博碩士論文 108223046 詳細資訊




以作者查詢圖書館館藏 以作者查詢臺灣博碩士 以作者查詢全國書目 勘誤回報 、線上人數:39 、訪客IP:18.116.118.198
姓名 陳柏宇(Bo-Yu Chen)  查詢紙本館藏   畢業系所 化學學系
論文名稱 開發精簡及迅速的方法應用於深度磷酸化蛋白質體學剖析
(Development of Streamlined and Rapid Method for Deep Phosphoproteomics Profiling)
檔案 [Endnote RIS 格式]    [Bibtex 格式]    [相關文章]   [文章引用]   [完整記錄]   [館藏目錄]   至系統瀏覽論文 (2026-12-31以後開放)
摘要(中) 蛋白質的磷酸化是細胞訊息傳遞網路中相當普遍的轉譯後修飾,不正常的磷酸化表現會使訊息傳遞異常,並且通常與人類疾病的發生或進展有關。磷酸肽純化及高性能質譜技術的進步已被應用於大規模細胞與臨床樣本的磷酸化蛋白質體分析。然而,目前在微量磷酸化蛋白質體學技術,大都還需要數百微克的蛋白質樣品,限制其分析臨床、稀少微量樣本(小於200微克蛋白質起始量,例如穿刺)的應用,開發具分析微量樣品能力之高靈敏度磷酸蛋白質體學方法為重要議題。近年來,微流體裝置已被用於蛋白質體樣品製備,但其在磷酸化蛋白質體學上的應用仍有相當大的挑戰及有待開發。因此具備的。
為了達到靈敏、簡易的散彈式磷酸化蛋白質體學流程,本篇論文中我們針對微量樣本開發了一個簡化的樣品製備流程,以1~200微克的肺癌PC9細胞株作為模型。首先,評估相容於胰蛋白酶裂解與層析質譜的介面活性劑脫氧膽酸鈉(SDC)的優化條件,從而省略蛋白質沉澱和水解後脫鹽步驟。在不除鹽的情況下,使用 1% 脫氧膽酸鈉 (SDC) 作為裂解緩衝液和 加入50% 異丙醇 (IPA) 在磷酸肽純化緩衝液中導致磷酸胜肽減少 1.5 倍。通過甲酸酸化濃度從 6%增加到 50%,專一性從 46% 增加到 84%,磷酸肽數目從 1725 增加到 2255。此外,乳酸助於防止非特異性結合,將純化磷酸肽的特異性從 84% 提高到 92%。 最後,我們使用 0.5% TFA 代替 50% FA 以減少樣品量。磷酸肽數目和專一性結果相似(0.5% TFA鑑定到 2071條,專一性 87%,50% FA鑑定到 2139條,專一性 88%)。其次,用蛋白質 LoBind 管 (500μL) 替換 StageTip (20μL) 可減少蛋白質吸附,並通過減少純化肽使用的二氧化鈦 (TiO2) 的量來防止非專一性結合,磷酸肽數目提高了 1.5 倍 ( 559 至 889 條磷酸肽),專一性從 72% 提高到 87%。此外,蛋白質與胰蛋白酶的比例從 1:50 優化到 1:10,以將消化效率從 16 小時加速到 2 小時。最後,結合數據非依賴性採集(DIA)方法,評估此樣品製備方法的靈敏度。我們從1-10µg細胞中鑑定出 1,093-4,478 個磷酸肽和 1,306-6,560 個磷酸位點,而library-based DIA 方法將鑑定覆蓋率提高了 5.3 倍(5,804-14,968磷酸肽和 1,441-6,560)。總結來說,本篇研究成功開發了一種簡化、快速和穩定的磷酸蛋白質體學分析策略。與最近發表的從 1μg 裂解物中鑑定 600 種磷酸肽(1011 個磷酸位點)的最佳鑑定結果相比。我們的結果顯示出優越的鑑定結果。此方法可應用於臨床或稀有樣品的磷酸蛋白質體學分析,研究異常磷酸化與疾病潛在機制的相關性。
摘要(英) Protein phosphorylation is the most ubiquitous post-translational modification (PTM) for regulation of cellular signaling networks. Aberrant protein phosphorylation results in abnormal signaling cascades and is often associated with initiation or progression of human diseases. Advancements in phosphopeptide enrichment and high-performance mass spectrometry-based shotgun have demonstrated large-scale phosphoproteomics analysis for various applications from cell line to clinical specimens. However, current sample preparation workflow in the phosphoproteomics analysis often requires samples at a hundred-microgram level of protein, which limits its implementation for micro-scale sample, i.e. < 200µg protein starting amount (i.e. < 200µg protein starting amount [1]) to study clinical or rare samples such as biopsy. Recently, microfluidic platforms have been developed for low-loss proteome sample preparation, while the application of phosphoproteomics still remains an enormous challenge. Therefore, a highly sensitive sample processing pipeline of phosphoproteomics is an urgent need and is worth development.
To achieve a sensitive and simplified shotgun phosphoproteomic workflow, in this thesis, we developed a streamlined sample preparation workflow for microscale sample (microgram). The PC9 cell line from 1-200µg was used as a model in this study. Firstly, optimized conditions for the detergent sodium deoxycholate (SDC) were evaluated to be compatible with tryptic digestion and LC-MS/MS, which allows bypassing the protein precipitation and post-digestion desalting steps. Without desalting, the use of 1% sodium deoxycholate (SDC) as lysis buffer and 50% isopropanol (IPA) in phosphopeptide enrichment buffer caused 1.5-fold less phosphopeptides. By more acidification concentration from 6% to 50% Formic acid, resulting in the specificity increased from 46% to 84% and phosphopeptides identification increased from 1725 to 2255. The use of lactic acid helped to prevent non-specific binding and improved the specificity from 84% to 92% with phosphopeptide identification similar to control (1932 for control and 1923 for adding lactic acid). In the end, we use 0.5% TFA to replace 50% FA for reducing sample volume. The phosphopeptide ID and specificity was similar between two acids (2071, 87% for 0.5% TFA and 2139, 88% for 50% FA). Secondarily, minimizing protein absorption by replacing the the StageTip (20µL) with Protein LoBind Tube (500µL) and prevent non-specific binding was achieved by reducing the amount of titania oxide (TiO2) beads from 1.5mg to 0.625mg for 7.5µg peptide, which enhanced specificity from 72% to 87% and 1.5-fold identification of 559 to 889 phosphopeptides. Moreover, the protein-to-trypsin ratio was optimized from 1:50 to 1:10 to accelerate the tryptic digestion efficiency from 16 hours to 2 hours. The total time spent of the entire workflow was successfully reduced to 4hrs. Finally, the sensitivity of this majorized workflow was examined by using a series dilution sample with data dependent acquisition (DDA) and data independent acquisition (DIA). For phosphopeptide identification, we identified 1,093-4,478 phosphopeptides and 1,306-6,560 phosphosites from 1-10 µg lysate, while the library-based DIA approach improved the identification coverage up to 5.3-fold (5,804-14,968 phosphopeptides and 1,441-6,474 phosphosites. In summary, this study successfully developed a streamlined and rapid and robust strategy for minimalistic phosphoproteomics analysis. Compared with recently published best identification of 600 phosphopeptides (1011 phosphosites) from 1µg lysate. Our results showed superior identification results. This pipeline can be applied to clinical or rare samples for phosphoproteomic profiling, which may provide a better understanding on the correlation of the aberrant phosphorylation and disease’s underlying mechanism.
關鍵字(中) ★ 磷酸化蛋白質體學
★ 二氧化鈦
★ 簡易方法
關鍵字(英)
論文目次 摘要 i
Abstract iii
Table of Contents ⅴ
List of Figures vii
List of Table ⅸ
Chapter 1 Introduction 1
1.1 The phosphorylation of Proteins 1
1.2 MS-based Phosphoproteomics 2
1.3 Enrichment Strategies for Phosphoproteome Analysis - Affinity-Based Chromatography 3
1.4 The Dilemma and Breakthrough in Phosphoproteomics Analysis 5
1.5 Current Status of Microscale Phosphoproteomics Analysis 6
1.6 Thesis Objective 8
Chapter 2 Materials and Method 10
2.1 Cell Culture 10
2.2 Cell Lysis 10
2.3 Protein Precipitation 10
2.4 BCA Protein Assay 11
2.5 Reduction, Alkylation and In-solution Digestion 11
2.6 Desalting by SDB-XC Stage-Tip 12
2.7 Titanium dioxide (TiO2) Phosphopeptide Enrichment 12
2.8 Liquid Chromatography - Tandem Mass Spectrometry (LC-MS/MS) Analysis 13
2.8.1 AB SCIEX TripleTOF 5600 13
2.8.2 Orbitrap Fusion Lumos 13
2.9 Database Search 14
2.10 Bioinformatics Analysis 15
Chapter 3 Results and Discussion 16
3.1 Protein Extraction and Tryptic Peptide Purification in Minimalistic Sample Preparation 16
3.1.1 Evaluating the Compatibility and Efficiency of Lysis Buffer in Enzymatic Digestion and Mass Spectrometry Detection 16
3.1.2 Enrichment Efficiency Dependent on Solution pH Condition 17
3.1.3 Improvement of Enriched Specificity by Lactic Acid-Assisted Sample Loading 18
3.1.4 Comparison of the Sample Acidification Using 0.5% TFA and 50% FA in TiO2 Enrichment 19
3.2 Further Optimization of TiO2 Enrichment in Minimalistic Phosphoproteomics 20
3.2.1 Reducing the Usage of Beads Amount Improving Micro-Scale Enrichment 20
3.2.2 Replacement of Experimental Container from Tip to Tube Improves Identification Coverage 21
3.2.3 Bypassing Post-Enrichment Desalting by Changing Elution Buffer 21
3.3 Digestion Time and Enzyme Ratio for Microscale Sample 22
3.4 Streamlined Workflow in Minimalistic Phosphoproteomics 23
3.4.1 Sensitivity and Reproducibility of SR-mPhos Platform 23
3.4.2 Pathway Mapping of Phosphoproteome in Non-Small Cell Lung Cancer 25
3.5 Minimalistic Phosphoproteomics Analysis Using DIA Approach 27
3.5.1 Improvement of Phosphoproteome Coverage Using DIA Approaches 28
3.5.2 Quantitation f by LibDIA and Direct DIA 29
Chapter 4 Conclusion 31
Reference 64
參考文獻 1. Humphrey, S.J., et al., “High-throughput and high-sensitivity phosphoproteomics with the EasyPhos platform.” Nat Protoc. 13(9), Sep 2018, p. 1897-1916.
2. Duan, G. and D. Walther, “The roles of post-translational modifications in the context of protein interaction networks.” PLoS Comput Biol. 11(2), Feb 2015, p. e1004049.
3. Cohen, P., “The origins of protein phosphorylation.” Nature Cell Biology. 4(5), 2002/05/01 2002, p. E127-E130.
4. Adam, K. and T. Hunter, “Histidine kinases and the missing phosphoproteome from prokaryotes to eukaryotes.” Lab Invest. 98(2), Feb 2018, p. 233-247.
5. Rikova, K., et al., “Global Survey of Phosphotyrosine Signaling Identifies Oncogenic Kinases in Lung Cancer.” Cell. 131(6), 2007/12/14/ 2007, p. 1190-1203.
6. Blume-Jensen, P. and T. Hunter, “Oncogenic kinase signalling.” Nature. 411(6835), 2001/05/01 2001, p. 355-365.
7. Zanivan, S., et al., “In Vivo SILAC-Based Proteomics Reveals Phosphoproteome Changes during Mouse Skin Carcinogenesis.” Cell Reports. 3(2), 2013/02/21/ 2013, p. 552-566.
8. Grundke-Iqbal, I., et al., “Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology.” Proceedings of the National Academy of Sciences of the United States of America. 83(13), 1986, p. 4913-4917.
9. Eidenmüller, J., et al., “Phosphorylation-mimicking glutamate clusters in the proline-rich region are sufficient to simulate the functional deficiencies of hyperphosphorylated tau protein.” The Biochemical journal. 357(Pt 3), 2001, p. 759-767.
10. Meyerovitch, J., J.M. Backer, and C.R. Kahn, “Hepatic phosphotyrosine phosphatase activity and its alterations in diabetic rats.” The Journal of clinical investigation. 84(3), 1989, p. 976-983.
11. Danielsson, A., et al., “Attenuation of Insulin-stimulated Insulin Receptor Substrate-1 Serine 307 Phosphorylation in Insulin Resistance of Type 2 Diabetes*.” Journal of Biological Chemistry. 280(41), 2005/10/14/ 2005, p. 34389-34392.
12. Giansanti, P., et al., “Six alternative proteases for mass spectrometry–based proteomics beyond trypsin.” Nature Protocols. 11(5), 2016/05/01 2016, p. 993-1006.
13. Giorgianni, F. and S. Beranova-Giorgianni, “Phosphoproteome Discovery in Human Biological Fluids.” Proteomes. 4(4), 2016, p. 37.
14. Engholm-Keller, K. and M.R. Larsen, “Technologies and challenges in large-scale phosphoproteomics.” PROTEOMICS. 13(6), 2013/03/01 2013, p. 910-931.
15. Yang, C., X. Zhong, and L. Li, “Recent advances in enrichment and separation strategies for mass spectrometry-based phosphoproteomics.” Electrophoresis. 35(24), 2014, p. 3418-3429.
16. von Stechow, L., C. Francavilla, and J.V. Olsen, “Recent findings and technological advances in phosphoproteomics for cells and tissues.” Expert review of proteomics. 12(5), 2015, p. 469-487.
17. Low, T.Y., et al., “WIDENING THE BOTTLENECK OF PHOSPHOPROTEOMICS: EVOLVING STRATEGIES FOR PHOSPHOPEPTIDE ENRICHMENT.” Mass Spectrometry Reviews. n/a(n/a), 2020/06/03 2020, p.
18. Oda, Y., T. Nagasu, and B.T. Chait, “Enrichment analysis of phosphorylated proteins as a tool for probing the phosphoproteome.” Nature Biotechnology. 19(4), 2001/04/01 2001, p. 379-382.
19. Jensen, S.S. and M.R. Larsen, “Evaluation of the impact of some experimental procedures on different phosphopeptide enrichment techniques.” Rapid Communications in Mass Spectrometry. 21(22), 2007/11/30 2007, p. 3635-3645.
20. Larsen, M.R., et al., “Highly Selective Enrichment of Phosphorylated Peptides from Peptide Mixtures Using Titanium Dioxide Microcolumns *.” Molecular & Cellular Proteomics. 4(7), 2005, p. 873-886.
21. Sugiyama, N., et al., “Phosphopeptide Enrichment by Aliphatic Hydroxy Acid-modified Metal Oxide Chromatography for Nano-LC-MS/MS in Proteomics Applications * S.” Molecular & Cellular Proteomics. 6(6), 2007, p. 1103-1109.
22. Larsen, M.R., et al., “Highly selective enrichment of phosphorylated peptides from peptide mixtures using titanium dioxide microcolumns.” Mol Cell Proteomics. 4(7), Jul 2005, p. 873-86.
23. Cantin, G.T., et al., “Optimizing TiO2-Based Phosphopeptide Enrichment for Automated Multidimensional Liquid Chromatography Coupled to Tandem Mass Spectrometry.” Analytical Chemistry. 79(12), 2007/06/01 2007, p. 4666-4673.
24. Kuroda, I., et al., “Phosphopeptide-selective column-switching RP-HPLC with a titania precolumn.” Anal Sci. 20(9), Sep 2004, p. 1313-9.
25. Pinkse, M.W., et al., “Selective isolation at the femtomole level of phosphopeptides from proteolytic digests using 2D-NanoLC-ESI-MS/MS and titanium oxide precolumns.” Anal Chem. 76(14), Jul 15 2004, p. 3935-43.
26. Kyono, Y., et al., “Successive and Selective Release of Phosphorylated Peptides Captured by Hydroxy Acid-Modified Metal Oxide Chromatography.” Journal of Proteome Research. 7(10), 2008/10/03 2008, p. 4585-4593.
27. Simon, E.S., et al., “Improved enrichment strategies for phosphorylated peptides on titanium dioxide using methyl esterification and pH gradient elution.” Anal Biochem. 377(2), Jun 15 2008, p. 234-42.
28. Rappsilber, J., M. Mann, and Y. Ishihama, “Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips.” Nat Protoc. 2(8), 2007, p. 1896-906.
29. Imami, K., et al., “Automated Phosphoproteome Analysis for Cultured Cancer Cells by Two-Dimensional NanoLC-MS Using a Calcined Titania/C18 Biphasic Column.” Analytical Sciences. 24(1), 2008, p. 161-166.
30. Mohammed, S., et al., “Chip-Based Enrichment and NanoLC-MS/MS Analysis of Phosphopeptides from Whole Lysates.” J Proteome Res. 7(4), Apr 2008, p. 1565-71.
31. Tsai, C.-F., et al., “Immobilized Metal Affinity Chromatography Revisited: pH/Acid Control toward High Selectivity in Phosphoproteomics.” Journal of Proteome Research. 7(9), 2008/09/05 2008, p. 4058-4069.
32. Rush, J., et al., “Immunoaffinity profiling of tyrosine phosphorylation in cancer cells.” Nat Biotechnol. 23(1), Jan 2005, p. 94-101.
33. Bodenmiller, B., et al., “Reproducible isolation of distinct, overlapping segments of the phosphoproteome.” Nat Methods. 4(3), Mar 2007, p. 231-7.
34. Dickhut, C., et al., “Impact of digestion conditions on phosphoproteomics.” J Proteome Res. 13(6), Jun 6 2014, p. 2761-70.
35. Kim, J., D.G. Camp Ii, and R.D. Smith, “Improved detection of multi-phosphorylated peptides in the presence of phosphoric acid in liquid chromatography/mass spectrometry.” Journal of Mass Spectrometry. 39(2), 2004/02/01 2004, p. 208-215.
36. Nakamura, T., K.T. Myint, and Y. Oda, “Ethylenediaminetetraacetic acid increases identification rate of phosphoproteomics in real biological samples.” Journal of proteome research. 9(3), 2010/03// 2010, p. 1385-1391.
37. Winter, D., et al., “Citrate Boosts the Performance of Phosphopeptide Analysis by UPLC-ESI-MS/MS.” Journal of Proteome Research. 8(1), 2009/01/02 2009, p. 418-424.
38. Choi, H., H.-s. Lee, and Z.-Y. Park, “Detection of Multiphosphorylated Peptides in LC−MS/MS Analysis under Low pH Conditions.” Analytical Chemistry. 80(8), 2008/04/15 2008, p. 3007-3015.
39. Beausoleil, S.A., et al., “A probability-based approach for high-throughput protein phosphorylation analysis and site localization.” Nat Biotechnol. 24(10), Oct 2006, p. 1285-92.
40. Boersema, P.J., S. Mohammed, and A.J. Heck, “Phosphopeptide fragmentation and analysis by mass spectrometry.” J Mass Spectrom. 44(6), Jun 2009, p. 861-78.
41. Palumbo, A.M. and G.E. Reid, “Evaluation of gas-phase rearrangement and competing fragmentation reactions on protein phosphorylation site assignment using collision induced dissociation-MS/MS and MS3.” Anal Chem. 80(24), Dec 15 2008, p. 9735-47.
42. Thingholm, T.E., et al., “SIMAC (sequential elution from IMAC), a phosphoproteomics strategy for the rapid separation of monophosphorylated from multiply phosphorylated peptides.” Mol Cell Proteomics. 7(4), Apr 2008, p. 661-71.
43. Gessulat, S., et al., “Prosit: proteome-wide prediction of peptide tandem mass spectra by deep learning.” Nat Methods. 16(6), Jun 2019, p. 509-518.
44. Bekker-Jensen, D.B., et al., “Rapid and site-specific deep phosphoproteome profiling by data-independent acquisition without the need for spectral libraries.” Nature Communications. 11(1), 2020/02/07 2020, p. 787.
45. Kitata, R.B., et al., “A data-independent acquisition-based global phosphoproteomics system enables deep profiling.” Nature Communications. 12(1), 2021/05/05 2021, p. 2539.
46. Masuda, T., et al., “Microscale phosphoproteome analysis of 10,000 cells from human cancer cell lines.” Anal Chem. 83(20), Oct 15 2011, p. 7698-703.
47. Chen, W., L. Chen, and R. Tian, “An integrated strategy for highly sensitive phosphoproteome analysis from low micrograms of protein samples.” Analyst. 143(15), Jul 23 2018, p. 3693-3701.
48. Tsougeni, K., et al., “TiO2–ZrO2 affinity chromatography polymeric microchip for phosphopeptide enrichment and separation.” Lab on a Chip. 11(18), 2011, p. 3113-3120.
49. Zhu, Y., et al., “Nanodroplet processing platform for deep and quantitative proteome profiling of 10–100 mammalian cells.” Nature Communications. 9(1), 2018/02/28 2018, p. 882.
50. Williams, S.M., et al., “Automated Coupling of Nanodroplet Sample Preparation with Liquid Chromatography–Mass Spectrometry for High-Throughput Single-Cell Proteomics.” Analytical Chemistry. 92(15), 2020/08/04 2020, p. 10588-10596.
51. Yu, Y.-Q., et al., “Enzyme-Friendly, Mass Spectrometry-Compatible Surfactant for In-Solution Enzymatic Digestion of Proteins.” Analytical Chemistry. 75(21), 2003/11/01 2003, p. 6023-6028.
