Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ping Yan is active.

Publication


Featured researches published by Ping Yan.


Nature | 2016

Proteogenomics connects somatic mutations to signalling in breast cancer

Philipp Mertins; D. R. Mani; Kelly V. Ruggles; Michael A. Gillette; Karl R. Clauser; Pei Wang; Xianlong Wang; Jana W. Qiao; Song Cao; Francesca Petralia; Emily Kawaler; Filip Mundt; Karsten Krug; Zhidong Tu; Jonathan T. Lei; Michael L. Gatza; Matthew D. Wilkerson; Charles M. Perou; Venkata Yellapantula; Kuan Lin Huang; Chenwei Lin; Michael D. McLellan; Ping Yan; Sherri R. Davies; R. Reid Townsend; Steven J. Skates; Jing Wang; Bing Zhang; Christopher R. Kinsinger; Mehdi Mesri

Summary Somatic mutations have been extensively characterized in breast cancer, but the effects of these genetic alterations on the proteomic landscape remain poorly understood. We describe quantitative mass spectrometry-based proteomic and phosphoproteomic analyses of 105 genomically annotated breast cancers of which 77 provided high-quality data. Integrated analyses allowed insights into the somatic cancer genome including the consequences of chromosomal loss, such as the 5q deletion characteristic of basal-like breast cancer. The 5q trans effects were interrogated against the Library of Integrated Network-based Cellular Signatures, thereby connecting CETN3 and SKP1 loss to elevated expression of EGFR, and SKP1 loss also to increased SRC. Global proteomic data confirmed a stromal-enriched group in addition to basal and luminal clusters and pathway analysis of the phosphoproteome identified a G Protein-coupled receptor cluster that was not readily identified at the mRNA level. Besides ERBB2, other amplicon-associated, highly phosphorylated kinases were identified, including CDK12, PAK1, PTK2, RIPK2 and TLK2. We demonstrate that proteogenomic analysis of breast cancer elucidates functional consequences of somatic mutations, narrows candidate nominations for driver genes within large deletions and amplified regions, and identifies therapeutic targets.


Nature Biotechnology | 2011

A targeted proteomics–based pipeline for verification of biomarkers in plasma

Jeffrey R. Whiteaker; Chenwei Lin; Jacob Kennedy; Liming Hou; Mary Trute; Izabela Sokal; Ping Yan; Regine M. Schoenherr; Lei Zhao; Uliana J. Voytovich; Karen S. Kelly-Spratt; Alexei L. Krasnoselsky; Philip R. Gafken; Jason M. Hogan; Lisa A. Jones; Pei Wang; Lynn M. Amon; Lewis A. Chodosh; Peter S. Nelson; Martin W. McIntosh; Christopher J. Kemp; Amanda G. Paulovich

High-throughput technologies can now identify hundreds of candidate protein biomarkers for any disease with relative ease. However, because there are no assays for the majority of proteins and de novo immunoassay development is prohibitively expensive, few candidate biomarkers are tested in clinical studies. We tested whether the analytical performance of a biomarker identification pipeline based on targeted mass spectrometry would be sufficient for data-dependent prioritization of candidate biomarkers, de novo development of assays and multiplexed biomarker verification. We used a data-dependent triage process to prioritize a subset of putative plasma biomarkers from >1,000 candidates previously identified using a mouse model of breast cancer. Eighty-eight novel quantitative assays based on selected reaction monitoring mass spectrometry were developed, multiplexed and evaluated in 80 plasma samples. Thirty-six proteins were verified as being elevated in the plasma of tumor-bearing animals. The analytical performance of this pipeline suggests that it should support the use of an analogous approach with human samples.


Nature Methods | 2014

Demonstrating the feasibility of large-scale development of standardized assays to quantify human proteins

Jacob Kennedy; Susan E. Abbatiello; Kyunggon Kim; Ping Yan; Jeffrey R. Whiteaker; Chenwei Lin; Jun Seok Kim; Yuzheng Zhang; Xianlong Wang; Richard G. Ivey; Lei Zhao; Hophil Min; Youngju Lee; Myeong Hee Yu; Eun Gyeong Yang; Cheolju Lee; Pei Wang; Henry Rodriguez; Youngsoo Kim; Steven A. Carr; Amanda G. Paulovich

Multiple reaction monitoring (MRM) mass spectrometry has been successfully applied to monitor targeted proteins in biological specimens, raising the possibility that assays could be configured to measure all human proteins. We report the results of a pilot study designed to test the feasibility of a large-scale, international effort for MRM assay generation. We have configured, validated across three laboratories and made publicly available as a resource to the community 645 novel MRM assays representing 319 proteins expressed in human breast cancer. Assays were multiplexed in groups of >150 peptides and deployed to quantify endogenous analytes in a panel of breast cancer–related cell lines. The median assay precision was 5.4%, with high interlaboratory correlation (R2 > 0.96). Peptide measurements in breast cancer cell lines were able to discriminate among molecular subtypes and identify genome-driven changes in the cancer proteome. These results establish the feasibility of a large-scale effort to develop an MRM assay resource.


Journal of Proteome Research | 2014

Panorama: A Targeted Proteomics Knowledge Base

Vagisha Sharma; Josh Eckels; Greg Taylor; Nicholas J. Shulman; Andrew B. Stergachis; Shannon A. Joyner; Ping Yan; Jeffrey R. Whiteaker; Goran N. Halusa; Birgit Schilling; Bradford W. Gibson; Christopher M. Colangelo; Amanda G. Paulovich; Steven A. Carr; Jacob D. Jaffe; Michael J. MacCoss; Brendan MacLean

Panorama is a web application for storing, sharing, analyzing, and reusing targeted assays created and refined with Skyline,1 an increasingly popular Windows client software tool for targeted proteomics experiments. Panorama allows laboratories to store and organize curated results contained in Skyline documents with fine-grained permissions, which facilitates distributed collaboration and secure sharing of published and unpublished data via a web-browser interface. It is fully integrated with the Skyline workflow and supports publishing a document directly to a Panorama server from the Skyline user interface. Panorama captures the complete Skyline document information content in a relational database schema. Curated results published to Panorama can be aggregated and exported as chromatogram libraries. These libraries can be used in Skyline to pick optimal targets in new experiments and to validate peak identification of target peptides. Panorama is open-source and freely available. It is distributed as part of LabKey Server,2 an open source biomedical research data management system. Laboratories and organizations can set up Panorama locally by downloading and installing the software on their own servers. They can also request freely hosted projects on https://panoramaweb.org, a Panorama server maintained by the Department of Genome Sciences at the University of Washington.


Nature Methods | 2014

CPTAC Assay Portal: a repository of targeted proteomic assays

Jeffrey R. Whiteaker; Goran N. Halusa; Andrew N. Hoofnagle; Vagisha Sharma; Brendan MacLean; Ping Yan; John A. Wrobel; Jacob Kennedy; D. R. Mani; Lisa J. Zimmerman; Matthew R. Meyer; Mehdi Mesri; Henry Rodriguez; Amanda G. Paulovich

To address these issues, the Clinical Proteomic Tumor Analysis Consortium (CPTAC) of the National Cancer Institute (NCI) has launched an Assay Portal (http://assays.cancer.gov) to serve as a public repository of well-characterized quantitative, MS-based, targeted proteomic assays. The purpose of the CPTAC Assay Portal is to facilitate widespread adoption of targeted MS assays by disseminating SOPs, reagents, and assay characterization data for highly characterized assays. A primary aim of the NCI-supported portal is to bring together clinicians or biologists and analytical chemists to answer hypothesis-driven questions using targeted, MS-based assays. Assay content is easily accessed through queries and filters, enabling investigators to find assays to proteins relevant to their areas of interest. Detailed characterization data are available for each assay, enabling researchers to evaluate assay performance prior to launching the assay in their own laboratory.


Molecular & Cellular Proteomics | 2012

Sequential Multiplexed Analyte Quantification Using Peptide Immunoaffinity Enrichment Coupled to Mass Spectrometry

Jeffrey R. Whiteaker; Lei Zhao; Chenwei Lin; Ping Yan; Pei Wang; Amanda G. Paulovich

Peptide immunoaffinity enrichment coupled to selected reaction monitoring (SRM) mass spectrometry (immuno-SRM) has emerged as a technology with great potential for quantitative proteomic assays. One advantage over traditional immunoassays is the tremendous potential for concurrent quantification of multiple analytes from a given sample (i.e. multiplex analysis). We sought to explore the capacity of the immuno-SRM technique for analyzing large numbers of analytes by evaluating the multiplex capabilities and demonstrating the sequential analysis of groups of peptides from a single sample. To evaluate multiplex analysis, immuno-SRM assays were arranged in groups of 10, 20, 30, 40, and 50 peptides using a common set of reagents. The multiplex immuno-SRM assays were used to measure synthetic peptides added to plasma covering several orders of magnitude concentration. Measurements made in large multiplex groups were highly correlated (r2 ≥ 0.98) and featured good agreement (bias ≤ 1%) compared with single-plex assays or a 10-plex configuration. The ability to sequentially enrich sets of analyte peptides was demonstrated by enriching groups of 10 peptides from a plasma sample in a sequential fashion. The data show good agreement (bias ≤ 1.5%) and similar reproducibility regardless of enrichment order. These significant advancements demonstrate the utility of immuno-SRM for analyzing large numbers of analytes, such as in large biomarker verification experiments or in pathway-based targeted analysis.


Proteomics | 2012

Multiplexed quantification of estrogen receptor and HER2/Neu in tissue and cell lysates by peptide immunoaffinity enrichment mass spectrometry

Regine M. Schoenherr; Jeffrey R. Whiteaker; Lei Zhao; Richard G. Ivey; Mary Trute; Jacob Kennedy; Uliana J. Voytovich; Ping Yan; Chenwei Lin; Amanda G. Paulovich

Access to a wider range of quantitative protein assays would significantly impact the number and use of tissue markers in guiding disease treatment. Quantitative mass spectrometry‐based peptide and protein assays, such as immuno‐SRM assays, have seen tremendous growth in recent years in application to protein quantification in biological fluids such as plasma or urine. Here, we extend the capability of the technique by demonstrating the application of a multiplexed immuno‐SRM assay for quantification of estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2) levels in cell line lysates and human surgical specimens. The performance of the assay was characterized using peptide response curves, with linear ranges covering approximately four orders of magnitude and limits of detection in the low fmol/mg lysate range. Reproducibility was acceptable with median coefficients of variation of approximately 10%. We applied the assay to measurements of ER and HER2 in well‐characterized cell line lysates with good discernment based on ER/HER2 status. Finally, the proteins were measured in surgically resected breast cancers, and the results showed good correlation with ER/HER2 status determined by clinical assays. This is the first implementation of the peptide‐based immuno‐SRM assay technology in cell lysates and human surgical specimens.


Molecular & Cellular Proteomics | 2015

Peptide Immunoaffinity Enrichment and Targeted Mass Spectrometry Enables Multiplex, Quantitative Pharmacodynamic Studies of Phospho-Signaling

Jeffrey R. Whiteaker; Lei Zhao; Ping Yan; Richard G. Ivey; Uliana J. Voytovich; Heather D. Moore; Chenwei Lin; Amanda G. Paulovich

In most cell signaling experiments, analytes are measured one Western blot lane at a time in a semiquantitative and often poorly specific manner, limiting our understanding of network biology and hindering the translation of novel therapeutics and diagnostics. We show the feasibility of using multiplex immuno-MRM for phospho-pharmacodynamic measurements, establishing the potential for rapid and precise quantification of cell signaling networks. A 69-plex immuno-MRM assay targeting the DNA damage response network was developed and characterized by response curves and determinations of intra- and inter-assay repeatability. The linear range was ≥3 orders of magnitude, the median limit of quantification was 2.0 fmol/mg, the median intra-assay variability was 10% CV, and the median interassay variability was 16% CV. The assay was applied in proof-of-concept studies to immortalized and primary human cells and surgically excised cancer tissues to quantify exposure–response relationships and the effects of a genomic variant (ATM kinase mutation) or pharmacologic (kinase) inhibitor. The study shows the utility of multiplex immuno-MRM for simultaneous quantification of phosphorylated and nonmodified peptides, showing feasibility for development of targeted assay panels to cell signaling networks.


Molecular & Cellular Proteomics | 2016

Immobilized Metal Affinity Chromatography Coupled to Multiple Reaction Monitoring Enables Reproducible Quantification of Phospho-signaling

Jacob Kennedy; Ping Yan; Lei Zhao; Richard G. Ivey; Uliana J. Voytovich; Heather D. Moore; Chenwei Lin; Era L. Pogosova-Agadjanyan; Derek L. Stirewalt; Kerryn W. Reding; Jeffrey R. Whiteaker; Amanda G. Paulovich

A major goal in cell signaling research is the quantification of phosphorylation pharmacodynamics following perturbations. Traditional methods of studying cellular phospho-signaling measure one analyte at a time with poor standardization, rendering them inadequate for interrogating network biology and contributing to the irreproducibility of preclinical research. In this study, we test the feasibility of circumventing these issues by coupling immobilized metal affinity chromatography (IMAC)-based enrichment of phosphopeptides with targeted, multiple reaction monitoring (MRM) mass spectrometry to achieve precise, specific, standardized, multiplex quantification of phospho-signaling responses. A multiplex immobilized metal affinity chromatography- multiple reaction monitoring assay targeting phospho-analytes responsive to DNA damage was configured, analytically characterized, and deployed to generate phospho-pharmacodynamic curves from primary and immortalized human cells experiencing genotoxic stress. The multiplexed assays demonstrated linear ranges of ≥3 orders of magnitude, median lower limit of quantification of 0.64 fmol on column, median intra-assay variability of 9.3%, median inter-assay variability of 12.7%, and median total CV of 16.0%. The multiplex immobilized metal affinity chromatography- multiple reaction monitoring assay enabled robust quantification of 107 DNA damage-responsive phosphosites from human cells following DNA damage. The assays have been made publicly available as a resource to the community. The approach is generally applicable, enabling wide interrogation of signaling networks.


Molecular & Cellular Proteomics | 2015

Anti-Peptide Monoclonal Antibodies Generated for Immuno-Multiple Reaction Monitoring-Mass Spectrometry Assays Have a High Probability of Supporting Western blot and ELISA

Regine M. Schoenherr; Richard G. Saul; Jeffrey R. Whiteaker; Ping Yan; Gordon Whiteley; Amanda G. Paulovich

Immunoaffinity enrichment of peptides coupled to targeted, multiple reaction monitoring-mass spectrometry (immuno-MRM) has recently been developed for quantitative analysis of peptide and protein expression. As part of this technology, antibodies are generated to short, linear, tryptic peptides that are well-suited for detection by mass spectrometry. Despite its favorable analytical performance, a major obstacle to widespread adoption of immuno-MRM is a lack of validated affinity reagents because commercial antibody suppliers are reluctant to commit resources to producing anti-peptide antibodies for immuno-MRM while the market is much larger for conventional technologies, especially Western blotting and ELISA. Part of this reluctance has been the concern that affinity reagents generated to short, linear, tryptic peptide sequences may not perform well in traditional assays that detect full-length proteins. In this study, we test the feasibility and success rates of generating immuno-MRM monoclonal antibodies (mAbs) (targeting tryptic peptide antigens) that are also compatible with conventional, protein-based immuno-affinity technologies. We generated 40 novel, peptide immuno-MRM assays and determined that the cross-over success rates for using immuno-MRM monoclonals for Western blotting is 58% and for ELISA is 43%, which compare favorably to cross-over success rates amongst conventional immunoassay technologies. These success rates could most likely be increased if conventional and immuno-MRM antigen design strategies were combined, and we suggest a workflow for such a comprehensive approach. Additionally, the 40 novel immuno-MRM assays underwent fit-for-purpose analytical validation, and all mAbs and assays have been made available as a resource to the community via the Clinical Proteomic Tumor Analysis Consortiums (CPTAC) Antibody (http://antibodies.cancer.gov) and Assay Portals (http://assays.cancer.gov), respectively. This study also represents the first determination of the success rate (92%) for generating mAbs for immuno-MRM using a recombinant B cell cloning approach, which is considerably faster than the traditional hybridoma approach.

Collaboration


Dive into the Ping Yan's collaboration.

Top Co-Authors

Avatar

Amanda G. Paulovich

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jeffrey R. Whiteaker

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Chenwei Lin

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jacob Kennedy

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Lei Zhao

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Regine M. Schoenherr

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Pei Wang

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Richard G. Ivey

Fred Hutchinson Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Uliana J. Voytovich

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge