Network


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

Hotspot


Dive into the research topics where Petra Erdmann-Gilmore is active.

Publication


Featured researches published by Petra Erdmann-Gilmore.


Molecular & Cellular Proteomics | 2014

Ischemia in tumors induces early and sustained phosphorylation changes in stress kinase pathways but does not affect global protein levels

Philipp Mertins; Feng Yang; Tao Liu; D. R. Mani; Vladislav A. Petyuk; Michael A. Gillette; Karl R. Clauser; Jana W. Qiao; Marina A. Gritsenko; Ronald J. Moore; Douglas A. Levine; R. Reid Townsend; Petra Erdmann-Gilmore; Jacqueline Snider; Sherri R. Davies; Kelly V. Ruggles; David Fenyö; R. Thomas Kitchens; Shunqiang Li; Narcisco Olvera; Fanny Dao; Henry Rodriguez; Daniel W. Chan; Daniel C. Liebler; Forest M. White; Karin D. Rodland; Gordon B. Mills; Richard D. Smith; Amanda G. Paulovich; Matthew J. Ellis

Protein abundance and phosphorylation convey important information about pathway activity and molecular pathophysiology in diseases including cancer, providing biological insight, informing drug and diagnostic development, and guiding therapeutic intervention. Analyzed tissues are usually collected without tight regulation or documentation of ischemic time. To evaluate the impact of ischemia, we collected human ovarian tumor and breast cancer xenograft tissue without vascular interruption and performed quantitative proteomics and phosphoproteomics after defined ischemic intervals. Although the global expressed proteome and most of the >25,000 quantified phosphosites were unchanged after 60 min, rapid phosphorylation changes were observed in up to 24% of the phosphoproteome, representing activation of critical cancer pathways related to stress response, transcriptional regulation, and cell death. Both pan-tumor and tissue-specific changes were observed. The demonstrated impact of pre-analytical tissue ischemia on tumor biology mandates caution in interpreting stress-pathway activation in such samples and motivates reexamination of collection protocols for phosphoprotein analysis.


Blood | 2013

Genomic impact of transient low-dose decitabine treatment on primary AML cells.

Jeffery M. Klco; David H. Spencer; Tamara Lamprecht; Shawn M. Sarkaria; Todd Wylie; Vincent Magrini; Jasreet Hundal; Jason Walker; Nobish Varghese; Petra Erdmann-Gilmore; Cheryl F. Lichti; Matthew R. Meyer; R. Reid Townsend; Richard Wilson; Elaine R. Mardis; Timothy J. Ley

Acute myeloid leukemia (AML) is characterized by dysregulated gene expression and abnormal patterns of DNA methylation; the relationship between these events is unclear. Many AML patients are now being treated with hypomethylating agents, such as decitabine (DAC), although the mechanisms by which it induces remissions remain unknown. The goal of this study was to use a novel stromal coculture assay that can expand primary AML cells to identify the immediate changes induced by DAC with a dose (100nM) that decreases total 5-methylcytosine content and reactivates imprinted genes (without causing myeloid differentiation, which would confound downstream genomic analyses). Using array-based technologies, we found that DAC treatment caused global hypomethylation in all samples (with a preference for regions with higher levels of baseline methylation), yet there was limited correlation between changes in methylation and gene expression. Moreover, the patterns of methylation and gene expression across the samples were primarily determined by the intrinsic properties of the primary cells, rather than DAC treatment. Although DAC induces hypomethylation, we could not identify canonical target genes that are altered by DAC in primary AML cells, suggesting that the mechanism of action of DAC is more complex than previously recognized.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Hsp 70/Hsp 90 organizing protein as a nitrosylation target in cystic fibrosis therapy

Nadzeya V. Marozkina; Sean Yemen; Molly Borowitz; Lei Liu; Melissa Plapp; Fei Sun; Rafique Islam; Petra Erdmann-Gilmore; R. Reid Townsend; Cheryl F. Lichti; Sneha Mantri; Phillip W. Clapp; Scott H. Randell; Benjamin Gaston; Khalequz Zaman