52. León, I.R., et al., “Quantitative assessment of in-solution digestion efficiency identifies optimal protocols for unbiased protein analysis.” Molecular & cellular proteomics : MCP. 12(10), 2013, p. 2992-3005.
53. Masuda, T., M. Tomita, and Y. Ishihama, “Phase Transfer Surfactant-Aided Trypsin Digestion for Membrane Proteome Analysis.” Journal of Proteome Research. 7(2), 2008/02/01 2008, p. 731-740.
54. Connor, P.A. and A.J. McQuillan, “Phosphate Adsorption onto TiO2 from Aqueous Solutions:  An in Situ Internal Reflection Infrared Spectroscopic Study.” Langmuir. 15(8), 1999/04/01 1999, p. 2916-2921.
55. Li, Q.R., et al., “Effect of peptide-to-TiO2 beads ratio on phosphopeptide enrichment selectivity.” J Proteome Res. 8(11), Nov 2009, p. 5375-81.
56. Chen, Z., et al., “Non-small-cell lung cancers: a heterogeneous set of diseases.” Nat Rev Cancer. 14(8), Aug 2014, p. 535-46.
57. Lee, D.H., “Treatments for EGFR-mutant non-small cell lung cancer (NSCLC): The road to a success, paved with failures.” Pharmacol Ther. 174 Jun 2017, p. 1-21.
58. Nyati, M.K., et al., “Integration of EGFR inhibitors with radiochemotherapy.” Nat Rev Cancer. 6(11), Nov 2006, p. 876-85.
59. Ren, S. and B.J. Rollins, “Cyclin C/cdk3 promotes Rb-dependent G0 exit.” Cell. 117(2), Apr 16 2004, p. 239-51.
60. Pluquet, O., et al., “Endoplasmic reticulum stress accelerates p53 degradation by the cooperative actions of Hdm2 and glycogen synthase kinase 3beta.” Mol Cell Biol. 25(21), Nov 2005, p. 9392-405.
61. Bui, N.-L.-C., et al., “Bad phosphorylation as a target of inhibition in oncology.” Cancer Letters. 415 2018/02/28/ 2018, p. 177-186.
62. Casamayor, A., N.A. Morrice, and D.R. Alessi, “Phosphorylation of Ser-241 is essential for the activity of 3-phosphoinositide-dependent protein kinase-1: identification of five sites of phosphorylation in vivo.” Biochem J. 342 ( Pt 2)(Pt 2), Sep 1 1999, p. 287-92.
63. Michalski, A., J. Cox, and M. Mann, “More than 100,000 Detectable Peptide Species Elute in Single Shotgun Proteomics Runs but the Majority is Inaccessible to Data-Dependent LC−MS/MS.” Journal of Proteome Research. 10(4), 2011/04/01 2011, p. 1785-1793.
64. Gillet, L.C., et al., “Targeted Data Extraction of the MS/MS Spectra Generated by Data-independent Acquisition: A New Concept for Consistent and Accurate Proteome Analysis*.” Molecular & Cellular Proteomics. 11(6), 2012/06/01/ 2012, p. O111.016717.
65. Bruderer, R., et al., “High-precision iRT prediction in the targeted analysis of data-independent acquisition and its impact on identification and quantitation.” Proteomics. 16(15-16), Aug 2016, p. 2246-56.
66. Tsou, C.C., et al., “DIA-Umpire: comprehensive computational framework for data-independent acquisition proteomics.” Nat Methods. 12(3), Mar 2015, p. 258-64, 7 p following 264.
67. Keller, A., et al., “Opening a SWATH Window on Posttranslational Modifications: Automated Pursuit of Modified Peptides.” Mol Cell Proteomics. 15(3), Mar 2016, p. 1151-63.
68. Wang, J., et al., “MSPLIT-DIA: sensitive peptide identification for data-independent acquisition.” Nat Methods. 12(12), Dec 2015, p. 1106-8.
69. Li, Y., et al., “Group-DIA: analyzing multiple data-independent acquisition mass spectrometry data files.” Nat Methods. 12(12), Dec 2015, p. 1105-6.
指導教授 陳玉如 謝發坤 審核日期 2021-9-27
推文 facebook   plurk   twitter   funp   google   live   udn   HD   myshare   reddit   netvibes   friend   youpush   delicious   baidu   
網路書籤 Google bookmarks   del.icio.us   hemidemi   myshare   

若有論文相關問題,請聯絡國立中央大學圖書館推廣服務組 TEL:(03)422-7151轉57407,或E-mail聯絡  - 隱私權政策聲明