1
|
Lewis PA, Silajdžić E, Smith H, Bates N, Smith CA, Mancini FE, Knight D, Denning C, Brison DR, Kimber SJ. A secreted proteomic footprint for stem cell pluripotency. PLoS One 2024; 19:e0299365. [PMID: 38875182 PMCID: PMC11178176 DOI: 10.1371/journal.pone.0299365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 02/08/2024] [Indexed: 06/16/2024] Open
Abstract
With a view to developing a much-needed non-invasive method for monitoring the healthy pluripotent state of human stem cells in culture, we undertook proteomic analysis of the waste medium from cultured embryonic (Man-13) and induced (Rebl.PAT) human pluripotent stem cells (hPSCs). Cells were grown in E8 medium to maintain pluripotency, and then transferred to FGF2 and TGFβ deficient E6 media for 48 hours to replicate an early, undirected dissolution of pluripotency. We identified a distinct proteomic footprint associated with early loss of pluripotency in both hPSC lines, and a strong correlation with changes in the transcriptome. We demonstrate that multiplexing of four E8- against four E6- enriched secretome biomarkers provides a robust, diagnostic metric for the pluripotent state. These biomarkers were further confirmed by Western blotting which demonstrated consistent correlation with the pluripotent state across cell lines, and in response to a recovery assay.
Collapse
Affiliation(s)
- Philip A Lewis
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Edina Silajdžić
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Helen Smith
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Nicola Bates
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Christopher A Smith
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Fabrizio E Mancini
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - David Knight
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Chris Denning
- Biodiscovery Institute, Division of Cancer & Stem Cells, School of Medicine, University of Nottingham, University Park, Nottingham, United Kingdom
| | - Daniel R Brison
- Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Susan J Kimber
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
2
|
Grafting of iPS cell-derived tenocytes promotes motor function recovery after Achilles tendon rupture. Nat Commun 2021; 12:5012. [PMID: 34408142 PMCID: PMC8373964 DOI: 10.1038/s41467-021-25328-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 08/04/2021] [Indexed: 11/09/2022] Open
Abstract
Tendon self-renewal is a rare occurrence because of the poor vascularization of this tissue; therefore, reconstructive surgery using autologous tendon is often performed in severe injury cases. However, the post-surgery re-injury rate is relatively high, and the collection of autologous tendons leads to muscle weakness, resulting in prolonged rehabilitation. Here, we introduce an induced pluripotent stem cell (iPSC)-based technology to develop a therapeutic option for tendon injury. First, we derived tenocytes from human iPSCs by recapitulating the normal progression of step-wise narrowing fate decisions in vertebrate embryos. We used single-cell RNA sequencing to analyze the developmental trajectory of iPSC-derived tenocytes. We demonstrated that iPSC-tenocyte grafting contributed to motor function recovery after Achilles tendon injury in rats via engraftment and paracrine effects. The biomechanical strength of regenerated tendons was comparable to that of healthy tendons. We suggest that iPSC-tenocytes will provide a therapeutic option for tendon injury.
Collapse
|
3
|
Wang X, Zhu J, Yang C, Qin F, Zhang B. Segmented Microfluidics-Based Packing Technology for Chromatographic Columns. Anal Chem 2021; 93:8450-8458. [PMID: 34111926 DOI: 10.1021/acs.analchem.1c00545] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanoflow liquid chromatography-mass spectrometry (NanoLC-MS) has become the method of choice for the analysis of complex biological systems, especially when the available sample amount is limited. The preparation of high-performance capillary columns for nanoLC use is still a technical challenge. Here, we report a segmented microfluidic method for the preparation of packed capillary columns, where liquid segments were used as soft, dynamic, and well-dispersed slurry reservoirs for carrying and delivering micrometer packing particles. Based on this microfluidic packing technology, the column bed was assembled layer-by-layer at a 50 μm resolution, and ultralong capillary columns of 3, 5, and 10 m were fabricated in such a manner. The microfluidically packed columns demonstrated excellent separation efficiencies of 116 000 plates/m. The higher efficiencies obtained at higher slurry concentrations also indicate that a high-quality packed bed can be obtained without sacrificing the packing speed. Kinetic performance limit analysis shows that the microfluidic packed columns have higher peak capacity production efficiency in the high-resolution region, presenting an improved separation impedance of 2800, which is significantly better than columns packed with the conventional slurry packing method. In comparison with a commercial nanoLC column, a 5 m long microfluidic packed column was evaluated for proteomic analysis using a standard HeLa protein digest and presented 261% improvement in peptide identification capability, resulting in significantly enhanced protein identification confidence.
Collapse
Affiliation(s)
- Xiaofei Wang
- Department of Chemistry and The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Jue Zhu
- Department of Chemistry and The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Chenyuhu Yang
- Department of Chemistry and The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Fei Qin
- Xiamen Medical College, Xiamen 361023, China
| | - Bo Zhang
- Department of Chemistry and The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
4
|
Glycoproteomic analysis of the changes in protein N-glycosylation during neuronal differentiation in human-induced pluripotent stem cells and derived neuronal cells. Sci Rep 2021; 11:11169. [PMID: 34045517 PMCID: PMC8160270 DOI: 10.1038/s41598-021-90102-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/04/2021] [Indexed: 11/09/2022] Open
Abstract
N-glycosylation of glycoproteins, a major post-translational modification, plays a crucial role in various biological phenomena. In central nervous systems, N-glycosylation is thought to be associated with differentiation and regeneration; however, the state and role of N-glycosylation in neuronal differentiation remain unclear. Here, we conducted sequential LC/MS/MS analyses of tryptic digest, enriched glycopeptides, and deglycosylated peptides of proteins derived from human-induced pluripotent stem cells (iPSCs) and iPSC-derived neuronal cells, which were used as a model of neuronal differentiation. We demonstrate that the production profiles of many glycoproteins and their glycoforms were altered during neuronal differentiation. Particularly, the levels of glycoproteins modified with an N-glycan, consisting of five N-acetylhexosamines, three hexoses, and a fucose (HN5H3F), increased in dopaminergic neuron-rich cells (DAs). The N-glycan was deduced to be a fucosylated and bisected biantennary glycan based on product ion spectra. Interestingly, the HN5H3F-modified proteins were predicted to be functionally involved in neural cell adhesion, axon guidance, and the semaphorin-plexin signaling pathway, and protein modifications were site-selective and DA-selective regardless of protein production levels. Our integrated method for glycoproteome analysis and resultant profiles of glycoproteins and their glycoforms provide valuable information for further understanding the role of N-glycosylation in neuronal differentiation and neural regeneration.
Collapse
|
5
|
Marissen R, Palmblad M. mzRecal: universal MS1 recalibration in mzML using identified peptides in mzIdentML as internal calibrants. Bioinformatics 2021; 37:2768-2769. [PMID: 33538780 DOI: 10.1093/bioinformatics/btab056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/31/2020] [Accepted: 01/26/2021] [Indexed: 11/13/2022] Open
Abstract
SUMMARY In mass spectrometry-based proteomics, accurate peptide masses improve identifications, alignment and quantitation. Getting the most out of any instrument therefore requires proper calibration. Here we present a new stand-alone software, mzRecal, for universal automatic recalibration of data from all common mass analyzers using standard open formats and based on physical principles. AVAILABILITY AND IMPLEMENTATION mzRecal is implemented in Go and freely available on https://github.com/524D/mzRecal. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Rob Marissen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, RC Leiden, The Netherlands
| | - Magnus Palmblad
- Center for Proteomics and Metabolomics, Leiden University Medical Center, RC Leiden, The Netherlands
| |
Collapse
|
6
|
Kimura A, Toyoda T, Iwasaki M, Hirama R, Osafune K. Combined Omics Approaches Reveal the Roles of Non-canonical WNT7B Signaling and YY1 in the Proliferation of Human Pancreatic Progenitor Cells. Cell Chem Biol 2020; 27:1561-1572.e7. [PMID: 33125912 DOI: 10.1016/j.chembiol.2020.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023]
Abstract
The proliferation of human pancreatic progenitor cells (PPCs) is critical for developing cell therapies for diabetes. Here, using transcriptome analysis combined with small interfering RNA (siRNA) screening, we revealed that WNT7B is a downstream growth factor of AT7867, a compound known to promote the proliferation of PPCs generated from human pluripotent stem cells. Feeder cell lines stably expressing mouse Wnt7a or Wnt7b, but not other Wnts, enhanced PPC proliferation in the absence of AT7867. Importantly, Wnt7a/b ligands did not activate the canonical Wnt pathway, and PPC proliferation depended on the non-canonical Wnt/PKC pathway. A comparison of the phosphoproteome in response to AT7867 or a newly synthesized AT7867 derivative uncovered the function of YY1 as a transcriptional regulator of WNT7B. Overall, our data highlight unknown roles of non-canonical WNT7B/PKC signaling and YY1 in human PPC proliferation and will contribute to the stable supply of a cell source for pancreatic disease modeling and therapeutic applications.
