., and outcome information available for every sample. Although unlocking the proteome of these tissues is still a challenge, new approaches are being developed. In this review, we summarize the different mass spectrometry platforms that are used in human clin. studies to unravel the FFPE proteome. The different ways of extg. crosslinked proteins and the anal. strategies are pointed out. Also, the pitfalls and challenges concerning the quality of FFPE proteomic approaches are depicted. We also evaluated the potential of these anal. methods for future clin. FFPE proteomics applications.
- 55Proungvitaya, S.; Klinthong, W.; Proungvitaya, T.; Limpaiboon, T.; Jearanaikoon, P.; Roytrakul, S.; Wongkham, C.; Nimboriboonporn, A.; Wongkham, S. High Expression of CCDC25 in Cholangiocarcinoma Tissue Samples. Oncol. Lett. 2017, 14 (2), 2566– 2572, DOI: 10.3892/ol.2017.644655https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cflslWhsg%253D%253D&md5=dbbf13668447ea01c723e4bf965dca50High expression of CCDC25 in cholangiocarcinoma tissue samplesProungvitaya Siriporn; Klinthong Wachiraya; Proungvitaya Tanakorn; Limpaiboon Temduang; Jearanaikoon Patcharee; Proungvitaya Siriporn; Proungvitaya Tanakorn; Limpaiboon Temduang; Jearanaikoon Patcharee; Wongkham Chaisiri; Nimboriboonporn Anongporn; Wongkham Sopit; Roytrakul Sittiruk; Nimboriboonporn AnongpornOncology letters (2017), 14 (2), 2566-2572 ISSN:1792-1074.Cholangiocarcinoma (CCA) is a malignant transformation of biliary epithelial cells. It is a slow growing tumor, but is also highly metastatic with a poor prognosis. Bile acids are known to transactivate the epidermal growth factor receptor (EGFR) in cholangiocytes and induce cyclooxygenase-2 expression. The protein expression profiles of bile acid-treated CCA cells were studied using a proteomic approach. To elucidate the possible mechanisms involved in the bile acid-mediated enhancement of CCA cell migration, the effects of six bile acids, including cholic, deoxycholic, taurocholic, taurodeoxycholic, glycocholic and glycodeoxycholic acid, on the migration of CCA cells were examined in vitro using wound healing assays. Subsequently, the possible proteins involved in enhanced CCA cell migration were investigated using a proteomic approach. Changes to the protein expression profiles of CCA cells following bile acid treatment was examined using two-dimensional electrophoresis and mass spectrometry. The results demonstrated that cholic and deoxycholic acid significantly enhanced the migration of CCA cells, compared with the treated MMNK-1 control cells. CCA cells had 77 overexpressed protein spots following cholic acid treatment, and 50 protein spots following deoxycholic acid treatment, compared with the treated MMNK-1 control cells. Liquid chromatography tandem-mass spectrometry analysis revealed that coiled-coil domain containing 25 (CCDC25) was significantly overexpressed in cholic acid-treated CCA cells compared with in cholic acid-treated control cells. When the expression levels of CCDC25 were investigated using western blot analysis, CCDC25 was demonstrated to be highly expressed in CCA tissues, but not in the adjacent non-cancerous tissue samples. The identified proteins were further analyzed for protein-chemical interactions using STITCH version 3.1 software. CCDC25 protein was identified to be associated with Son of sevenless homolog 1 and growth factor receptor-bound protein 2, which are involved in EGFR signaling. The results of the present study demonstrated that following cholic acid treatment, CCDC25 is overexpressed in CCA cells, which is associated with significantly enhanced cell migration. This suggests that CCDC25 is a potential therapeutic target for the treatment of patients with CCA.
- 56Brychtova, V.; Vojtesek, B.; Hrstka, R. Anterior Gradient 2: A Novel Player in Tumor Cell Biology. Cancer Lett. 2011, 304 (1), 1– 7, DOI: 10.1016/j.canlet.2010.12.02356https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXjsFWru7g%253D&md5=6baccba91e15dfacd80ed874ae409574Anterior gradient 2: A novel player in tumor cell biologyBrychtova, Veronika; Vojtesek, Borivoj; Hrstka, RomanCancer Letters (New York, NY, United States) (2011), 304 (1), 1-7CODEN: CALEDQ; ISSN:0304-3835. (Elsevier)A review. AGR2 has evolutionarily conserved roles in development and tissue regeneration and is linked with several human cancers. The exact functions and regulation of AGR2 are poorly understood, but current data identify AGR2 as a clin. relevant factor that modulates the behavior and response of hormone-dependent cancers (breast, prostate) and hormone-independent cancers (colorectal, pancreatic, esophageal and other common cancers). AGR2 protein expression induces metastasis, acts as a p53 tumor suppressor inhibitor and survival factor, participates directly in neoplastic transformation and is involved in drug resistance. Thus, AGR2 is an important tumor biomarker and neg. prognostic factor potentially exploitable in clin. practice.
- 57Suwanmanee, G.; Yosudjai, J.; Phimsen, S.; Wongkham, S.; Jirawatnotai, S.; Kaewkong, W. Upregulation of AGR2vH Facilitates Cholangiocarcinoma Cell Survival under Endoplasmic Reticulum Stress via the Activation of the Unfolded Protein Response Pathway. Int. J. Mol. Med. 2019, 45 (2), 669– 677, DOI: 10.3892/ijmm.2019.4432There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jproteome.0c00629.
Figure S1: Venn diagram illustrating the shared number of identified proteins among different sample types, as well as proteins that are unique to each sample type; Figure S2: Number of missed cleavages are shown for all replicates and are expressed in percentage; Figure S3: Qualitative assessment of reproducibility of the microPOTS system; Figure S4: Quantitative assessment of reproducibility of the microPOTS system; Table S1: Median coefficient of variation (CV) for quantile normalized protein LFQ values for each sample type; Box S1: Sample preparation and LC-MS analysis for bulk proteomics data set; Box S2: Bulk data analysis and comparison to microPOTS data; Figure S5: Scatter plot with associated Pearson’s correlation coefficient (R = 0.625) between microPOTS and bulk proteomics for all overlapping 1066 proteins that were identified; Figure S6: Assessment of physicochemical characteristics; Figure S7: Assessment of physicochemical characteristics; Figure S8: Subcellular localization; Table S2: List of differentially expressed proteins between CP-A dKO cells with or without LCA treatment; Table S3: List of differentially expressed proteins between CP-A dKO cells with or without X-ray treatment (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.