The endogenous signaling molecule S-nitrosoglutathione (GSNO) and other S-nitrosylating agents can cause full maturation of the abnormal gene product ΔF508 cystic fibrosis (CF) transmembrane conductance regulator (CFTR). However, the molecular mechanism of action is not known. Here we show that Hsp70/Hsp90 organizing protein (Hop) is a critical target of GSNO, and its S-nitrosylation results in ΔF508 CFTR maturation and cell surface expression. S-nitrosylation by GSNO inhibited the association of Hop with CFTR in the endoplasmic reticulum. This effect was necessary and sufficient to mediate GSNO-induced cell-surface expression of ΔF508 CFTR. Hop knockdown using siRNA recapitulated the effect of GSNO on ΔF508 CFTR maturation and expression. Moreover, GSNO acted additively with decreased temperature, which promoted mutant CFTR maturation through a Hop-independent mechanism. We conclude that GSNO corrects ΔF508 CFTR trafficking by inhibiting Hop expression, and that combination therapies—using differing mechanisms of action—may have additive benefits in treating CF.


Molecular & Cellular Proteomics | 2016

An Analysis of the Sensitivity of Proteogenomic Mapping of Somatic Mutations and Novel Splicing Events in Cancer

Kelly V. Ruggles; Zuojian Tang; Xuya Wang; Himanshu Grover; Manor Askenazi; Jennifer Teubl; Song Cao; Michael D. McLellan; Karl R. Clauser; David L. Tabb; Philipp Mertins; Robbert J. C. Slebos; Petra Erdmann-Gilmore; Shunqiang Li; Harsha P. Gunawardena; Ling Xie; Tao Liu; Jian Ying Zhou; Shisheng Sun; Katherine A. Hoadley; Charles M. Perou; Xian Chen; Sherri R. Davies; Christopher A. Maher; Christopher R. Kinsinger; Karen D. Rodland; Hui Zhang; Zhen Zhang; Li Ding; R. Reid Townsend

Improvements in mass spectrometry (MS)-based peptide sequencing provide a new opportunity to determine whether polymorphisms, mutations, and splice variants identified in cancer cells are translated. Herein, we apply a proteogenomic data integration tool (QUILTS) to illustrate protein variant discovery using whole genome, whole transcriptome, and global proteome datasets generated from a pair of luminal and basal-like breast-cancer-patient-derived xenografts (PDX). The sensitivity of proteogenomic analysis for singe nucleotide variant (SNV) expression and novel splice junction (NSJ) detection was probed using multiple MS/MS sample process replicates defined here as an independent tandem MS experiment using identical sample material. Despite analysis of over 30 sample process replicates, only about 10% of SNVs (somatic and germline) detected by both DNA and RNA sequencing were observed as peptides. An even smaller proportion of peptides corresponding to NSJ observed by RNA sequencing were detected (<0.1%). Peptides mapping to DNA-detected SNVs without a detectable mRNA transcript were also observed, suggesting that transcriptome coverage was incomplete (∼80%). In contrast to germline variants, somatic variants were less likely to be detected at the peptide level in the basal-like tumor than in the luminal tumor, raising the possibility of differential translation or protein degradation effects. In conclusion, this large-scale proteogenomic integration allowed us to determine the degree to which mutations are translated and identify gaps in sequence coverage, thereby benchmarking current technology and progress toward whole cancer proteome and transcriptome analysis.


Molecular & Cellular Proteomics | 2016

Integrated Bottom-up and Top-down Proteomics of Patient-derived Breast Tumor Xenografts

Ioanna Ntai; Richard D. LeDuc; Ryan T. Fellers; Petra Erdmann-Gilmore; Sherri R. Davies; Jeanne M. Rumsey; Bryan P. Early; Paul M. Thomas; Shunqiang Li; Philip D. Compton; Matthew J. Ellis; Kelly V. Ruggles; David Fenyö; Emily S. Boja; Henry Rodriguez; R. Reid Townsend; Neil L. Kelleher

Bottom-up proteomics relies on the use of proteases and is the method of choice for identifying thousands of protein groups in complex samples. Top-down proteomics has been shown to be robust for direct analysis of small proteins and offers a solution to the “peptide-to-protein” inference problem inherent with bottom-up approaches. Here, we describe the first large-scale integration of genomic, bottom-up and top-down proteomic data for the comparative analysis of patient-derived mouse xenograft models of basal and luminal B human breast cancer, WHIM2 and WHIM16, respectively. Using these well-characterized xenograft models established by the National Cancer Institutes Clinical Proteomic Tumor Analysis Consortium, we compared and contrasted the performance of bottom-up and top-down proteomics to detect cancer-specific aberrations at the peptide and proteoform levels and to measure differential expression of proteins and proteoforms. Bottom-up proteomic analysis of the tumor xenografts detected almost 10 times as many coding nucleotide polymorphisms and peptides resulting from novel splice junctions than top-down. For proteins in the range of 0–30 kDa, where quantitation was performed using both approaches, bottom-up proteomics quantified 3,519 protein groups from 49,185 peptides, while top-down proteomics quantified 982 proteoforms mapping to 358 proteins. Examples of both concordant and discordant quantitation were found in a ∼60:40 ratio, providing a unique opportunity for top-down to fill in missing information. The two techniques showed complementary performance, with bottom-up yielding eight times more identifications of 0–30 kDa proteins in xenograft proteomes, but failing to detect differences in certain posttranslational modifications (PTMs), such as phosphorylation pattern changes of alpha-endosulfine. This work illustrates the potency of a combined bottom-up and top-down proteomics approach to deepen our knowledge of cancer biology, especially when genomic data are available.


Nature Communications | 2017

Proteogenomic integration reveals therapeutic targets in breast cancer xenografts.

Kuan-lin Huang; Shunqiang Li; Philipp Mertins; Song Cao; Harsha P. Gunawardena; Kelly V. Ruggles; D. R. Mani; Karl R. Clauser; Maki Tanioka; Jerry Usary; Shyam M. Kavuri; Ling Xie; Christopher Yoon; Jana W. Qiao; John A. Wrobel; Matthew A. Wyczalkowski; Petra Erdmann-Gilmore; Jacqueline Snider; Jeremy Hoog; Purba Singh; Beifang Niu; Zhanfang Guo; Sam Q. Sun; Souzan Sanati; Emily Kawaler; Xuya Wang; Adam Scott; Kai Ye; Michael D. McLellan; Michael C. Wendl

Recent advances in mass spectrometry (MS) have enabled extensive analysis of cancer proteomes. Here, we employed quantitative proteomics to profile protein expression across 24 breast cancer patient-derived xenograft (PDX) models. Integrated proteogenomic analysis shows positive correlation between expression measurements from transcriptomic and proteomic analyses; further, gene expression-based intrinsic subtypes are largely re-capitulated using non-stromal protein markers. Proteogenomic analysis also validates a number of predicted genomic targets in multiple receptor tyrosine kinases. However, several protein/phosphoprotein events such as overexpression of AKT proteins and ARAF, BRAF, HSP90AB1 phosphosites are not readily explainable by genomic analysis, suggesting that druggable translational and/or post-translational regulatory events may be uniquely diagnosed by MS. Drug treatment experiments targeting HER2 and components of the PI3K pathway supported proteogenomic response predictions in seven xenograft models. Our study demonstrates that MS-based proteomics can identify therapeutic targets and highlights the potential of PDX drug response evaluation to annotate MS-based pathway activities.