Collapse
Affiliation(s)
- Azuma Kimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Taro Toyoda
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Mio Iwasaki
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Ryusuke Hirama
- Research Institute for Bioscience Products and Fine Chemicals, Ajinomoto Co., Inc., Kawasaki, Kanagawa 210-8681, Japan
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
7
|
Affiliation(s)
- Yasushi ISHIHAMA
- Department of Molecular & Cellular BioAnalysis, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
8
|
Lindoso RS, Kasai-Brunswick TH, Monnerat Cahli G, Collino F, Bastos Carvalho A, Campos de Carvalho AC, Vieyra A. Proteomics in the World of Induced Pluripotent Stem Cells. Cells 2019; 8:cells8070703. [PMID: 31336746 PMCID: PMC6678893 DOI: 10.3390/cells8070703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 02/05/2023] Open
Abstract
Omics approaches have significantly impacted knowledge about molecular signaling pathways driving cell function. Induced pluripotent stem cells (iPSC) have revolutionized the field of biological sciences and proteomics and, in particular, has been instrumental in identifying key elements operating during the maintenance of the pluripotent state and the differentiation process to the diverse cell types that form organisms. This review covers the evolution of conceptual and methodological strategies in proteomics; briefly describes the generation of iPSC from a historical perspective, the state-of-the-art of iPSC-based proteomics; and compares data on the proteome and transcriptome of iPSC to that of embryonic stem cells (ESC). Finally, proteomics of healthy and diseased cells and organoids differentiated from iPSC are analyzed.
Collapse
Affiliation(s)
- Rafael Soares Lindoso
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
| | - Tais H Kasai-Brunswick
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
| | - Gustavo Monnerat Cahli
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
- Laboratory of Proteomics, LADETEC, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-598, Brazil
| | - Federica Collino
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
- Department of Biomedical Sciences, University of Padova, 35131 Padua, Italy
| | - Adriana Bastos Carvalho
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil
| | - Antonio Carlos Campos de Carvalho
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil.
| | - Adalberto Vieyra
- Carlos Chagas Filho Institute of Biophysics and National Center for Structural Biology and Bioimaging/CENABIO, Federal University of Rio de Janeiro, Rio de Janeiro 21941-102, Brazil.
- Graduate Program in Translational Biomedicine, Grande Rio University, Duque de Caxias 25071-202, Brazil.
| |
Collapse
|
9
|
Towards a high peak capacity of 130 using nanoflow hydrophilic interaction liquid chromatography. Anal Chim Acta 2019; 1062:147-155. [DOI: 10.1016/j.aca.2019.01.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 11/21/2022]
|
10
|
Liang Y, Zhang L, Zhang Y. Well-Defined Materials for High-Performance Chromatographic Separation. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2019; 12:451-473. [PMID: 30939031 DOI: 10.1146/annurev-anchem-061318-114854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Chromatographic separation has been widely applied in various fields, such as chemical engineering, precision medicine, energy, and biology. Because chromatographic separation is based on differential partitioning between the mobile phase and stationary phase and affected by band dispersion and mass transfer resistance from these two phases, the materials used as the stationary phase play a decisive role in separation performance. In this review, we discuss the design of separation materials to achieve the separation with high efficiency and high resolution and highlight the well-defined materials with uniform pore structure and unique properties. The achievements, recent developments, challenges, and future trends of such materials are discussed. Furthermore, the surface functionalization of separation ma-terials for further improvement of separation performance is reviewed. Finally, future research directions and the challenges of chromatographic separation are presented.
Collapse
Affiliation(s)
- Yu Liang
- CAS Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic Research and Analysis Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
| | - Lihua Zhang
- CAS Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic Research and Analysis Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
| | - Yukui Zhang
- CAS Key Lab of Separation Sciences for Analytical Chemistry, National Chromatographic Research and Analysis Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
| |
Collapse
|
11
|
Iwasaki M, Tabata T, Kawahara Y, Ishihama Y, Nakagawa M. Removal of Interference MS/MS Spectra for Accurate Quantification in Isobaric Tag-Based Proteomics. J Proteome Res 2019; 18:2535-2544. [PMID: 31039306 DOI: 10.1021/acs.jproteome.9b00078] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rapid progress in mass spectrometry (MS) has made comprehensive analyses of the proteome possible, but accurate quantification remains challenging. Isobaric tags for relative and absolute quantification (iTRAQ) is widely used as a tool to quantify proteins expressed in different cell types and various cellular conditions. The quantification precision of iTRAQ is quite high, but the accuracy dramatically decreases in the presence of interference peptides that are coeluted and coisolated with the target peptide. Here, we developed "removal of interference mixture MS/MS spectra (RiMS)" to improve the quantification accuracy of isobaric tag approaches. The presence of spectrum interference is judged by examining the overlap in the elution time of all scanned precursor ions. Removal of this interference decreased protein identification (11% loss) but improved quantification accuracy. Further, RiMS does not require any specialized equipment, such as MS3 instruments or an additional ion separation mode. Finally, we demonstrated that RiMS can be used to quantitatively compare human-induced pluripotent stem cells and human dermal fibroblasts, as it revealed differential protein expressions that reflect the biological characteristics of the cells.
Collapse
Affiliation(s)
- Mio Iwasaki
- Center for iPS Cell Research and Application , Kyoto University , Kyoto 606-8507 , Japan
| | - Tsuyoshi Tabata
- Center for iPS Cell Research and Application , Kyoto University , Kyoto 606-8507 , Japan.,Graduate school of Pharmaceutical Sciences , Kyoto University , Kyoto 606-8501 , Japan
| | - Yuka Kawahara
- Center for iPS Cell Research and Application , Kyoto University , Kyoto 606-8507 , Japan
| | - Yasushi Ishihama
- Graduate school of Pharmaceutical Sciences , Kyoto University , Kyoto 606-8501 , Japan
| | - Masato Nakagawa
- Center for iPS Cell Research and Application , Kyoto University , Kyoto 606-8507 , Japan
| |
Collapse
|
12
|
Krieger JR, Wybenga-Groot LE, Tong J, Bache N, Tsao MS, Moran MF. Evosep One Enables Robust Deep Proteome Coverage Using Tandem Mass Tags while Significantly Reducing Instrument Time. J Proteome Res 2019; 18:2346-2353. [DOI: 10.1021/acs.jproteome.9b00082] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
| | | | | | | | - Ming S. Tsao
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, Canada M5G 2C1
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King’s College Circle, Toronto, ON, Canada M5S 1A8
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, Canada M5G 1L7
| | - Michael F. Moran
- Department of Molecular Genetics, University of Toronto, 1 King’s College Circle, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
13
|
Baud A, Little D, Wen TQ, Heywood WE, Gissen P, Mills K. An Optimized Method for the Proteomic Analysis of Low Volumes of Cell Culture Media and the Secretome: The Application and the Demonstration of Altered Protein Expression in iPSC-Derived Neuronal Cell Lines from Parkinson's Disease Patients. J Proteome Res 2019; 18:1198-1207. [PMID: 30562036 DOI: 10.1021/acs.jproteome.8b00831] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Traditionally, cell culture medium in iPSC-derived cell work is not the main focus of the research and often is considered as just "food for cells". We demonstrate that by manipulation of the media and optimized methodology, it is possible to use this solution to study the proteins that the cell secretes (the "secretome"). This is particularly useful in the study of iPSC-derived neurons, which require long culture time. We demonstrate that media can be used to model diseases with optimized incubation and sampling times. The ability not to sacrifice cells allows significant cost and research benefits. In this manuscript we describe an optimized method for the analysis of the cell media from iPSC-derived neuronal lines from control and Parkinson's disease patients. We have evaluated the use of standard and supplement B27-free cell media as well as five different sample preparation techniques for proteomic analysis of the cell secretome. Mass spectral analysis of culture media allowed for the identification of >500 proteins, in 500 μL of media, which is less volume than reported previously (20-40 mL). Using shorter incubation times and our optimized methodology, we describe the use of this technique to study and describe potential disease mechanisms in Parkinson's disease.
Collapse
Affiliation(s)
- Anna Baud
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| | - Daniel Little
- MRC Laboratory for Molecular Cell Biology , University College London , London , WC1E 6BT , U.K
| | - Teo Qi Wen
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| | - Wendy E Heywood
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| | - Paul Gissen
- MRC Laboratory for Molecular Cell Biology , University College London , London , WC1E 6BT , U.K
| | - Kevin Mills
- Centre for Translational Omics , UCL Great Ormond Street Institute of Child Health , London , WC1N 1EH , U.K
| |
Collapse
|
14
|
Wilson RS, Nairn AC. Cell-Type-Specific Proteomics: A Neuroscience Perspective. Proteomes 2018; 6:51. [PMID: 30544872 PMCID: PMC6313874 DOI: 10.3390/proteomes6040051] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/18/2022] Open
Abstract
Cell-type-specific analysis has become a major focus for many investigators in the field of neuroscience, particularly because of the large number of different cell populations found in brain tissue that play roles in a variety of developmental and behavioral disorders. However, isolation of these specific cell types can be challenging due to their nonuniformity and complex projections to different brain regions. Moreover, many analytical techniques used for protein detection and quantitation remain insensitive to the low amounts of protein extracted from specific cell populations. Despite these challenges, methods to improve proteomic yield and increase resolution continue to develop at a rapid rate. In this review, we highlight the importance of cell-type-specific proteomics in neuroscience and the technical difficulties associated. Furthermore, current progress and technological advancements in cell-type-specific proteomics research are discussed with an emphasis in neuroscience.
Collapse
Affiliation(s)
- Rashaun S Wilson
- Yale/NIDA Neuroproteomics Center, 300 George St., New Haven, CT 06511, USA.
| | - Angus C Nairn
- Yale/NIDA Neuroproteomics Center, 300 George St., New Haven, CT 06511, USA.
- Department of Psychiatry, Yale School of Medicine, Connecticut Mental Health Center, New Haven, CT 06511, USA.
| |
Collapse
|
15
|
Dams M, Dores-Sousa JL, Lamers RJ, Treumann A, Eeltink S. High-Resolution Nano-Liquid Chromatography with Tandem Mass Spectrometric Detection for the Bottom-Up Analysis of Complex Proteomic Samples. Chromatographia 2018. [DOI: 10.1007/s10337-018-3647-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
16
|
Ito Y, Nakamura S, Sugimoto N, Shigemori T, Kato Y, Ohno M, Sakuma S, Ito K, Kumon H, Hirose H, Okamoto H, Nogawa M, Iwasaki M, Kihara S, Fujio K, Matsumoto T, Higashi N, Hashimoto K, Sawaguchi A, Harimoto KI, Nakagawa M, Yamamoto T, Handa M, Watanabe N, Nishi E, Arai F, Nishimura S, Eto K. Turbulence Activates Platelet Biogenesis to Enable Clinical Scale Ex Vivo Production. Cell 2018; 174:636-648.e18. [PMID: 30017246 DOI: 10.1016/j.cell.2018.06.011] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 03/30/2018] [Accepted: 05/23/2018] [Indexed: 12/14/2022]
Abstract
The ex vivo generation of platelets from human-induced pluripotent cells (hiPSCs) is expected to compensate donor-dependent transfusion systems. However, manufacturing the clinically required number of platelets remains unachieved due to the low platelet release from hiPSC-derived megakaryocytes (hiPSC-MKs). Here, we report turbulence as a physical regulator in thrombopoiesis in vivo and its application to turbulence-controllable bioreactors. The identification of turbulent energy as a determinant parameter allowed scale-up to 8 L for the generation of 100 billion-order platelets from hiPSC-MKs, which satisfies clinical requirements. Turbulent flow promoted the release from megakaryocytes of IGFBP2, MIF, and Nardilysin to facilitate platelet shedding. hiPSC-platelets showed properties of bona fide human platelets, including circulation and hemostasis capacities upon transfusion in two animal models. This study provides a concept in which a coordinated physico-chemical mechanism promotes platelet biogenesis and an innovative strategy for ex vivo platelet manufacturing.
Collapse
Affiliation(s)
- Yukitaka Ito
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Kyoto Development Center, Megakaryon Corporation, Kyoto, Japan
| | - Sou Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | | | - Yoshikazu Kato
- Mixing Technology Laboratory, SATAKE Chemical Equipment Manufacturing Ltd., Saitama, Japan
| | - Mikiko Ohno
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan
| | - Shinya Sakuma
- Department of Micro-Nano Systems Engineering, Nagoya University, Nagoya, Japan
| | - Keitaro Ito
- Department of Micro-Nano Systems Engineering, Nagoya University, Nagoya, Japan
| | - Hiroki Kumon
- Department of Micro-Nano Systems Engineering, Nagoya University, Nagoya, Japan
| | - Hidenori Hirose
- Kyoto Development Center, Megakaryon Corporation, Kyoto, Japan
| | - Haruki Okamoto
- Kyoto Development Center, Megakaryon Corporation, Kyoto, Japan
| | - Masayuki Nogawa
- Center for Transfusion Medicine and Cell Therapy, Keio University School of Medicine, Tokyo, Japan
| | - Mio Iwasaki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Shunsuke Kihara
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kosuke Fujio
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Takuya Matsumoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Natsumi Higashi
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kazuya Hashimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Akira Sawaguchi
- Department of Anatomy, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Ken-Ichi Harimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masato Nakagawa
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; AMED-CREST, AMED, Tokyo, Japan
| | - Makoto Handa
- Center for Transfusion Medicine and Cell Therapy, Keio University School of Medicine, Tokyo, Japan
| | - Naohide Watanabe
- Center for Transfusion Medicine and Cell Therapy, Keio University School of Medicine, Tokyo, Japan
| | - Eiichiro Nishi
- Department of Pharmacology, Shiga University of Medical Science, Otsu, Japan
| | - Fumihito Arai
- Department of Micro-Nano Systems Engineering, Nagoya University, Nagoya, Japan
| | - Satoshi Nishimura
- Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan; Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
17
|
Nakashima Y, Miyagi-Shiohira C, Noguchi H, Omasa T. Atorvastatin Inhibits the HIF1α-PPAR Axis, Which Is Essential for Maintaining the Function of Human Induced Pluripotent Stem Cells. Mol Ther 2018; 26:1715-1734. [PMID: 29929789 PMCID: PMC6036234 DOI: 10.1016/j.ymthe.2018.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 12/31/2022] Open
Abstract
We herein report a novel mechanism of action of statin preparations using a new drug discovery method. Milk fat globule-EGF factor 8 protein (MFG-E8) was identified from the secretory component of mouse embryonic fibroblast (MEF) as a cell adhesion-promoting factor effective for screening active cellular agents of human induced pluripotent stem cells (hiPSCs) in vitro using electrochemical impedance. Our analyses showed that atorvastatin did not cause death in myocardial cells differentiated from hiPSCs but reduced the pluripotent cell survival in vitro when using serum- and albumin-free media, and inhibited the ability to form teratomas in mice. This result could have been already the cytopathic effect of atorvastatin, and complete elimination of hiPSCs was confirmed in the xenotransplantation assay. The administration of atorvastatin to hiPSCs caused the expression of hypoxia inducible factor (HIF)1α mRNA to be unchanged at 6 hr and downregulated at 24 hr. In addition, the inhibition of the survival of hiPSCs was confirmed by HIF1α-peroxisome proliferator-activated receptor (PPAR) axis inhibition. These results suggest that the addition of atorvastatin to hiPSC cultures reduces the survival of pluripotent cells by suppressing the HIF1α-PPAR axis. In summary, the HIF1α-PPAR axis has an important role in maintaining the survival of pluripotent hiPSCs.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8506, Japan; Department of Material and Life Science, Graduate School of Engineering, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara-cho, Okinawa 903-0215, Japan.
| | - Chika Miyagi-Shiohira
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara-cho, Okinawa 903-0215, Japan
| | - Hirofumi Noguchi
- Department of Regenerative Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara-cho, Okinawa 903-0215, Japan
| | - Takeshi Omasa
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8506, Japan; Department of Material and Life Science, Graduate School of Engineering, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
18
|
Andjelković U, Tufegdžić S, Popović M. Use of monolithic supports for high-throughput protein and peptide separation in proteomics. Electrophoresis 2017; 38:2851-2869. [DOI: 10.1002/elps.201700260] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/02/2017] [Accepted: 09/03/2017] [Indexed: 01/08/2023]
Affiliation(s)
- Uroš Andjelković
- Department of Chemistry-Institute of Chemistry; Technology and Metallurgy; University of Belgrade; Belgrade Serbia
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Srdjan Tufegdžić
- Department of Chemistry-Institute of Chemistry; Technology and Metallurgy; University of Belgrade; Belgrade Serbia
| | - Milica Popović
- Faculty of Chemistry; University of Belgrade; Belgrade Serbia
| |
Collapse
|
19
|
Combining membrane proteomics and computational three-way pathway analysis revealed signalling pathways preferentially regulated in human iPSCs and human ESCs. Sci Rep 2017; 7:15055. [PMID: 29118436 PMCID: PMC5678157 DOI: 10.1038/s41598-017-15347-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022] Open
Abstract
Owing to the clinical potential of human induced pluripotent stem cells (hiPSCs) in regenerative medicine, a thorough examination of the similarities and differences between hiPSCs and human embryonic stem cells (hESCs) has become indispensable. Moreover, as the important roles of membrane proteins in biological signalling, functional analyses of membrane proteome are therefore promising. In this study, a pathway analysis by the bioinformatics tool GSEA was first performed to identify significant pathways associated with the three comparative membrane proteomics experiments: hiPSCs versus precursor human foreskin fibroblasts (HFF), hESCs versus precursor HFF, and hiPSCs versus hESCs. A following three-way pathway comparison was conducted to identify the differentially regulated pathways that may contribute to the differences between hiPSCs and hESCs. Our results revealed that pathways related to oxidative phosphorylation and focal adhesion may undergo incomplete regulations during the reprogramming process. This hypothesis was supported by another public proteomics dataset to a certain degree. The identified pathways and their core enriched proteins could serve as the starting point to explore the possible ways to make hiPSCs closer to hESCs.
Collapse
|
20
|
Aburaya S, Aoki W, Minakuchi H, Ueda M. Definitive screening design enables optimization of LC-ESI-MS/MS parameters in proteomics. Biosci Biotechnol Biochem 2017; 81:2237-2243. [PMID: 29068257 DOI: 10.1080/09168451.2017.1391685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In proteomics, more than 100,000 peptides are generated from the digestion of human cell lysates. Proteome samples have a broad dynamic range in protein abundance; therefore, it is critical to optimize various parameters of LC-ESI-MS/MS to comprehensively identify these peptides. However, there are many parameters for LC-ESI-MS/MS analysis. In this study, we applied definitive screening design to simultaneously optimize 14 parameters in the operation of monolithic capillary LC-ESI-MS/MS to increase the number of identified proteins and/or the average peak area of MS1. The simultaneous optimization enabled the determination of two-factor interactions between LC and MS. Finally, we found two parameter sets of monolithic capillary LC-ESI-MS/MS that increased the number of identified proteins by 8.1% or the average peak area of MS1 by 67%. The definitive screening design would be highly useful for high-throughput analysis of the best parameter set in LC-ESI-MS/MS systems.
Collapse
Affiliation(s)
- Shunsuke Aburaya
- a Division of Applied Life Sciences, Graduate School of Agriculture , Kyoto University , Kyoto , Japan.,b Japan Society for the Promotion of Science , Kyoto , Japan
| | - Wataru Aoki
- a Division of Applied Life Sciences, Graduate School of Agriculture , Kyoto University , Kyoto , Japan.,c Kyoto Integrated Science and Technology Bio-Analysis Center , Kyoto , Japan.,d JST, PRESTO , Kyoto , Japan
| | | | - Mitsuyoshi Ueda
- a Division of Applied Life Sciences, Graduate School of Agriculture , Kyoto University , Kyoto , Japan.,c Kyoto Integrated Science and Technology Bio-Analysis Center , Kyoto , Japan.,d JST, PRESTO , Kyoto , Japan
| |
Collapse
|
21
|
Ikeda T, Uchiyama I, Iwasaki M, Sasaki T, Nakagawa M, Okita K, Masui S. Artificial acceleration of mammalian cell reprogramming by bacterial proteins. Genes Cells 2017; 22:918-928. [PMID: 28776863 DOI: 10.1111/gtc.12519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 07/06/2017] [Indexed: 11/26/2022]
Abstract
The molecular mechanisms of cell reprogramming and differentiation involve various signaling factors. Small molecule compounds have been identified to artificially influence these factors through interacting cellular proteins. Although such small molecule compounds are useful to enhance reprogramming and differentiation and to show the mechanisms that underlie these events, the screening usually requires a large number of compounds to identify only a very small number of hits (e.g., one hit among several tens of thousands of compounds). Here, we show a proof of concept that xenospecific gene products can affect the efficiency of cell reprogramming to pluripotency. Thirty genes specific for the bacterium Wolbachia pipientis were forcibly expressed individually along with reprogramming factors (Oct4, Sox2, Klf4 and c-Myc) that can generate induced pluripotent stem cells in mammalian cells, and eight were found to affect the reprogramming efficiency either positively or negatively (hit rate 26.7%). Mechanistic analysis suggested one of these proteins interacted with cytoskeleton to promote reprogramming. Our results raise the possibility that xenospecific gene products provide an alternative way to study the regulatory mechanism of cell identity.
Collapse
Affiliation(s)
- Takashi Ikeda
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ikuo Uchiyama
- National Institute for Basic Biology, National Institutes of Natural Sciences, Nishigonaka 38, Myodaiji, Okazaki, Aichi, 444-8585, Japan
| | - Mio Iwasaki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tetsuhiko Sasaki
- Honeybee Science Research Center, Research Institute, Tamagawa University, 6-1-1 Tamagawagakuen, Machida, Tokyo, 194-8610, Japan
| | - Masato Nakagawa
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Keisuke Okita
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shinji Masui
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
22
|
Bekker-Jensen DB, Kelstrup CD, Batth TS, Larsen SC, Haldrup C, Bramsen JB, Sørensen KD, Høyer S, Ørntoft TF, Andersen CL, Nielsen ML, Olsen JV. An Optimized Shotgun Strategy for the Rapid Generation of Comprehensive Human Proteomes. Cell Syst 2017; 4:587-599.e4. [PMID: 28601559 PMCID: PMC5493283 DOI: 10.1016/j.cels.2017.05.009] [Citation(s) in RCA: 308] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/03/2017] [Accepted: 05/11/2017] [Indexed: 01/08/2023]
Abstract
This study investigates the challenge of comprehensively cataloging the complete human proteome from a single-cell type using mass spectrometry (MS)-based shotgun proteomics. We modify a classical two-dimensional high-resolution reversed-phase peptide fractionation scheme and optimize a protocol that provides sufficient peak capacity to saturate the sequencing speed of modern MS instruments. This strategy enables the deepest proteome of a human single-cell type to date, with the HeLa proteome sequenced to a depth of ∼584,000 unique peptide sequences and ∼14,200 protein isoforms (∼12,200 protein-coding genes). This depth is comparable with next-generation RNA sequencing and enables the identification of post-translational modifications, including ∼7,000 N-acetylation sites and ∼10,000 phosphorylation sites, without the need for enrichment. We further demonstrate the general applicability and clinical potential of this proteomics strategy by comprehensively quantifying global proteome expression in several different human cancer cell lines and patient tissue samples. Multi-shot proteomics quantifies the protein levels of 12,200+ genes in HeLa cells This essentially complete HeLa proteome has coverage similar to next-gen RNA-seq Deep coverage of major PTMs is achieved without specific enrichment The approach is extendable to other human cell lines and patient samples
Collapse
Affiliation(s)
- Dorte B Bekker-Jensen
- Proteomics Program, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Christian D Kelstrup
- Proteomics Program, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| | - Tanveer S Batth
- Proteomics Program, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Sara C Larsen
- Proteomics Program, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Christa Haldrup
- Departments of Molecular Medicine and Clinical Medicine, Aarhus University Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Jesper B Bramsen
- Departments of Molecular Medicine and Clinical Medicine, Aarhus University Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Karina D Sørensen
- Departments of Molecular Medicine and Clinical Medicine, Aarhus University Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Søren Høyer
- Institute of Pathology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Torben F Ørntoft
- Departments of Molecular Medicine and Clinical Medicine, Aarhus University Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Claus L Andersen
- Departments of Molecular Medicine and Clinical Medicine, Aarhus University Hospital, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Michael L Nielsen
- Proteomics Program, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Jesper V Olsen
- Proteomics Program, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark.
| |
Collapse
|
23
|
Baud A, Wessely F, Mazzacuva F, McCormick J, Camuzeaux S, Heywood WE, Little D, Vowles J, Tuefferd M, Mosaku O, Lako M, Armstrong L, Webber C, Cader MZ, Peeters P, Gissen P, Cowley SA, Mills K. Multiplex High-Throughput Targeted Proteomic Assay To Identify Induced Pluripotent Stem Cells. Anal Chem 2017; 89:2440-2448. [PMID: 28192931 DOI: 10.1021/acs.analchem.6b04368] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Induced pluripotent stem cells have great potential as a human model system in regenerative medicine, disease modeling, and drug screening. However, their use in medical research is hampered by laborious reprogramming procedures that yield low numbers of induced pluripotent stem cells. For further applications in research, only the best, competent clones should be used. The standard assays for pluripotency are based on genomic approaches, which take up to 1 week to perform and incur significant cost. Therefore, there is a need for a rapid and cost-effective assay able to distinguish between pluripotent and nonpluripotent cells. Here, we describe a novel multiplexed, high-throughput, and sensitive peptide-based multiple reaction monitoring mass spectrometry assay, allowing for the identification and absolute quantitation of multiple core transcription factors and pluripotency markers. This assay provides simpler and high-throughput classification into either pluripotent or nonpluripotent cells in 7 min analysis while being more cost-effective than conventional genomic tests.
Collapse
Affiliation(s)
- Anna Baud
- Centre for Translational Omics, UCL Great Ormond Street Institute of Child Health , London, WC1N 1EH, United Kingdom
| | - Frank Wessely
- Department of Physiology, Anatomy & Genetics, Oxford University , Oxford, OX1 3PT, United Kingdom
| | - Francesca Mazzacuva
- Centre for Translational Omics, UCL Great Ormond Street Institute of Child Health , London, WC1N 1EH, United Kingdom
| | - James McCormick
- Centre for Translational Omics, UCL Great Ormond Street Institute of Child Health , London, WC1N 1EH, United Kingdom
| | - Stephane Camuzeaux
- Centre for Translational Omics, UCL Great Ormond Street Institute of Child Health , London, WC1N 1EH, United Kingdom
| | - Wendy E Heywood
- Centre for Translational Omics, UCL Great Ormond Street Institute of Child Health , London, WC1N 1EH, United Kingdom
| | - Daniel Little
- MRC Laboratory for Molecular Cell Biology, University College London , London, WC1E 6BT, United Kingdom
| | - Jane Vowles
- Oxford Parkinson's Disease Centre, University of Oxford , Oxford, OX1 3QX, United Kingdom
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford , Oxford, OX1 3RE, United Kingdom
| | | | - Olukunbi Mosaku
- MRC Laboratory for Molecular Cell Biology, University College London , London, WC1E 6BT, United Kingdom
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University , Newcastle, NE1 3BZ, United Kingdom
| | - Lyle Armstrong
- Institute of Genetic Medicine, Newcastle University , Newcastle, NE1 3BZ, United Kingdom
| | - Caleb Webber
- Department of Physiology, Anatomy & Genetics, Oxford University , Oxford, OX1 3PT, United Kingdom
| | - M Zameel Cader
- The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital , Oxford, OX3 9DS, United Kingdom
| | - Pieter Peeters
- Janssen Research and Development , Beerse, 2340, Belgium
| | - Paul Gissen
- MRC Laboratory for Molecular Cell Biology, University College London , London, WC1E 6BT, United Kingdom
| | - Sally A Cowley
- Oxford Parkinson's Disease Centre, University of Oxford , Oxford, OX1 3QX, United Kingdom
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford , Oxford, OX1 3RE, United Kingdom
| | - Kevin Mills
- Centre for Translational Omics, UCL Great Ormond Street Institute of Child Health , London, WC1N 1EH, United Kingdom
| |
Collapse
|
24
|
Urisman A, Levin RS, Gordan JD, Webber JT, Hernandez H, Ishihama Y, Shokat KM, Burlingame AL. An Optimized Chromatographic Strategy for Multiplexing In Parallel Reaction Monitoring Mass Spectrometry: Insights from Quantitation of Activated Kinases. Mol Cell Proteomics 2017; 16:265-277. [PMID: 27940637 PMCID: PMC5294213 DOI: 10.1074/mcp.m116.058172] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 12/01/2016] [Indexed: 01/06/2023] Open
Abstract
Reliable quantitation of protein abundances in defined sets of cellular proteins is critical to numerous biological applications. Traditional immunodetection-based methods are limited by the quality and availability of specific antibodies, especially for site-specific post-translational modifications. Targeted proteomic methods, including the recently developed parallel reaction monitoring (PRM) mass spectrometry, have enabled accurate quantitative measurements of up to a few hundred specific target peptides. However, the degree of practical multiplexing in label-free PRM workflows remains a significant limitation for the technique. Here we present a strategy for significantly increasing multiplexing in label-free PRM that takes advantage of the superior separation characteristics and retention time stability of meter-scale monolithic silica-C18 column-based chromatography. We show the utility of the approach in quantifying kinase abundances downstream of previously developed active kinase enrichment methodology based on multidrug inhibitor beads. We examine kinase activation dynamics in response to three different MAP kinase inhibitors in colorectal carcinoma cells and demonstrate reliable quantitation of over 800 target peptides from over 150 kinases in a single label-free PRM run. The kinase activity profiles obtained from these analyses reveal compensatory activation of TGF-β family receptors as a response to MAPK blockade. The gains achieved using this label-free PRM multiplexing strategy will benefit a wide array of biological applications.
Collapse
Affiliation(s)
- Anatoly Urisman
- From the ‡Department of Pathology, University of California San Francisco, San Francisco, California;
- §Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Rebecca S Levin
- §Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
- ¶Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California
| | - John D Gordan
- ¶Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California
- ‖Department of Medicine, University of California San Francisco, San Francisco, California
| | - James T Webber
- **Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California
| | - Hilda Hernandez
- §Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| | - Yasushi Ishihama
- ‡‡Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kevan M Shokat
- ¶Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California
- §§Howard Hughes Medical Institute, University of California San Francisco, San Francisco, California
| | - Alma L Burlingame
- §Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| |
Collapse
|
25
|
Nat1 promotes translation of specific proteins that induce differentiation of mouse embryonic stem cells. Proc Natl Acad Sci U S A 2016; 114:340-345. [PMID: 28003464 DOI: 10.1073/pnas.1617234114] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Novel APOBEC1 target 1 (Nat1) (also known as "p97," "Dap5," and "Eif4g2") is a ubiquitously expressed cytoplasmic protein that is homologous to the C-terminal two thirds of eukaryotic translation initiation factor 4G (Eif4g1). We previously showed that Nat1-null mouse embryonic stem cells (mES cells) are resistant to differentiation. In the current study, we found that NAT1 and eIF4G1 share many binding proteins, such as the eukaryotic translation initiation factors eIF3 and eIF4A and ribosomal proteins. However, NAT1 did not bind to eIF4E or poly(A)-binding proteins, which are critical for cap-dependent translation initiation. In contrast, compared with eIF4G1, NAT1 preferentially interacted with eIF2, fragile X mental retardation proteins (FMR), and related proteins and especially with members of the proline-rich and coiled-coil-containing protein 2 (PRRC2) family. We also found that Nat1-null mES cells possess a transcriptional profile similar, although not identical, to the ground state, which is established in wild-type mES cells when treated with inhibitors of the ERK and glycogen synthase kinase 3 (GSK3) signaling pathways. In Nat1-null mES cells, the ERK pathway is suppressed even without inhibitors. Ribosome profiling revealed that translation of mitogen-activated protein kinase kinase kinase 3 (Map3k3) and son of sevenless homolog 1 (Sos1) is suppressed in the absence of Nat1 Forced expression of Map3k3 induced differentiation of Nat1-null mES cells. These data collectively show that Nat1 is involved in the translation of proteins that are required for cell differentiation.
Collapse
|
26
|
Krokhin OV, Spicer V. Generation of accurate peptide retention data for targeted and data independent quantitative LC-MS analysis: Chromatographic lessons in proteomics. Proteomics 2016; 16:2931-2936. [PMID: 27701844 DOI: 10.1002/pmic.201600283] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 09/30/2016] [Indexed: 11/06/2022]
Abstract
The emergence of data-independent quantitative LC-MS/MS analysis protocols further highlights the importance of high-quality reproducible chromatographic procedures. Knowing, controlling and being able to predict the effect of multiple factors that alter peptide RP-HPLC separation selectivity is critical for successful data collection for the construction of ion libraries. Proteomic researchers have often regarded RP-HPLC as a "black box", while vast amount of research on peptide separation is readily available. In addition to obvious parameters, such as the type of ion-pairing modifier, stationary phase and column temperature, we describe the "mysterious" effects of gradient slope, column size and flow rate on peptide separation selectivity. Retention time variations due to these parameters are governed by the linear solvent strength (LSS) theory on a peptide level by the value of its slope S in the basic LSS equation-a parameter that can be accurately predicted. Thus, the application of shallower gradients, higher flow rates, or smaller columns will each increases the relative retention of peptides with higher S-values (long species with multiple positively charged groups). Simultaneous changes to these parameters that each drive shifts in separation selectivity in the same direction should be avoided. The unification of terminology represents another pressing issue in this field of applied proteomics that should be addressed to facilitate further progress.
Collapse
Affiliation(s)
- Oleg V Krokhin
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, Canada.,Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Vic Spicer
- Manitoba Centre for Proteomics and Systems Biology, Winnipeg, Canada
| |
Collapse
|
27
|
de Graaf EL, Pellegrini D, McDonnell LA. Set of Novel Automated Quantitative Microproteomics Protocols for Small Sample Amounts and Its Application to Kidney Tissue Substructures. J Proteome Res 2016; 15:4722-4730. [PMID: 27809536 DOI: 10.1021/acs.jproteome.6b00889] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Here we assessed the ability of an automated sample preparation device equipped with disposable microcolumns to prepare mass-limited samples for high-sensitivity quantitative proteomics, using both label-free and isobaric labeling approaches. First, we compared peptide label-free quantification reproducibility for 1.5-150 μg of cell lysates and found that labware preconditioning was essential for reproducible quantification of <7.5 μg digest. Second, in-solution and on-column tandem mass tag (TMT) labeling protocols were compared and optimized for 1 μg of sample. Surprisingly, standard methods for in-solution and on-column labeling showed poor TMT labeling (50-85%); however, novel optimized and automated protocols restored efficient labeling to >98%. Third, compared with a single long gradient experiment, a simple robotized high-pH fractionation protocol using only 6 μg of starting material doubled the number of unique peptides and increased proteome coverage 1.43-fold. To facilitate the analysis of heterogeneous tissue samples, such as those obtained from laser capture microdissection, a modified BCA protein assay was developed that consumes and detects down to 15 ng of protein. As a proof-of-principle, the modular automated workflow was applied to 0.5 and 1 mm2 mouse kidney cortex and medulla microdissections to show the method's potential for real-life small sample sources and to create kidney substructure-specific proteomes.
Collapse
Affiliation(s)
| | - Davide Pellegrini
- Fondazione Pisana per la Scienza ONLUS , Pisa 56121, Italy.,NEST, Scuola Normale Superiore , Pisa 56127, Italy
| | - Liam A McDonnell
- Fondazione Pisana per la Scienza ONLUS , Pisa 56121, Italy.,Leiden University Medical Center , Leiden 2333 ZA, The Netherlands
| |
Collapse
|
28
|
Tang D, Chen Y, He H, Huang J, Chen W, Peng W, Lu Q, Dai Y. Integrated analysis of mRNA, microRNA and protein in systemic lupus erythematosus-specific induced pluripotent stem cells from urine. BMC Genomics 2016; 17:488. [PMID: 27402083 PMCID: PMC4940874 DOI: 10.1186/s12864-016-2809-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 05/28/2016] [Indexed: 12/16/2022] Open
Abstract
Background In clinical practice, it is difficult to monitor the repeating relapse in patients who have been suffering from systemic lupus erythematosus (SLE). The underlying etiology remains largely unknown. Methods Aiming to understand the pathogenesis of SLE, a detailed study was conducted. Renal tubular cells–derived iPSCs were successfully obtained from the urine of SLE patients and healthy controls. With the purpose to identify simultaneous expression profiling of microRNA, mRNA and protein, Illumina HiSeq™ 2000 System and iTRAQ-coupled 2D LC-MS/MS analysis were utilized in systemic lupus erythematosus-specific induced pluripotent stem cells (SLE-iPSCs) and normal control-iPSCs (NC-iPSCs). The integration of multiple profiling datasets was realized since it could facilitate the identification of non-seed miRNA targets, as well as differentially expressed mRNAs and proteins. Results For this study, profiling datasets of 1099 differentially expressed mRNAs, 223 differentially expressed microRNAs and 94 differentially expressed proteins were integrated. In order to investigate the influence of miRNA on the processes of regulating mRNAs and proteins’ levels, potential targets of differentially expressed mRNAs and proteins were predicted using miRanda, TargetScan and Pictar. Multiple profiling datasets were integrated to facilitate the identification of miRNA targets, as well as differentially expressed mRNAs and proteins. Through gene ontology (GO) analysis of differentially expressed mRNAs and proteins, biological processes that drive proliferation were identified, such as mRNA processing and translation. Western blot and Q-PCR confirmed AK4 protein and mRNA up-regulation. The findings also showed that TAGLN’s protein and mRNA level were down-regulated in SLE-iPSCs, both miR-371a-5p and let-7a-5p in SLE-iPSC were down-regulated and verified using Q-PCR. The up-regulation of AK4 involved in nucleotide biosynthesis suggested a general acceleration of anabolic metabolism induced by down-regulated miR-371a-5p, which might contribute to SLE. Conclusion Based on high throughput analysis, integrated miRNA, mRNA, and protein expression data were generated. Differentially expressed dates were also adopted in conjunction with in-silico tools to identify potential candidates for SLE-iPSCs. Representative miRNA, mRNA and proteins were verified. It was also expected that the knowledge gained from this study can be applied to assess the usefulness of pathogenesis and novel biomarker candidates of SLE, which may develop a new way for SLE diagnosis. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2809-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Donge Tang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, People's Republic of China
| | - Yuyu Chen
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, People's Republic of China
| | - Huiyan He
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, People's Republic of China
| | - Jianrong Huang
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, People's Republic of China
| | - Wenbiao Chen
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, People's Republic of China
| | - Wujian Peng
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, People's Republic of China
| | - Qianjin Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha, Hunan, 410011, People's Republic of China
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, 518020, People's Republic of China.
| |
Collapse
|
29
|
Nakashima Y, Omasa T. What Kind of Signaling Maintains Pluripotency and Viability in Human-Induced Pluripotent Stem Cells Cultured on Laminin-511 with Serum-Free Medium? Biores Open Access 2016; 5:84-93. [PMID: 27096107 PMCID: PMC4834485 DOI: 10.1089/biores.2016.0001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Xeno-free medium contains no animal-derived components, but is composed of minimal growth factors and is serum free; the medium may be supplemented with insulin, transferrin, and selenium (ITS medium). Serum-free and xeno-free culture of human-induced pluripotent stem cells (hiPSCs) uses a variety of components based on ITS medium and Dulbecco's modified Eagle's medium/Ham's nutrient mixture F12 (DMEM/F12) that contain high levels of iron salt and glucose. Culture of hiPSCs also requires scaffolding materials, such as extracellular matrix, collagen, fibronectin, laminin, proteoglycan, and vitronectin. The scaffolding component laminin-511, which is composed of α5, β1, and γ1 chains, binds to α3β1, α6β1, and α6β4 integrins on the cell membrane to induce activation of the PI3K/AKT- and Ras/MAPK-dependent signaling pathways. In hiPSCs, the interaction of laminin-511/α6β1 integrin with the cell–cell adhesion molecule E-cadherin confers protection against apoptosis through the Ras homolog gene family member A (RhoA)/Rho kinase (ROCK) signaling pathway (the major pathways for cell death) and the proto-oncogene tyrosine-protein kinase Fyn (Fyn)-RhoA-ROCK signaling pathway. The expression levels of α6β1 integrin and E-cadherin on cell membranes are controlled through the activation of insulin receptor/insulin, FGF receptor/FGF2, or activin-like kinase 5 (ALK5)-dependent TGF-β signaling. A combination of growth factors, medium constituents, cell membrane-located E-cadherin, and α6β1 integrin-induced signaling is required for pluripotent cell proliferation and for optimal cell survival on a laminin-511 scaffold. In this review, we discuss and explore the influence of growth factors on the cadherin and integrin signaling pathways in serum-free and xeno-free cultures of hiPSCs during the preparation of products for regenerative medicinal therapies. In addition, we suggest the optimum serum-free medium components for use with laminin-511, a new scaffold for hiPSC culture.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Department of Material and Life Science, Graduate School of Engineering, Osaka University , Osaka, Japan
| | - Takeshi Omasa
- Department of Material and Life Science, Graduate School of Engineering, Osaka University , Osaka, Japan
| |
Collapse
|
30
|
Moravcová D, Rantamäki AH, Duša F, Wiedmer SK. Monoliths in capillary electrochromatography and capillary liquid chromatography in conjunction with mass spectrometry. Electrophoresis 2016; 37:880-912. [PMID: 26800083 DOI: 10.1002/elps.201500520] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/18/2015] [Accepted: 12/21/2015] [Indexed: 12/29/2022]
Abstract
Here, we have reviewed separation studies utilizing monolithic capillary columns for separation of compounds preceding MS analysis. The review is divided in two parts according to the used separation method, namely CEC and capillary LC (cLC). Based on our overview, monolithic CEC-MS technique have been more focused on the syntheses of highly specialized and selective separation phase materials for fast and efficient separation of specific types of analytes. In contrast, monolithic cLC-MS is more widely used and is often employed, for instance, in the analysis of oligonucleotides, metabolites, and peptides and proteins in proteomic studies. While poly(styrene-divinylbenzene)-based and silica-based monolithic capillaries found their place in proteomic analyses, the other laboratory-synthesized monoliths still wait for their wider utilization in routine analyses. The development of new monolithic materials will most likely continue due to the demand of more efficient and rapid separation of increasingly complex samples.
Collapse
Affiliation(s)
- Dana Moravcová
- Institute of Analytical Chemistry of the CAS, v. v. i, Brno, Czech Republic
| | | | - Filip Duša
- Institute of Analytical Chemistry of the CAS, v. v. i, Brno, Czech Republic
| | | |
Collapse
|
31
|
Affiliation(s)
- Jennifer S Brodbelt
- Department of Chemistry, University of Texas at Austin , Austin, Texas 78712, United States
| |
Collapse
|
32
|
Shinoda K, Ohyama K, Hasegawa Y, Chang HY, Ogura M, Sato A, Hong H, Hosono T, Sharp LZ, Scheel DW, Graham M, Ishihama Y, Kajimura S. Phosphoproteomics Identifies CK2 as a Negative Regulator of Beige Adipocyte Thermogenesis and Energy Expenditure. Cell Metab 2015; 22:997-1008. [PMID: 26525534 PMCID: PMC4670581 DOI: 10.1016/j.cmet.2015.09.029] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 07/21/2015] [Accepted: 09/29/2015] [Indexed: 01/18/2023]
Abstract
Catecholamines promote lipolysis both in brown and white adipocytes, whereas the same stimuli preferentially activate thermogenesis in brown adipocytes. Molecular mechanisms for the adipose-selective activation of thermogenesis remain poorly understood. Here, we employed quantitative phosphoproteomics to map global and temporal phosphorylation profiles in brown, beige, and white adipocytes under β3-adrenenoceptor activation and identified kinases responsible for the adipose-selective phosphorylation profiles. We found that casein kinase2 (CK2) activity is preferentially higher in white adipocytes than brown/beige adipocytes. Genetic or pharmacological blockade of CK2 in white adipocytes activates the thermogenic program in response to cAMP stimuli. Such activation is largely through reduced CK2-mediated phosphorylation of class I HDACs. Notably, inhibition of CK2 promotes beige adipocyte biogenesis and leads to an increase in whole-body energy expenditure and ameliorates diet-induced obesity and insulin resistance. These results indicate that CK2 is a plausible target to rewire the β3-adrenenoceptor signaling cascade that promotes thermogenesis in adipocytes.
Collapse
Affiliation(s)
- Kosaku Shinoda
- UCSF Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143-0669, USA
| | - Kana Ohyama
- UCSF Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143-0669, USA
| | - Yutaka Hasegawa
- UCSF Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143-0669, USA
| | - Hsin-Yi Chang
- Department of Molecular and Cellular Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Mayu Ogura
- Department of Molecular and Cellular Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Ayaka Sato
- Department of Molecular and Cellular Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Haemin Hong
- UCSF Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143-0669, USA
| | - Takashi Hosono
- UCSF Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143-0669, USA
| | - Louis Z Sharp
- UCSF Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143-0669, USA
| | - David W Scheel
- UCSF Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143-0669, USA
| | - Mark Graham
- Cardiovascular Group, Department of Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, CA 92008, USA
| | - Yasushi Ishihama
- Department of Molecular and Cellular Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Shingo Kajimura
- UCSF Diabetes Center and Department of Cell and Tissue Biology, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143-0669, USA.
| |
Collapse
|
33
|
Wakabayashi M, Kyono Y, Sugiyama N, Ishihama Y. Extended Coverage of Singly and Multiply Phosphorylated Peptides from a Single Titanium Dioxide Microcolumn. Anal Chem 2015; 87:10213-21. [DOI: 10.1021/acs.analchem.5b01216] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Masaki Wakabayashi
- Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29, Yoshida-Shimo-Adachi-Cho,
Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yutaka Kyono
- Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29, Yoshida-Shimo-Adachi-Cho,
Sakyo-ku, Kyoto, 606-8501, Japan
| | - Naoyuki Sugiyama
- Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29, Yoshida-Shimo-Adachi-Cho,
Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yasushi Ishihama
- Graduate School of Pharmaceutical
Sciences, Kyoto University, 46-29, Yoshida-Shimo-Adachi-Cho,
Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
34
|
Holl S, Mohammed Y, Zimmermann O, Palmblad M. Scientific workflow optimization for improved peptide and protein identification. BMC Bioinformatics 2015; 16:284. [PMID: 26335531 PMCID: PMC4558836 DOI: 10.1186/s12859-015-0714-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/24/2015] [Indexed: 01/18/2023] Open
Abstract
Background Peptide-spectrum matching is a common step in most data processing workflows for mass spectrometry-based proteomics. Many algorithms and software packages, both free and commercial, have been developed to address this task. However, these algorithms typically require the user to select instrument- and sample-dependent parameters, such as mass measurement error tolerances and number of missed enzymatic cleavages. In order to select the best algorithm and parameter set for a particular dataset, in-depth knowledge about the data as well as the algorithms themselves is needed. Most researchers therefore tend to use default parameters, which are not necessarily optimal. Results We have applied a new optimization framework for the Taverna scientific workflow management system (http://ms-utils.org/Taverna_Optimization.pdf) to find the best combination of parameters for a given scientific workflow to perform peptide-spectrum matching. The optimizations themselves are non-trivial, as demonstrated by several phenomena that can be observed when allowing for larger mass measurement errors in sequence database searches. On-the-fly parameter optimization embedded in scientific workflow management systems enables experts and non-experts alike to extract the maximum amount of information from the data. The same workflows could be used for exploring the parameter space and compare algorithms, not only for peptide-spectrum matching, but also for other tasks, such as retention time prediction. Conclusion Using the optimization framework, we were able to learn about how the data was acquired as well as the explored algorithms. We observed a phenomenon identifying many ammonia-loss b-ion spectra as peptides with N-terminal pyroglutamate and a large precursor mass measurement error. These insights could only be gained with the extension of the common range for the mass measurement error tolerance parameters explored by the optimization framework.
Collapse
Affiliation(s)
- Sonja Holl
- Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, 52425, Jülich, Germany.
| | - Yassene Mohammed
- Center for Proteomics and Metabolomics, Leiden University Medical Center, PO Box 9600, 2300, RC, Leiden, The Netherlands.
| | - Olav Zimmermann
- Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich, 52425, Jülich, Germany.
| | - Magnus Palmblad
- Center for Proteomics and Metabolomics, Leiden University Medical Center, PO Box 9600, 2300, RC, Leiden, The Netherlands.
| |
Collapse
|
35
|
Abstract
In proteomics, nano-LC is arguably the most common tool for separating peptides/proteins prior to MS. The main advantage of nano-LC is enhanced sensitivity, as compounds enter the MS in more concentrated bands. This is particularly relevant for determining low abundant compounds in limited samples. Nano-LC columns can produce peak capacities of 1000 or more, and very narrow columns can be used to perform proteomics of 1000 cells or less. Also, nano-LC can be coupled with online add-ons such as selective trap columns or enzymatic reactors, for faster and more automated analysis. Nano-LC is today an established tool for research laboratories; but can nano-LC-based systems soon be ready for more routine settings, such as in clinics?
Collapse
|
36
|
Savitski MM, Wilhelm M, Hahne H, Kuster B, Bantscheff M. A Scalable Approach for Protein False Discovery Rate Estimation in Large Proteomic Data Sets. Mol Cell Proteomics 2015; 14:2394-404. [PMID: 25987413 DOI: 10.1074/mcp.m114.046995] [Citation(s) in RCA: 291] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Indexed: 02/06/2023] Open
Abstract
Calculating the number of confidently identified proteins and estimating false discovery rate (FDR) is a challenge when analyzing very large proteomic data sets such as entire human proteomes. Biological and technical heterogeneity in proteomic experiments further add to the challenge and there are strong differences in opinion regarding the conceptual validity of a protein FDR and no consensus regarding the methodology for protein FDR determination. There are also limitations inherent to the widely used classic target-decoy strategy that particularly show when analyzing very large data sets and that lead to a strong over-representation of decoy identifications. In this study, we investigated the merits of the classic, as well as a novel target-decoy-based protein FDR estimation approach, taking advantage of a heterogeneous data collection comprised of ∼19,000 LC-MS/MS runs deposited in ProteomicsDB (https://www.proteomicsdb.org). The "picked" protein FDR approach treats target and decoy sequences of the same protein as a pair rather than as individual entities and chooses either the target or the decoy sequence depending on which receives the highest score. We investigated the performance of this approach in combination with q-value based peptide scoring to normalize sample-, instrument-, and search engine-specific differences. The "picked" target-decoy strategy performed best when protein scoring was based on the best peptide q-value for each protein yielding a stable number of true positive protein identifications over a wide range of q-value thresholds. We show that this simple and unbiased strategy eliminates a conceptual issue in the commonly used "classic" protein FDR approach that causes overprediction of false-positive protein identification in large data sets. The approach scales from small to very large data sets without losing performance, consistently increases the number of true-positive protein identifications and is readily implemented in proteomics analysis software.
Collapse
Affiliation(s)
| | - Mathias Wilhelm
- §Chair for Proteomics and Bioanalytics, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany; ¶SAP SE, Dietmar-Hopp-Allee 16, 69190 Walldorf, Germany
| | - Hannes Hahne
- §Chair for Proteomics and Bioanalytics, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
| | - Bernhard Kuster
- §Chair for Proteomics and Bioanalytics, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany; ‖Center for Integrated Protein Science Munich, Emil Erlenmeyer Forum 5, 85354 Freising, Germany
| | - Marcus Bantscheff
- From the ‡Cellzome GmbH, Meyerhofstrasse 1, 69117 Heidelberg, Germany;
| |
Collapse
|
37
|
Wang G, He Z, Shi G, Wang H, Zhang Q, Li Y. Controllable construction of Titanium dioxide-Zirconium dioxide@Zinc hydroxyfluoride networks in micro-capillaries for bio-analysis. J Colloid Interface Sci 2015; 446:290-7. [DOI: 10.1016/j.jcis.2015.01.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 01/15/2015] [Accepted: 01/20/2015] [Indexed: 11/27/2022]
|
38
|
Pripuzova NS, Getie-Kebtie M, Grunseich C, Sweeney C, Malech H, Alterman MA. Development of a protein marker panel for characterization of human induced pluripotent stem cells (hiPSCs) using global quantitative proteome analysis. Stem Cell Res 2015; 14:323-38. [PMID: 25840413 PMCID: PMC5778352 DOI: 10.1016/j.scr.2015.01.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 01/23/2015] [Accepted: 01/30/2015] [Indexed: 12/13/2022] Open
Abstract
The emergence of new methods for reprogramming of adult somatic cells into induced pluripotent stem cells (iPSC) led to the development of new approaches in drug discovery and regenerative medicine. Investigation of the molecular mechanisms underlying the self-renewal, expansion and differentiation of human iPSC (hiPSC) should lead to improvements in the manufacture of safe and reliable cell therapy products. The goal of our study was qualitative and quantitative proteomic characterizations of hiPSC by means of electrospray ionization (ESI)-MSe and MALDI-TOF/TOF mass spectrometry (MS). Proteomes of hiPSCs of different somatic origins: fibroblasts and peripheral blood CD34+ cells, reprogrammed by the same technique, were compared with the original somatic cells and hESC. Quantitative proteomic comparison revealed approximately 220 proteins commonly up-regulated in all three pluripotent stem cell lines compared to the primary cells. Expression of 21 proteins previously reported as pluripotency markers was up-regulated in both hiPSCs (8 were confirmed by Western blot). A number of novel candidate marker proteins with the highest fold-change difference between hiPSCs/hESC and somatic cells discovered by MS were confirmed by Western blot. A panel of 22 candidate marker proteins of hiPSC was developed and expression of these proteins was confirmed in 8 additional hiPSC lines.
Collapse
Affiliation(s)
- Natalia S Pripuzova
- Tumor Vaccine and Biotechnology Branch, Division of Cellular and Gene Therapies, FDA, Center for Biologics Evaluation and Research, Bethesda, MD 20892-4555, USA
| | - Melkamu Getie-Kebtie
- Tumor Vaccine and Biotechnology Branch, Division of Cellular and Gene Therapies, FDA, Center for Biologics Evaluation and Research, Bethesda, MD 20892-4555, USA
| | - Christopher Grunseich
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3705, USA
| | - Colin Sweeney
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1456, USA
| | - Harry Malech
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1456, USA
| | - Michail A Alterman
- Tumor Vaccine and Biotechnology Branch, Division of Cellular and Gene Therapies, FDA, Center for Biologics Evaluation and Research, Bethesda, MD 20892-4555, USA.
| |
Collapse
|
39
|
Camerini S, Mauri P. The role of protein and peptide separation before mass spectrometry analysis in clinical proteomics. J Chromatogr A 2014; 1381:1-12. [PMID: 25618357 DOI: 10.1016/j.chroma.2014.12.035] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 11/25/2022]
Abstract
The purpose of clinical proteomics is to characterise protein profiles of a plethora of diseases with the aim of finding specific biomarkers. These are particularly valuable for early diagnosis, and represent key molecules suitable to elucidate pathogenic mechanisms. Samples deriving from patients (i.e. blood, urine, cerebrospinal fluid, biopsies) are the sources for clinical proteomics. Due to the complexity of the extracted samples their direct analysis is unachievable. Any analytical clinical proteomics study should start with the choice of the optimal combination of strategies with respect to both sample preparations and MS approaches. Protein or peptide fractionation (off-line or on-line) is essential to reduce complexity of biological samples and to achieve the most complete and reproducible analysis. The aim of this review is to introduce the readers to a functional range of strategies to help scientists in their proteomics set up. In particular, the separation approaches of proteins or peptides (both gel-based and gel-free) are reviewed with special attention paid to their advantages and limitations, and to the different liquid chromatography techniques used to peptide fractionation after protein enzymatic digestion and before their detection. Finally, the role of mass spectrometry (MS) for protein identification and quantification is discussed including emerging MS data acquisition strategies.
Collapse
Affiliation(s)
- Serena Camerini
- Dept of Cell Biology and Neurosciences Higher Institute of Health (ISS), Rome, Italy
| | - Pierluigi Mauri
- Institute for Biomedical Technologies (ITB-CNR), Segrate, and Institute of Life Science - Scuola Superiore Sant'Anna, Pisa, Italy.
| |
Collapse
|
40
|
Han J, Ye L, Xu L, Zhou Z, Gao F, Xiao Z, Wang Q, Zhang B. Towards high peak capacity separations in normal pressure nanoflow liquid chromatography using meter long packed capillary columns. Anal Chim Acta 2014; 852:267-73. [DOI: 10.1016/j.aca.2014.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/31/2014] [Accepted: 09/07/2014] [Indexed: 10/24/2022]
|
41
|
Imamura H, Sugiyama N, Wakabayashi M, Ishihama Y. Large-Scale Identification of Phosphorylation Sites for Profiling Protein Kinase Selectivity. J Proteome Res 2014; 13:3410-9. [DOI: 10.1021/pr500319y] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Haruna Imamura
- Graduate
School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Naoyuki Sugiyama
- Graduate
School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masaki Wakabayashi
- Graduate
School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yasushi Ishihama
- Graduate
School of Pharmaceutical
Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
42
|
Horie K, Kamakura T, Ikegami T, Wakabayashi M, Kato T, Tanaka N, Ishihama Y. Hydrophilic Interaction Chromatography Using a Meter-Scale Monolithic Silica Capillary Column for Proteomics LC-MS. Anal Chem 2014; 86:3817-24. [DOI: 10.1021/ac4038625] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kanta Horie
- Eisai Co., Ltd, Pharmaceutical Science and Technology
Core Function Unit, Global Formulation Research, Kawashima, Kakamigahara, Gifu 501-6195, Japan
- Graduate
School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| | - Takeo Kamakura
- Graduate
School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| | - Tohru Ikegami
- Department
of Biomolecular Engineering, Kyoto Institute of Technology, Sakyo-ku, Kyoto, Kyoto 606-8585, Japan
| | - Masaki Wakabayashi
- Graduate
School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| | - Takashi Kato
- Eisai Co., Ltd, Pharmaceutical Science and Technology
Core Function Unit, Global Formulation Research, Kawashima, Kakamigahara, Gifu 501-6195, Japan
| | - Nobuo Tanaka
- Department
of Biomolecular Engineering, Kyoto Institute of Technology, Sakyo-ku, Kyoto, Kyoto 606-8585, Japan
- GL Sciences Inc., 237-2
Sayamagahara, Iruma, Saitama 358-0032, Japan
| | - Yasushi Ishihama
- Graduate
School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Kyoto 606-8501, Japan
| |
Collapse
|
43
|
Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells. Nat Methods 2014; 11:319-24. [PMID: 24487582 DOI: 10.1038/nmeth.2834] [Citation(s) in RCA: 1197] [Impact Index Per Article: 119.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 12/20/2013] [Indexed: 12/13/2022]
Abstract
Mass spectrometry (MS)-based proteomics typically employs multistep sample-preparation workflows that are subject to sample contamination and loss. We report an in-StageTip method for performing sample processing, from cell lysis through elution of purified peptides, in a single, enclosed volume. This robust and scalable method largely eliminates contamination or loss. Peptides can be eluted in several fractions or in one step for single-run proteome analysis. In one day, we obtained the largest proteome coverage to date for budding and fission yeast, and found that protein copy numbers in these cells were highly correlated (R(2) = 0.78). Applying the in-StageTip method to quadruplicate measurements of a human cell line, we obtained copy-number estimates for 9,667 human proteins and observed excellent quantitative reproducibility between replicates (R(2) = 0.97). The in-StageTip method is straightforward and generally applicable in biological or clinical applications.
Collapse
|
44
|
Marino F, Cristobal A, Binai NA, Bache N, Heck AJR, Mohammed S. Characterization and usage of the EASY-spray technology as part of an online 2D SCX-RP ultra-high pressure system. Analyst 2014; 139:6520-8. [DOI: 10.1039/c4an01568a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The EASY-spray technology can now be implemented as a simple online 2D SCX-RP ultra-high pressure system, which allows one to reach deep proteome coverages.
Collapse
Affiliation(s)
- Fabio Marino
- Biomolecular Mass Spectrometry and Proteomics
- Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences
- University of Utrecht
- 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Centre
| | - Alba Cristobal
- Biomolecular Mass Spectrometry and Proteomics
- Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences
- University of Utrecht
- 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Centre
| | - Nadine A. Binai
- Biomolecular Mass Spectrometry and Proteomics
- Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences
- University of Utrecht
- 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Centre
| | | | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics
- Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences
- University of Utrecht
- 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Centre
| | - Shabaz Mohammed
- Biomolecular Mass Spectrometry and Proteomics
- Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences
- University of Utrecht
- 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Centre
| |
Collapse
|
45
|
IWASAKI M, ISHIHAMA Y. Challenges Facing Complete Human Proteome Analysis. CHROMATOGRAPHY 2014. [DOI: 10.15583/jpchrom.2014.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Mio IWASAKI
- Center for iPS Cell Research and Application, Kyoto University
| | | |
Collapse
|
46
|
Mayne J, Starr AE, Ning Z, Chen R, Chiang CK, Figeys D. Fine Tuning of Proteomic Technologies to Improve Biological Findings: Advancements in 2011–2013. Anal Chem 2013; 86:176-95. [DOI: 10.1021/ac403551f] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Janice Mayne
- Ottawa Institute of
Systems Biology, Department of Biochemistry, Microbiology
and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada K1H8M5
| | - Amanda E. Starr
- Ottawa Institute of
Systems Biology, Department of Biochemistry, Microbiology
and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada K1H8M5
| | - Zhibin Ning
- Ottawa Institute of
Systems Biology, Department of Biochemistry, Microbiology
and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada K1H8M5
| | - Rui Chen
- Ottawa Institute of
Systems Biology, Department of Biochemistry, Microbiology
and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada K1H8M5
| | - Cheng-Kang Chiang
- Ottawa Institute of
Systems Biology, Department of Biochemistry, Microbiology
and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada K1H8M5
| | - Daniel Figeys
- Ottawa Institute of
Systems Biology, Department of Biochemistry, Microbiology
and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, Canada K1H8M5
| |
Collapse
|
47
|
Grigoryan M, Shamshurin D, Spicer V, Krokhin OV. Unifying Expression Scale for Peptide Hydrophobicity in Proteomic Reversed Phase High-Pressure Liquid Chromatography Experiments. Anal Chem 2013; 85:10878-86. [DOI: 10.1021/ac402310t] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Marine Grigoryan
- Manitoba Centre for Proteomics and Systems
Biology and ‡Department of Internal Medicine, University of Manitoba, 799 JBRC,
715 McDermot Avenue, Winnipeg, R3E 3P4, Canada
| | - Dmitry Shamshurin
- Manitoba Centre for Proteomics and Systems
Biology and ‡Department of Internal Medicine, University of Manitoba, 799 JBRC,
715 McDermot Avenue, Winnipeg, R3E 3P4, Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems
Biology and ‡Department of Internal Medicine, University of Manitoba, 799 JBRC,
715 McDermot Avenue, Winnipeg, R3E 3P4, Canada
| | - Oleg V. Krokhin
- Manitoba Centre for Proteomics and Systems
Biology and ‡Department of Internal Medicine, University of Manitoba, 799 JBRC,
715 McDermot Avenue, Winnipeg, R3E 3P4, Canada
| |
Collapse
|
48
|
Zhang Y, Fonslow BR, Shan B, Baek MC, Yates JR. Protein analysis by shotgun/bottom-up proteomics. Chem Rev 2013; 113:2343-94. [PMID: 23438204 PMCID: PMC3751594 DOI: 10.1021/cr3003533] [Citation(s) in RCA: 986] [Impact Index Per Article: 89.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Yaoyang Zhang
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bryan R. Fonslow
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bing Shan
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Moon-Chang Baek
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Molecular Medicine, Cell and Matrix Biology Research Institute, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - John R. Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|