Molecular & Cellular Proteomics | 2015

An Integrated Multiomics Approach to Identify Candidate Antigens for Serodiagnosis of Human Onchocerciasis

Samantha N. McNulty; Bruce A. Rosa; Peter U. Fischer; Jeanne M. Rumsey; Petra Erdmann-Gilmore; Kurt C. Curtis; Sabine Specht; R. Reid Townsend; Gary J. Weil; Makedonka Mitreva

Improved diagnostic methods are needed to support ongoing efforts to eliminate onchocerciasis (river blindness). This study used an integrated approach to identify adult female Onchocerca volvulus antigens that can be explored for developing serodiagnostic tests. The first step was to develop a detailed multi-omics database of all O. volvulus proteins deduced from the genome, gene transcription data for different stages of the parasite including eight individual female worms (providing gene expression information for 94.8% of all protein coding genes), and the adult female worm proteome (detecting 2126 proteins). Next, female worm proteins were purified with IgG antibodies from onchocerciasis patients and identified using LC-MS with a high-resolution hybrid quadrupole-time-of-flight mass spectrometer. A total of 241 immunoreactive proteins were identified among those bound by IgG from infected individuals but not IgG from uninfected controls. These included most of the major diagnostic antigens described over the past 25 years plus many new candidates. Proteins of interest were prioritized for further study based on a lack of conservation with orthologs in the human host and other helminthes, their expression pattern across the life cycle, and their consistent expression among individual female worms. Based on these criteria, we selected 33 proteins that should be carried forward for testing as serodiagnostic antigens to supplement existing diagnostic tools. These candidates, together with the extensive pan-omics dataset generated in this study are available to the community (http://nematode.net) to facilitate basic and translational research on onchocerciasis.


Science Signaling | 2017

Breast tumors educate the proteome of stromal tissue in an individualized but coordinated manner

Xuya Wang; Arshag D. Mooradian; Petra Erdmann-Gilmore; Qiang Zhang; Rosa Viner; Sherri R. Davies; Kuan-lin Huang; Ryan Bomgarden; Brian A. Van Tine; Jieya Shao; Li Ding; Shunqiang Li; Matthew J. Ellis; John C. Rogers; R. Reid Townsend; David Fenyö; Jason M. Held

Proteomic analysis of the tumor-associated stroma reveals extensive and coordinated regulation by breast cancers. Profiling the tumor stroma proteome Communication between a tumor and cells in the surrounding stroma contributes to tumor growth, progression, and drug resistance. Thus, targeting this communication, in the primary tumor and especially in metastatic niches, may be an effective way to treat cancer. Wang et al. grew patient breast tumors subcutaneously in mice and obtained species-distinguished proteomic profiles of the tumors (human) and tumor-associated stroma (mouse). The authors found that all breast tumors consistently altered clustered subsets of the stromal proteome, particularly proteins involved in immune signaling, but that these varied in a subtype- and stage-specific manner. These findings may have future implications for treatment stratification and provide a platform from which to understand this experimental model and tumor-stroma interactions on a large-scale protein level. Cancer forms specialized microenvironmental niches that promote local invasion and colonization. Engrafted patient-derived xenografts (PDXs) locally invade and colonize naïve stroma in mice while enabling unambiguous molecular discrimination of human proteins in the tumor from mouse proteins in the microenvironment. To characterize how patient breast tumors form a niche and educate naïve stroma, subcutaneous breast cancer PDXs were globally profiled by species-specific quantitative proteomics. Regulation of PDX stromal proteins by breast tumors was extensive, with 35% of the stromal proteome altered by tumors consistently across different animals and passages. Differentially regulated proteins in the stroma clustered into six signatures, which included both known and previously unappreciated contributors to tumor invasion and colonization. Stromal proteomes were coordinately regulated; however, the sets of proteins altered by each tumor were highly distinct. Integrated analysis of tumor and stromal proteins, a comparison made possible in these xenograft models, indicated that the known hallmarks of cancer contribute pleiotropically to establishing and maintaining the microenvironmental niche of the tumor. Education of the stroma by the tumor is therefore an intrinsic property of breast tumors that is highly individualized, yet proceeds by consistent, nonrandom, and defined tumor-promoting molecular alterations.


Cancer Research | 2018

Mass spectrometry-based proteomics reveals potential roles of NEK9 and MAP2K4 in resistance to PI3K inhibitors in triple negative breast cancers

Filip Mundt; Sandeep Rajput; Shunqiang Li; Kelly V. Ruggles; Arshag D. Mooradian; Philipp Mertins; Michael A. Gillette; Karsten Krug; Zhanfang Guo; Jeremy Hoog; Petra Erdmann-Gilmore; Tina Primeau; Shixia Huang; Dean P. Edwards; Xiaowei Wang; Xuya Wang; Emily Kawaler; D. R. Mani; Karl R. Clauser; Feng Gao; Jingqin Luo; Sherri R. Davies; Gary L. Johnson; Kuan-lin Huang; Christopher Yoon; Li Ding; David Fenyö; Matthew J. Ellis; R. Reid Townsend; Jason M. Held

Activation of PI3K signaling is frequently observed in triple-negative breast cancer (TNBC), yet PI3K inhibitors have shown limited clinical activity. To investigate intrinsic and adaptive mechanisms of resistance, we analyzed a panel of patient-derived xenograft models of TNBC with varying responsiveness to buparlisib, a pan-PI3K inhibitor. In a subset of patient-derived xenografts, resistance was associated with incomplete inhibition of PI3K signaling and upregulated MAPK/MEK signaling in response to buparlisib. Outlier phosphoproteome and kinome analyses identified novel candidates functionally important to buparlisib resistance, including NEK9 and MAP2K4. Knockdown of NEK9 or MAP2K4 reduced both baseline and feedback MAPK/MEK signaling and showed synthetic lethality with buparlisib in vitro A complex in/del frameshift in PIK3CA decreased sensitivity to buparlisib via NEK9/MAP2K4-dependent mechanisms. In summary, our study supports a role for NEK9 and MAP2K4 in mediating buparlisib resistance and demonstrates the value of unbiased omic analyses in uncovering resistance mechanisms to targeted therapy.Significance: Integrative phosphoproteogenomic analysis is used to determine intrinsic resistance mechanisms of triple-negative breast tumors to PI3K inhibition. Cancer Res; 78(10); 2732-46. ©2018 AACR.


Journal of Proteome Research | 2017

Quality Assessments of Long-Term Quantitative Proteomic Analysis of Breast Cancer Xenograft Tissues

Jianying Zhou; Lijun Chen; Bai Zhang; Yuan Tian; Tao Liu; Stefani N. Thomas; Li Chen; Michael Schnaubelt; Emily S. Boja; Tara Hiltke; Christopher R. Kinsinger; Henry Rodriguez; Sherri R. Davies; Shunqiang Li; Jacqueline Snider; Petra Erdmann-Gilmore; David L. Tabb; R. Reid Townsend; Matthew J. Ellis; Karin D. Rodland; Richard D. Smith; Steven A. Carr; Zhen Zhang; Daniel W. Chan; Hui Zhang

Clinical proteomics requires large-scale analysis of human specimens to achieve statistical significance. We evaluated the long-term reproducibility of an iTRAQ (isobaric tags for relative and absolute quantification)-based quantitative proteomics strategy using one channel for reference across all samples in different iTRAQ sets. A total of 148 liquid chromatography tandem mass spectrometric (LC-MS/MS) analyses were completed, generating six 2D LC-MS/MS data sets for human-in-mouse breast cancer xenograft tissues representative of basal and luminal subtypes. Such large-scale studies require the implementation of robust metrics to assess the contributions of technical and biological variability in the qualitative and quantitative data. Accordingly, we derived a quantification confidence score based on the quality of each peptide-spectrum match to remove quantification outliers from each analysis. After combining confidence score filtering and statistical analysis, reproducible protein identification and quantitative results were achieved from LC-MS/MS data sets collected over a 7-month period. This study provides the first quality assessment on long-term stability and technical considerations for study design of a large-scale clinical proteomics project.

Collaboration


Dive into the Petra Erdmann-Gilmore's collaboration.

Top Co-Authors

Avatar

R. Reid Townsend

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Shunqiang Li

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Sherri R. Davies

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matthew J. Ellis

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kuan-lin Huang

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Li Ding

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge