7:30 AM - 7:00 PM - Registration
|
|
Registration and Information Desk
|
8:30 AM - 9:05 AM - Plenary Session
|
|
Donald F. Hunt Distinguished Contribution in Proteomics Award Plenary Session
Sponsored By: JPR
Presented By:
|
Joshua Coon
(Bio)
Josh Coon grew up in rural Michigan, where he enjoyed fly fishing and woodworking, even building several riverboats during high school and college. His interest in Analytical Chemistry stemmed from a love of building, not boats, but chemical instrumentation. To escape the cold, he joined the Chemistry graduate program at the University of Florida. After graduating in 2002, he moved to Charlottesville, Virginia, to join Professor Don Hunt's lab, where he co-invented electron transfer dissociation (ETD).
In 2005, Coon moved to Wisconsin as an Assistant Professor. He currently holds the Thomas and Margaret Pyle Chair at the Morgridge Institute for Research and is a Professor of Chemistry and Biomolecular Chemistry at the University of Wisconsin-Madison. His program specializes in developing and applying novel chemical instrumentation and molecular analysis methodologies. The team uses these technologies for studies ranging from basic biochemical questions in model organisms to translational work in human subjects. To date, he has published nearly 400 peer-reviewed manuscripts, which have collectively received over 35,000 citations. His work has been recognized with numerous awards, including the Distinguished Achievement in Proteomic Sciences Award (Human Proteome Organization), the H.I. Romnes Faculty Fellowship (UW-Madison), the Biemann Medal (American Society for Mass Spectrometry), the Pittsburgh Conference Achievement Award (Pittcon Society), the Ken Standing Award (University of Manitoba), and the ACS Chemical Instrumentation Award, among many others. Coon has mentored over 40 Ph.D. graduates and around 15 postdoctoral scholars, many of whom are now faculty members at top academic institutions or leaders in industry.
|
|
9:15 AM - 10:35 AM - Parallel Sessions
|
|
Parallel Session 01: New Discoveries in Molecular Metabolism
Use of integrated omic methods and in vivo imaging to understand molecular mechanisms underlying metabolic health
In utero exposures during fetal development lead to developmental programming (DP) of offspring with increased risk of cardiometabolic disease. Maternal overnutrition (MON) during gestation is a common adverse exposure that programs offspring metabolically, and disproportionally affects individuals from disadvantaged communities. The long-term impact of MON on offspring are important to public health, but difficult to determine in humans due to confounding by socioeconomic factors such as access to health care and healthy foods. Skeletal muscle (SKM) comprises ~40% of total body mass, is a major metabolic tissue, and important for quality of life. Most prior studies to identify mechanisms underlying DP by MON in adult offspring have been conducted on rodents which limits our understanding of relevant processes and potential mechanisms in humans. Nonhuman primates (NHP) complement rodent studies for translation to understand human aging-related diseases. In this study we compared young adult MON baboon offspring born to mothers fed an obesogenic diet prior to and during pregnancy and lactation with age-matched controls (CON) to identify early SKM molecular and metabolic changes. We generated untargeted integrated-omics data (transcriptomic, proteomic, metabolomic) from SKM biopsies, imaging data from SKM in vivo magnetic resonance spectroscopy (MRS), and clinical blood measures of metabolic status, and used unbiased clustering analyses to identify modules of molecules that correlate with in vivo and clinical metabolic measures that differ between MON and CON. We found that circulating lipoproteins positively correlated with SKM metabolism in CON, but were negatively correlated in MON young adults. These results indicate that standard clinical measures of metabolic status differ in adult offspring of suboptimal pregnancies compared with healthy pregnancies, and highlight the need to develop relevant clinical measures for offspring of suboptimal in utero environments. Our results also show the power of integrating multi-omics with in vivo imaging to better understand metabolic health/disease.
|
|
Laura Cox, Professor, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
(Bio)
The focus of my research is understanding the impact of maternal under-nutrition and maternal obesity during pregnancy on offspring cardiovascular health and aging using genomic and other omic methods. In recent work, my research group is using integrated "omic" approaches to better understand molecular networks underlying cardiovascular health and to identify molecules dysregulated in these networks prior to onset of clinical measures indicative of cardiovascular disease.
|
OA01.01 | Single-Organoid Proteomics Defines Molecular Features at the Intersection of Cancer, Cell Bioenergetics, and Virus Infection
Historically, the study of many cancer types has been limited by an absence of experimental models that accurately exhibit characteristics of tumorigenesis in patients. As self-organizing systems that recapitulate the structural complexity of human tissues, three-dimensional (3D) organoid models provide new opportunities to determine how tumors spatially and temporally reprogram inter- and intra-cellular signaling. Understanding dysregulated signaling cascades is further fundamental to deciphering how tumor microenvironments are impacted by host-pathogen interactions during oncomodulatory viral infections. Defining these altered features is relevant for cancers lacking effective standard-of-care treatments, as there are pressing needs to identify molecular dependencies for targeting in new treatment strategies. We harnessed organoids to investigate cellular remodeling events at the interface of cancer, metabolism, and virus infection. We investigated two cancer organoid models, high-grade serous tubo-ovarian cancer and patient-derived colorectal cancer. We applied a multidisciplinary approach that integrated quantitative single-organoid proteomics, global protein-protein interaction characterizations by thermal proximity co-aggregation profiling (TPCA-MS), live metabolic flux assays, live super-resolution microscopy, and virology. In ovarian cancer organoids, we uncovered functional enrichments in mitochondria-associated metabolism, which were exacerbated in 3D organoids relative to their 2D monolayer counterparts. Follow-up targeted proteomics by parallel reaction monitoring revealed abundance changes in proteins mediating organelle-organelle contacts (e.g., mitochondria and endoplasmic reticulum, ER). TPCA-MS uncovered altered thermal stabilities of protein complexes that drive mitochondrial metabolism (e.g., F1F0-ATP synthase). Live metabolic flux assays and microscopy identified that the most aggressive cancer subtype exhibited higher degrees of mitochondria-ER encapsulations, coincident with elevated calcium signaling, lipid droplet clustering, and bioenergetic respiration through oxidative phosphorylation. Infections with oncomodulatory and metabolically-active cytomegalovirus revealed significant differences in viral susceptibility, suggesting that heightened tumor metabolism confers a viral replication advantage. Our findings explain how cancer types harness the metabolic capacities of remodeled organelles to promote tumorigenesis, which alters infection outcomes within the tumor microenvironment.
|
|
Krystal Lum, Princeton University, Princeton, NJ, United States
|
OA01.02 | Multi-omic Analysis Identifies Modulations in Amino Acid and Fatty Acid Metabolism Associated with Cardiovascular Dysfunction in RAB27a Mutant Mice
In humans, mutations in the small GTPase RAB27a are linked to Griscelli syndrome, a rare and fatal autosomal dominant disease associated with hypopigmentation, immune system abnormalities, and cardiac deficits. RAB27a is involved in protein and vesicle trafficking and secretion of exosomes, placing it at the center of cell-cell communication. Although not produced in cardiac tissue in mice, RAB27a is expressed within local perivascular adipose tissue (PVAT), and we hypothesized that global loss of RAB27a would impact homeostatic signaling from PVAT to the heart and vasculature. We created Rab27a global null mice and found an age-related cardiomyopathy phenotype including altered vasocontractile responses, decreased left ventricular ejection fraction, and changes in PVAT and aorta proteomic signatures. To uncover the mechanism behind this phenotype we performed large-scale untargeted metabolomics and proteomics analysis, and integrated the datasets, across blood plasma, adipose tissue, and cardiac tissue from the Rab27a null mouse model. Unbiased proteomics on plasma was performed with the Seer enrichment platform couple to the Orbitrap Astral Mass Spectrometry platform for maximum coverage. We identified 54,020 peptides corresponding to 5,003 protein groups. Unbiased metabolomics on plasma and tissues found 1923 lipid and polar metabolites (1505 identified). In perivascular adipose tissue 1623 metabolites were found (992 identified), while in perivascular aorta tissue 915 metabolites were found (535 identified). Following multi-omic integration of these datasets, pathway analysis was performed uncovering that ablation of Rab27adownregulated amino acid and fatty acid metabolism in plasma as well as PVAT and cardiac tissues. Specifically, energy metabolism as well as plasma membrane microdomain lipids are affected by Rab27a loss, which may help explain the cardiovascular phenotype observed in the mouse model. Ongoing work is leveraging validation and flux workflows to tie loss of Rab27a to these specific functions.
|
|
Calvin Vary, MaineHealth Institute for Research, Scarborough, ME, United States
|
OA01.03 | An atlas of the zinc binding human cysteine proteome identifies glutathione reductase (GSR) as zinc-targetable cancer vulnerability
Zinc is an essential micronutrient that regulates a wide range of physiological processes, principally through Zn2+ binding to protein cysteine residues. Despite being critical for modulation of protein function, for the vast majority of the human proteome, the cysteine sites subject to Zn2+ binding remain undefined. To address this, we developed ZnCPT, a deep and quantitative mapping of the zinc binding cysteine proteome that quantifies the zinc binding status across over 58,000 cysteine sites. We define 6173 zinc binding protein cysteines, uncovering protein families across major domains of biology that are subject to either constitutive or inducible zinc binding. The systematic structural analysis of zinc binding site environments identified distinct features providing a structural basis for constitutive and inducible zinc coordination, including physicochemical determinants. Cross-referencing ZnCPT with published redox proteomes revealed that most cysteines belong to distinct subpopulations, either redox regulated or zinc binding, which substantially enhances our understanding of cysteine functionality. ZnCPT further enables systematic discovery of zinc-regulated structural, enzymatic, and allosteric functional domains. On this basis, we identify 52 cancer genetic dependencies subject to zinc binding and nominate malignancies sensitive to zinc-induced cytotoxicity. In doing so, we discover a mechanism of zinc regulation over Glutathione Reductase (GSR) that drives cell death in GSR-dependent lung cells and tumors in vivo. Mechanistic analyses revealed zinc-mediated inhibition of GSR by binding to its active site, which leads to disruption of cellular redox homeostasis and pronounced cytotoxicity that can be alleviated through pharmacological or genetic elevation of the antioxidant thiol pool. Importantly, zinc supplementation significantly reduced the growth of GSR-dependent lung cancer cells in a murine tumor xenograft model. We provide ZnCPT as a resource for understanding mechanisms of zinc regulation of protein function.
|
|
Nils Burger, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, United States
|
|
|
Parallel Session 02: (Pre-)clinical Proteomics - From Pre-Phase 1 to Phase 3
Functional proteomics for clinical insight in TNBC and GBM
Functional proteomics can provide unprecedented insight into biological regulation in the context of health and disease. To identify potential therapeutic targets and therapeutic strategies for tumors with few treatment options, we applied functional proteomics with multiplexed isobaric tags (TMT) for quantification to gain insight into activated signaling networks. Application of this approach to triple negative breast cancer (TNBC) patient-derived xenograft tumors resulted in the identification of Src-family kinase signaling networks as potential targets for selected tumors. Extending these analyses to 150 human patient tumor specimens, we were able to identify activated networks in each patient tumor and to stratify patients into different categories based on their respective signaling networks and putative therapeutic targets. To enable reproducible quantification of low-abundance phosphorylation sites we developed SureQuant pTyr, a method enabling the targeted quantification of ~400 pTyr sites. SureQuant pTyr provides highly accurate and reproducible quantification that should be amenable to clinical proteomics.
|
|
Forest White
(Bio)
Forest White is a Professor in the Department of Biological Engineering at the Massachusetts Institute of Technology (MIT). He received Ph.D. from Florida State University, completed a post-doc at the University of Virginia, and then joined MDS Proteomics where he developed phosphoproteomics capabilities in the company. In 2003 he joined the Department of Biological Engineering at MIT. Research in his lab is focused on quantification of protein phosphorylation-mediated signaling networks and MHC peptide presentation in normal and pathophysiological conditions. Applications include novel drug target discovery in glioblastoma, melanoma, and triple negative breast cancer, as well as analysis of mechanisms underlying therapeutic resistance and metastasis. Forest is a member of the Koch Institute for Integrative Cancer Research at MIT.
|
OA02.01 | Integrated Multi-Omics and Clinical Covariate Analysis Uncover Abnormal Adaptive Immune Responses in Human Adult Congenital Heart Valve Disease
PURPOSE: Calcific aortic valve disease (CAVD) occurs in two distinct anatomies of heart valve leaflets: tricuspid aortic valve (TAV) and bicuspid (BAV). In BAV patients, two of three valve leaflets are fused during embryonic development, leading to rapid disease progression with increased fibrosis and calcification. The pathogenic mechanisms underlying accelerated CAVD in BAVs remain unknown. Fibrocalcific-burdened tissue is i) difficult for proteomics to access, and ii) suffers from reduced correlation between RNA and protein levels. METHODS: Clinical human aortic valve tissue samples (TAV and BAV; n=83 donors) were analyzed by bulk multi-omics (mass spectrometric proteomics, miRNA-seq, RNA-seq) and single-nuclei RNA-seq (snRNA-seq). TargetScan identified microRNA regulatory gene/protein targets. Patient characteristics and clinical imaging data were integrated with multi-omics datasets by latent factor-based approaches to identify unique molecular mechanisms of CAVD across TAV vs. BAV. RESULTS: Mass spectrometric proteomics coupled to pathway- and protein-protein interaction networks revealed significantly elevated platelet activation in BAV tissues and implicated altered developmental SLIT-ROBO signaling proteins in adult BAV-CAVD progression. Orthogonally, snRNA-seq of CAVD tissues identified 7 novel subpopulations of fibroblastic valve interstitial cells (65% of total cells) with distinct myofibrogenic (ACTA2/CARMN/MYH11-high) vs. osteogenic (RUNX2/CDH11/POSTN-high) lineages. snRNA-seq also revealed that transcriptional and cellular heterogeneity in BAV-CAVD was driven by significant accumulation of specific macrophage and B-cell states (p CONCLUSION: Multi-omics integration identifies aberrant adaptive immune responses as critical drivers of BAV-CAVD. Our findings delineate a novel strategy for therapeutic discovery in congenital heart disease and fibrocalcific tissues in general, with forthcoming target validation studies conducted in TAV- vs. BAV-derived primary human valve cell cohorts.
|
|
Mark Blaser, Brigham and Women's Hospital, Boston, MA, United States
|
OA02.02 | Using machine learning to integrate unbiased proteomics and functional testing to guide an interventional umbrella clinical trial
Acute myeloid leukemia (AML) is an aggressive blood cancer with a 26% 5-year survival rate. Current therapies, even when combined with newer agents, have not significantly enhanced long-term survival for many AML patients, and relapse remains a major problem. One major barrier in the development of effective therapies for AML has been the lack of accurate model systems to identify potentially useful novel agents and then predict therapeutic efficacy in molecularly defined subtypes of patients with AML. Current AML cell lines often harbor strong driver alterations that occur in only a small subpopulation of patients and therefore may not accurately predict agent sensitivity for most patients. With state-of-the-art culture conditions, we have raised over 50 patient-derived AML cell models and 19 American Type Culture Collection (ATCC) cell lines to measure cellular response across 40 AML therapeutics. Leveraging our optimized pipeline for LC-MS/MS and innovative data integration pipeline, we have created a patient knowledge landscape with embedded physical models for prospective therapeutic positioning. These new biomaterials offer immense benefits in the pursuit of understanding the diversity of AML, producing both rationally designed therapeutics and unbiased compound screens. Unbiased proteomics data was generated using peripheral blood samples from 100 AML patients and all of the ex-vivo models to robustly quantify more than 8,000 proteins per sample. Preliminary results using proteomics and cell surface markers show little drift from the original patient samples. Additionally, we have developed a custom deep learning architecture to reveal subsets of patients that have similar proteomic and compound sensitivity profiles and position these subsets to their nearest proteomic-matched ex vivo model. To drive a prospective clinical trial at the University of Washington/Fred Hutchinson Cancer Center, our model was used to select a minimal set of proteins that can accurately separate patient subsets for which there are differential sensitivity profiles across 4 AML therapeutics.
|
|
James Sorrentino, Yatiri Bio, San Diego, CA, United States
|
OA02.03 | Innovative Proteomic Approaches in Translational Medicine: Uncovering New Biomarkers for Therapeutic Development In Inflammatory Bowel Disease
Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn's disease (CD), is characterized by chronic gastrointestinal inflammation. Novel biomarkers are critical for monitoring disease activity, guiding treatment decisions, and improving patient outcomes. Biomarkers are indispensable in drug development, providing insights into target engagement, dose selection, and therapeutic mechanisms. A previous study by our group used data-independent acquisition LC-MS/MS (DIA-MS) to profile the human fecal proteome, identifying over 600 human proteins and approximately 100 differentially abundant proteins in IBD stool compared to healthy stool. Many of these proteins are associated with immune cells and ulceration, hallmarks of IBD pathobiology, and revealed new fecal proteins from multiple dysregulated pathways in IBD. To further investigate the biological mechanisms underlying IBD, this method was optimized using the EvoSep One System combined with a Thermo Fisher Orbitrap Astral Mass Spectrometer. Preliminary results indicate over 2000 human fecal proteins are now measurable using a 35-minute gradient, significantly enhancing the depth of proteome coverage while reducing analysis time. This approach effectively captures the dynamic range of proteins in fecal samples and this has been applied to a large clinical cohort of IBD patient samples to identify potential biomarkers correlating with treatment response and durability, as well as biomarkers that can inform on pathway modulation for early clinical candidates. By advancing the understanding of the fecal proteome in IBD through large-scale DIA-MS, this study aims to uncover novel biomarkers that can drive more effective drug development and ultimately improve patient outcomes in this disease.
|
|
Veronica Anania
|
|
10:35 AM - 11:00 AM - Break
|
|
Coffee Break
|
11:00 AM - 12:20 PM - Parallel Sessions
|
|
Parallel Session 03: Neuroproteomics
Sponsored By: New Objective
|
|
Tara Tracy, Assistant Professor, Buck Institute for Research on Aging, Novato, CA, United States
(Bio)
Dr. Tracy received her PhD in Neuroscience from the University of California, Berkeley where she studied synapse development. During her postdoctoral training at the Gladstone Institute of Neurological Disease and the University of California, San Francisco, Dr. Tracy investigated the toxic mechanisms that drive neuron dysfunction and cognitive decline in Alzheimer's disease. Dr. Tracy's laboratory at the Buck Institute is investigating the synapse dysfunction in the brain that causes cognitive decline in aging and in neurodegenerative diseases. Research from the Tracy laboratory has uncovered a synapse repair mechanism that promotes resilience to tauopathy-related memory loss. In 2022, Dr. Tracy was awarded the McKnight Brain Research Foundation Innovator Award in Cognitive Aging & Memory Loss from the American Federation of Aging Research.
|
OA03.01 | Highly Multiplexed Targeted Assay for Alzheimer's Disease and Related Dementias in Mag-Net Enriched Plasma
Neurodegenerative diseases with different etiologies can have overlap in clinical presentation, especially with cognitive impairment and dementia. This makes it challenging to make definitive diagnoses prior to post-mortem assessment, often requiring significant additional neuroimaging and clinical follow-up. Plasma is a preferred source for biomarker proteins due to its routine collection in clinical care. Previously we found a subset of proteins in Mag-Net enriched plasma that can distinguish individuals with Alzheimer’s disease dementia from those with Parkinson’s disease dementia, Parkinson’s disease cognitively normal, and healthy cognitively normal. Here we validate a subset of those proteins through a highly multiplex parallel reaction monitoring (PRM) assay on a linear ion trap instrument. Plasma from a small neurodegenerative disease sample set were enriched and digested using the Mag-Net method. A sample pool was acquired by 1 m/z window gas-phase fractionated (GPF) data-independent acquisition (DIA) on a Stellar MS. Using information from the Stellar GPF-DIA, proteins from a classifier built from a previous discovery experiment were selected for a preliminary PRM assay targeting 1006 precursors from 144 proteins. Overall the PRM assay is highly reproducible, with inter-QC sample correlation of >0.99. Additionally, each precursor is assessed by matched-matrix calibration curve figures of merit. The PRM assay was applied to a small validation set of 40 individuals with Alzheimer’s, Parkinson’s, or cognitively normal. We used the Boruta Package for feature selection to filter the 144 quantitative protein features further to a set of 21. These 21 proteins from the 144 preliminary proteins classify Alzheimer’s with a mean ROC AUC of 0.88. The protein features include proteins known to be involved in the biological processes or pathogenesis of AD or Lewy Body Disease. Further validation is planned with a large plasma cohort with Lewy body disease and Alzheimer’s disease dementia.
|
|
Deanna Plubell, University of Washington, Seattle, WA, United States
|
OA03.02 | Using Cross-linking MS to Investigate the Distribution of Long-lived Proteins Within Mitochondrial Protein Complexes in Mouse Brain
Continuous replenishment of mitochondrial proteins has long been considered essential for maintaining high-quality organelles. In agreement, the average half-life of mitochondrial proteins in the mouse brain has been estimated at less than three weeks. Recently, however, we uncovered that a discrete subset of the mitochondrial brain proteome exhibits lifespans of four months. These long-lived mitochondrial proteins, or mt-LLPs, are enriched at cristae invaginations and include OxPhos components, MICOS, mt-DNA proteins, chaperones, and cytochrome C. While the pathways involved in mitochondrial protein degradation have been studied, little is known about how the mitochondrial proteome is replenished post-removal and how newly synthesized proteins are integrated with existing protein pools. This is especially important for multiprotein OxPhos complexes, which, in addition to being long-lived, are assembled from proteins encoded by both the nuclear and mitochondrial genomes. To gain a deeper understanding of how new proteins are integrated into existing mitochondrial networks, we combined an in vivo whole rodent metabolic stable isotope pulse-chase labeling method with cross-linking mass spectrometry (XL-MS) on intact immuno-isolated mitochondria from brain extracts. Using this method, we were able to examine protein-protein interactions between the old (long-lived) proteins and newly synthesized copies, as well as how they interact over time. Our analysis revealed that once assembled, mitochondrial OxPhos complexes are preserved with limited subunit exchange throughout their lifetime and are spatially restricted within the same mitochondrial cristae. Since cristae stability is intimately linked to mitochondrial function, we propose that mt-LLPs could play a previously unrecognized role in the long-term stabilization of mitochondrial ultrastructure, which, in turn, might be imperative for mitochondrial fitness. The combination of pulse-chase protein labeling with cross-linking mass spectrometry developed here allows for the interrogation of protein-protein interactions in both space and time, thus enabling both structural and temporal insights into complex assembly pathways.
|
|
Ewa Bomba-Warczak, University of Pennsylvania, Philadelphia, PA, United States
|
OA03.03 | Multi-epitope Immunocapture of Huntingtin Reveals Striatum-selective Molecular Signatures
Huntington’s disease (HD) is a debilitating neurodegenerative disorder affecting an individual’s cognitive and motor abilities. HD is caused by mutation in the huntingtin gene producing a toxic polyglutamine-expanded protein (mHTT) and leading to degeneration in the striatum and cortex. Yet, the molecular signatures that underlie tissue-specific vulnerabilities remain unclear. Here, we investigate this aspect by leveraging multi-epitope protein interaction assays, subcellular fractionation, thermal proteome profiling, and genetic modifier assays. Use of human cell, mouse, and fly models afforded capture of distinct subcellular pools of epitope-enriched and tissue-dependent interactions linked to dysregulated cellular pathways and disease relevance. First, we characterized the cell-wide impact of HTT loss or polyglutamine expansion in an HD model of liver pathobiology using thermal proximity coaggregation assay mass spectrometry (TPCA-MS) to capture global protein interaction networks and dynamics. Our results point to divergent alterations in the assembly of critical protein complexes in KO versus mHTT, including chromatin remodeling and mitochondrial complexes. To place these findings in the broader context of HD pathobiology, we characterized determinants of striatal vulnerability in the brain by defining HTT protein interactions at different stages of disease progression. We established an HTT association with nearly all subunits of the transcriptional regulatory Mediator complex (20/26), with preferential enrichment of MED15 in the tail domain. Using HD and KO models, we find HTT modulates the subcellular localization and assembly of Mediator. We demonstrated striatal enriched and functional interactions with regulators of calcium homeostasis and chromatin remodeling and established their disease relevance using HD fly genetic modifiers assays. Lastly, we implement our previously developed web-based HD protein network and multi-omics platform, HTT-OMNI, to catalyze interrogation of these tissue-dependent and epitope-enriched HTT protein interactions and facilitate multi-omic analyses within the HD community. Altogether, we offer insights into tissue- and localization-dependent (m)HTT functions and pathobiology.
|
|
Todd Greco, Princeton University, Princeton, NJ, United States
|
|
|
Parallel Session 04: Proteomics Meets Pharma: Compound, Phenotypic and MoA Screens
Adventures in Chemoproteomic Profiling of Small Molecules
|
|
Jarrod Marto, Principal Investigator and Professor, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States
(Bio)
Jarrod Marto, Ph.D., is a Principal Investigator at the Dana-Farber Cancer Institute in the Department of Cancer Biology and an Associate Professor of Pathology at Brigham and Women's Hospital and Harvard Medical School. Since 2006 Dr. Marto has served as Director of the Blais Proteomics Center at Dana-Farber and more recently launched the Center for Emergent Drug Targets. Dr. Marto's research is focused on the development and use of state-of-the-art mass spectrometry and other bioanalytical techniques to understand how genomic alterations as well as the activity of chemical probes or clinical drugs manifest at the level of individual proteins, signaling pathways, or other compartments throughout the functional proteome.
Dr. Marto has authored 200 peer-reviewed papers across the fields of bioanalytical chemistry, scientific instrumentation, mass-informatics, chemical biology, and cancer cell signaling. In addition, he is a founding member of Entact Bio and serves on the SAB of 908 Devices.
|
OA04.01 | Development of a Multi-omics Platform to Generate Cellular Phenotypic Compound 'Signatures'
In Discovery, demonstrating translational biology for small molecules is often a bottleneck and as we become more efficient at identifying chemical starting points, understanding biological mechanism of action for these molecules is increasingly becoming the rate limiting step in compound progression. To overcome this hurdle, a suite of existing and newly developed high content experimental platforms and data pipelines, High Content Profiling platforms, including Cell Paint Imaging, Transcriptomics, Proteomics and Metabolomics, have been applied to multiple projects to facilitate small molecule screening and selection. Here, we present the application of multi-omics strategy to generate compound ‘Signatures’ in the cellular systems. These can be compared to genomic signatures, or signatures derived from tools molecules, to help define MOA and ensure translatability of the signatures. We applied a multi-omics strategy to characterize compounds that inhibit a synthetic lethal target that contributes to kinetochore-microtubule attachment and mitotic spindle assembly checkpoint fidelity. Program compounds along with well characterized tools were used to treat both sensitive and insensitive cell lines. Each compound was dosed in triplicate, at 10 µM for 24 hours. Vehicle treated cells acted as a reference for differential expression analysis as well the origin for scalar projection analysis. Data processing pipelines were built in-house, encompassing feature extraction, identification, alignment, normalization, and statistical analysis. Strong positive correlation was observed across the platforms. A subset of compounds showed robust cellular activity with the desired phenotype in the sensitive cell line but not in the insensitive cell line as expected. Of target phenotypes also were identified Two distinct chemical series demonstrate on phenotype activity and cluster with tool compounds. This indicates these series have high potential for further chemistry exploration. Overall, a multi-omics platform is developed to generate cellular phenotypic compound 'Signatures'. This allows us to prioritize compound series in multiple
|
|
Tao Wang, GSK, Collegeville, United States
|
OA04.02 | Novel Screening Strategies to Characterize Ligandable Cysteines and Binding Pockets of Deubiquitinases
Deubiquitinases (DUBs) are a compelling drug target-class due to their central role in regulating protein homeostasis. There have been successful efforts to discover and characterize inhibitors across the enzyme family via biochemical and chemoproteomic screens, with several DUB inhibitors currently undergoing clinical investigation. Beyond DUB inhibitors, there is tremendous therapeutic potential for DUB ligands with alternative mechanisms of action; however, there are few examples of such compounds and there have been no systematic studies to identify ligandable pockets across DUBs. Cysteine profiling has emerged as a means of studying ligandable sites throughout the proteome and is used to discover starting points for chemical probes when performed in an electrophilic fragment screening format. While covalent compounds have been de-prioritized because of off-target concerns, they have garnered renewed interest due in part to their ability to target non-conserved nucleophilic residues, effectively expanding the landscape of druggable proteins. To address the need for allosteric DUB-targeted chemical probes, I have developed a DUB-biased cysteine profiling assay with unparalleled coverage of the DUB cysteinome. Through an electrophilic fragment screen, we have discovered novel ligands targeting previously uncharacterized DUB cysteines, allowing us to predict the presence of ligandable pockets outside the catalytic domain and across the DUBome. We are poised to develop these hits into the next generation of DUB targeted probes with diverse mechanisms of action.
|
|
Nicholas Girardi, Harvard University, Boston, United States
|
OA04.03 | The first fully automated high-throughput cell-based assay system to screen drugs in a 96-well plate format and address biological perturbations
In line with our efforts to push into clinical proteomics, we developed a high throughout screening (HTS) system to investigate the effects of potential therapeutic drugs on perturbed cells metabolically. We previously showed the identification of important cardiac modulators in ischemic cardiomyocytes from hypoxic stress and reperfusion. We now present the first 96-well plate HTP cell-based assay to demonstrate the agonistic properties of PR-364, a selective activator of the cytosolic E3 ubiquitin ligase required for mitophagy and down-regulated during ischemia. AC16 human cardiomyocytes (Sigma) were seeded and overnight cultures were subsequently grown in a 96-well plate and assayed under experimental conditions. Hypoxia was induced by treating the cells with cobalt chloride for 4 hours after an initial 4 hours of normal growth, whereas reperfusion required an immediate ischemic stress for 4 hours and then followed by 4 hours of reoxygenation. Cell lysates were lysed in ammonium bicarbonate on an LE220 Plus ultrasonication system (Covaris) and subsequently subjected to Cys reduction, alkylation, and trypsin digestion via a Biomek i7 (Beckman) automated workstation. Peptides were loaded on an Ultimate 3000 (Thermo) chromatography system coupled to an Exploris Orbitrap (Thermo) mass spectrometer. Data-Independent Acquisition (DIA) was performed over a 45-minute gradient and searched using the Pan human library. Proteomic analysis revealed more than 1,300 unique proteins across the biological replicates in each condition, representing a diverse dynamic range and a uniform distribution for statistical comparison, with 70 % of proteins displaying a CV < 30. When treated with PR-364, we observed a a significant difference in the proteome of ischemic cells in hypoxic and reperfused cardiomyocytes. Differentially expressed proteins that were quantified in 75 % of the replicates in the hypoxic and reperfused cells that were treated with the parking activating drug demonstrated distinct upregulated proteins that were downregulated in the control conditions without drug treatment.
|
|
Saeed Seyedmohammad
|
|
1:40 PM - 3:00 PM - Parallel Sessions
|
|
Parallel Session 05: Protein Networks - From Signaling to Interactions
Signaling mechanisms and protein networks in innate immunity
Toll-like receptor (TLR) signaling in macrophages is essential for generating effective innate immune responses. Quantitative differences dependent on the dose and timing of the stimulus critically affect cell function and often involve proteins that are not components of widely shared transduction pathways. Mathematical modeling is an important approach to better understand how these signaling networks function in time and space. Wehave successfully modeled the S1P signaling pathway in macrophages using selected reaction monitoring (SRM) to measure the absolute abundance of the pathway proteins. The resulting values became parameters in a computational pathway model. To model the TLR signaling networks, we developed assays for the canonical TLR signaling pathway and related proteins and phosphoproteins and used parallel reaction monitoring (PRM) with heavy-labeled internal peptide standards to quantify protein and phosphorylated protein moleculenumbers per cell in untreated and LPS-stimulated macrophages. The absolute protein abundance values were entered into a model of the TLR pathway developed using Simmune, the rule-based modeling tool with a visual interface. To reach beyond basal level quantification, the TLR signaling network model is tested further and combined with global proteomic approaches to discover biologically important new proteins, protein complexes and PTMs involved in this innate immune pathway, of which some examples will be given. The protein and PTM levels are quantified in macrophages under diverse, but well-defined conditions (different TLR ligands, whole pathogens, and cells with mutations in specific signaling molecules). These data will allow to parameterize and test the TLR network model under a variety of conditions. Together, the interconnected projects will lead to the better understanding how the immune signaling pathways are regulated and activated during an infection. This research was supported by the Intramural Research Program of NIAID, NIH.
|
|
Aleksandra Nita-Lazar, Ph. D., National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, United States
(Bio)
Dr. Aleksandra Nita-Lazar received her Ph.D. in biochemistry in 2003 from the University of Basel for studies performed at the Friedrich Miescher Institute for Biomedical Research, where she analyzed atypical protein glycosylation using mass spectrometry and protein biochemistry methods. After postdoctoral training at Stony Brook University and Massachusetts Institute of Technology (Ludwig Cancer Foundation Fellow), where she continued to investigate post-translational protein modifications and their influence on cell signaling, she joined the Program in Systems Immunology and Infectious Disease Modeling, now the Laboratory of Immune System Biology, DIR, NIAID, NIH, in April 2009, as an independent investigator and Chief of the Cellular Networks Proteomics Unit. Dr. Nita-Lazar was granted tenure in December 2018 and she now continues her work as Senior Investigator and Chief of the Functional Cellular Networks Section. Her main research interests are protein state changes and networks regulating the host-pathogen interactions and macrophage activation.
|
OA05.01 | Exploring the Interplay Between Protein Turnover and Assembly States: Mechanistic Insights and Age-Related Changes
Protein homeostasis, a critical aspect of cellular function, deteriorates with age, contributing to the onset of age-related diseases. However, our understanding of the molecular mechanisms leading to proteome imbalances during aging remains extremely limited. One reason for this knowledge gap is that age-associated changes in protein abundance are often subtle, falling below the detection limit of standard proteomics methods. Proteome imbalances can occur without major changes in protein expression levels, driven by factors such as alterations in the integration of proteins into molecular complexes or imbalances between synthesis and degradation that impact protein turnover. This can result in the accumulation of supernumerary, damaged, or orphaned proteins, potentially leading to protein aggregation, a hallmark of aging-related pathologies. Despite the importance of both protein assembly and turnover in aging, few studies have simultaneously measured these processes from the same sample to explore their interdependence. In this study, we systematically investigate the relationship between protein assembly states and turnover rates in brain and liver tissues, and how these relationships shift with aging. By combining metabolic labeling with size-exclusion chromatography, we quantify protein half-lives in monomeric versus assembled forms and analyze how these dynamics change from young to aged mice. Our findings illuminate the intricate link between turnover and assembly-state regulation, highlighting tissue-specific differences and age-related changes, and offer new insights into the mechanisms of proteostasis in aging.
|
|
Ella Doron-Mandel, Columbia University, New York, NY, United States
|
OA05.02 | PrIUS: A Versatile Workflow for Identifying Ubiquitylation Sites and Distinguishing Non-Substrate Interactors in E3 Ligase Pathways
Protein ubiquitylation is a highly complex post-translational modification (PTM) involved in regulating a multitude of cellular processes. This complexity is reflected in the fact that the human genome encodes over 600 E3 ubiquitin ligases, and at least 100,000 ubiquitylation sites have been reported within the proteome. However, despite this knowledge, a critical gap remains: the vast majority of these ubiquitylation sites have not been directly linked to their associated E3 ligase(s). Identifying the specific locations of ubiquitylation sites on substrates, determining which E3 ligase(s) are responsible, and understanding the context in which specific ubiquitylation events occur are essential for deciphering the full impact of ubiquitin signaling. To address this gap and directly identify E3 ligase substrate sites in a cellular context, we developed an approach called Proximity-based Identification of Ubiquitin Substrates (PrIUS). PrIUS employs a standard BioID workflow to first generate a proximity-enriched E3 ligase interactome that includes both substrate and non-substrate interactions. From this interactome, substrates and their ubiquitylation sites can be directly identified through serial enrichment of diGly remnants. Using quantitative label-free mass spectrometry, we benchmarked PrIUS with the E3 ligase NEDD4L, successfully identifying NEDD4L substrates along with their diGly sites. Additionally, BioID analysis prior to diGly enrichment allowed us to identify non-substrate NEDD4L interactors. Furthermore, PrIUS enabled the identification of PROTAC-induced ubiquitylation sites in situ. For example, using the NSD2 degrader UNC8732, which recruits the E3 ligase FBXO22, we precisely identified and validated PROTAC-induced ubiquitylation sites on NSD2. In this context, PrIUS allows simultaneous characterization of PROTAC specificity at both the protein interaction (BioID) and substrate (diGly) levels within a cellular environment. Overall, PrIUS is a versatile approach for identifying E3 ligase substrate ubiquitylation sites while also distinguishing non-substrate interactors.
|
|
Tanner Tessier, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
|
OA05.03 | Quantifying loci-specific proteomes with oligonucleotide-directed proximity-interactome mapping
The accuracy of crucial nuclear processes, such as transcription, replication, and repair, depends largely on the local composition of chromatin and the post-translational modifications (PTMs) present on histone proteins. Current approaches suffer from significant technical limitations that hinder their wide-scale adaption and deployment. In order to meet these challenges, we have developed DNA oligonucleotide-directed proximity-interactome mapping (DNA OMAP). By combining proximity biotinylation with loci-specfic in situ hybridization, DNA-OMAP presents a flexible, high-throughput method to quantify the molecular neighborhoods associated with specific genomic loci. DNA OMAP utilizes programmable peroxidase-conjugated oligonucleotide probes to biotinylate proteins near specific genomic loci. This approach enables the purification and identification of DNA-associated proteins and interacting DNA regions with high specificity using ~5-10x fewer cells than comparable methods. To validate our method, we performed both single probe and multi-probe experiments. Using probes targeted to telomeric repeats, we were able to observe evidence of specific labeling via imaging as well as a strong enrichment of known telomere associated proteins such as the shelterin complex. Performing an experiment with three probes (mitochondrial, telomeric, and, peri-centromeric) shows OMAP capable of differentiating between both cellular compartments and sub-compartments, with strong enrichment known interactors quantified at each locus. While small, single-copy loci have not been used for proteomics experiments yet, DNA-DNA interactions have been captured and present exciting future directions for OMAP. DNA OMAP provides a powerful and flexible tool for exploring the intricate relationships between DNA sequence, chromatin structure, and cellular function, addressing longstanding challenges in the field.
|
|
Chris McGann, University of Washington, Seattle, United States
|
|
|
Parallel Session 06: High Throughput Proteomics - Tackling 1000+ Samples
|
|
Roman Fischer
(Bio)
Roman Fischer is an associate professor at the Target Discovery Institute (TDI) and Centre of Medicines' Discovery (CMD) at the Nuffield Department of Medicine at the University of Oxford. He leads 2 proteomics focussed laboratories: the Discovery Proteomics Facility (DisPro, Academic Lead) and the Clinical Proteomics Group (CPG, Principal Investigator). DisPro engages in fundamental research collaborations involving protein-protein interactions, deep proteomes and PTM analysis using top end proteomics equipment (Orbitrap Astral, Orbitrap Ascend, TimsTOF HT, Q-Exactive). In the Clinical Research Group, RF focusses on own research interests covering the technology driven disciplines of spatial and single cell proteomics as well as high-throughput proteomics. The dedicated lab uses a TimsTOF Ultra 2, TimsTOF Flex, Leica LMD7, Leica Mica and CellenOne to develop specific workflows to address spatial proteome organisation in pathology, with a focus on cancer and its triggered immune response in specific cell compartments of the immune system.
RF has published >250 peer reviewed manuscripts, which acquired >14000 citations (h-index 63) and is an established leader in the field of proteomics with >20 years of experience in this area. DisPro and CPG have links to all major hospitals in Oxford and long-standing collaborations with most departments across the University, facilitating close interaction and collaboration with clinical PIs with access to study and trial samples.
|
OA06.01 | Fast histone extraction combined with automation for high-throughput PTM analysis
Sample preparation is the most hands-on, time-consuming step for proteomics analysis and one of the most critical parts. The variability from person to person and the limitation on sample throughput is one of the disadvantages of manual sample preparation. Automation is already present in proteomics, mainly for total and clinical proteome analyses. Histone PTM analysis is still a challenge due to the complexity of the data analysis, but the sample preparation workflow is well-established for our lab. Here, we present an approach to reduce histone extraction time followed by automated chemical derivatization, fast trypsin digestion, and desalting on a 96-well format. The resulting peptides are analyzed using a Q-TOF on a 20-minute SWATH gradient, optimizing the instrument time. The histone extraction uses an HCl and H2SO4 quick acid extraction with no incubation time. The liquid handling system Andrew from Waters performed the automated histone propionylation, fast trypsin digestion, and desalting. The mass spectrometry analysis is conducted on an M-CLASS HPLC from Waters coupled to a 7600 Zeno-TOF Sciex mass spectrometer using a 20-minute gradient in SWATH mode. The data analysis is performed using an updated version of our EpiProfile software. The fast histone extraction not only shows a higher yield compared to the traditional 2 hr acid extraction but also reduces hands-on time by 80%. The automated sample preparation, followed by the fast-gradient mass spectrometry runs, and the data analysis can be completed in just around 45 minutes per sample, significantly saving time and effort. Establishing this workflow will help to achieve a robust platform for deep histone PTM analysis (~150 PTMs) with reproducible and throughput results, which will benefit biological insights by enabling more samples with high-quality results.
|
|
Francisca de Luna Vitorino, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
|
OA06.02 | Improved Proteome Coverage and Reproducibility in Large Scale Analyses using Packed Emitter Columns.
Presenting author: Danni Smith Authors: Vincent Albrecht1, Tim Heymann1, Danni Smith2, Greta Briedytė2, Johannes Bruno Müller-Reif1. Affiliations: 1Max Planck Institute of Biochemistry, 2IonOpticks Pty Ltd. Proteomic analyses of large cohorts of human patient samples plays a critical role in discovering novel biomarkers for diagnosis of disease and responses to treatment. Here, we introduce a novel packed emitter chromatography column, the IonOpticks Aurora Rapid 8x150 (8 cm x 150 μm inner diameter, 1.7 μm C18), designed to improve the performance of large cohort proteomic analyses. Quality control samples embedded within a large human patient cohort were used to assess the efficiency and reproducibility of the column. Patient plasma samples were analysed on Aurora Rapid 8 cm x 150 μm column on a 100SPD method using Evosep One and Thermo Fisher Orbitrap Astral Mass Spectrometer. Weekly quality control injections (200 ng HeLa tryptic digest) were included to monitor system performance (acquired using 60 samples per day method). Analysis of the HeLa quality control samples demonstrated robust protein identifications, with over 9,300 proteins and 100,000 unique peptides identified per QC run across multiple months and more than 10 individual columns. Coefficient of variation, Pearson correlation and average peak width were calculated to assess column reproducibility across the cohort. A median CV of 2.52 %, >0.98 Pearson correlation score and 2.9 second FWHM were achieved across the 257 QC analyses. Our data demonstrates that the novel packed emitter column combined with the Evosep One and Thermo Fisher Orbitrap Astral Mass Spectrometer enabled maximum protein identifications whilst ensuring robust and reproducible protein quantification across large cohort analyses.
|
|
Daniela-lee Smith, IonOpticks, Melbourne, Victoria, Australia
|
OA06.03 | Scalable single cell analysis towards 1000 samples per day using the Evosep Whisper zoom methods on the timsTOF Ultra 2
Scalability is crucial for meaningful data collection with reliable and robust statistical power in single-cell proteomics workflows. Multiplexing and fast chromatography help upscale proteome analyses. Here, we demonstrate the applicability of the new EvosepOne Whisper Zoom methods for speeding up label-free sample analysis up to 120 samples per day (SPD) and exceeding >1000 samples per day in a multiplexing approach with data acquisition on the timsTOF Ultra 2. HeLa cells were isolated into the proteoCHIP® EVO 96, directly lysed and digested using the cellenONE platform. Samples were transferred onto Evotips, separated in Whisper Zoom 120, 80 and 40 SPD, analyzed on timsTOF Ultra 2 in dia-PASEF® mode and processed with Spectronaut 19 using directDIA+. HeLa, HEK 293 and K562 protein digests were labeled with 16-plex TMTpro, combined into 9-plex single-cell equivalent samples (250pg per label), acquired in Whisper Zoom 120SPD on the timsTOF Ultra 2 in dda-PASEF and analyzed with Spectromine 4.5. Analyses of HeLa cell digest resulted in >4000 protein groups from 250 pg at 120SPD, 5000 proteins at 80SPD and 5500 protein groups at 40SPD speed; reaching 6500 protein groups at 120SPD, 7000 protein groups at 80SPD and 7700 protein groups at 40SPD from the higher loads. From single HeLa cells, protein identification rates correlated with gradient length, identifying on average 2500 protein groups at 120SPD, 3000 protein groups at 80SPD and 4000 protein groups at 40SPD. The 9-plex multiplexing approach run at Whisper Zoom 120SPD speed resulted in 1500 protein groups on average per TMT label/cell type. Excellent chromatographic reproducibility along with quantitative accuracy within and across TMT batches was observed, accurately separating the 360 HEK 293 samples from the 360 HeLa samples and the 360 K562 samples.
|
|
Dijana Vitko, Bruker Daltonics Inc., Austin, TX, United States
|
|
3:15 PM - 4:30 PM - Lightning Session
|
|
Lightning Talks - Round 01
Sponsored By:
Alamar Biosciences
|
4:30 PM - 6:00 PM - Poster Session
|
|
Poster Session 01 and Exhibitor Viewing
Sponsored By: QuantumSi
|
6:00 PM - 7:15 PM - Evening Workshops
|
|
Evening Workshops: NCI's Clinical Proteomic Tumor Analysis Consortium (CPTAC): Computational Tools for Multi-omics Integration Analysis
The National Cancer Institute's Clinical Proteomic Tumor Analysis Consortium (NCI's CPTAC) is a national effort to accelerate the understanding of the molecular basis of cancer through the integration of large-scale proteome and genome analysis, or proteogenomics. This goal is achieved through the application of large-scale proteome and genome analyses, followed by proteogenomic integration of genome, transcriptome, and proteome data sets. CPTAC is also supporting development of new proteogenomic data analysis tools. All data and analytical tools are made broadly available to the research community through public databases to maximize utility and public benefit.
Presented By:
|
Xu Zhang
(Bio)
Dr. Zhang is a Program Manager in the Office of Cancer Clinical Proteomics Research (OCCPR) at the National Cancer Institute (NCI), National Institutes of Health (NIH). She provides scientific expertise in Proteomics Data Science/Data Management, manages and oversees the grants and contracts that support proteomics data analysis, informatics and software tools, and data management activities for OCCPR's CPTAC, International Cancer Proteogenome Consortium (ICPC), and the Applied Proteogenomics Organizational Learning and Outcomes (APOLLO) network. Dr. Zhang has extensive experience in the proteogenomic field, especially in proteomics data management.
|
|
Alexey Nesvizhskii
(Bio)
Prof. Alexey Nesvizhskii is the Godfrey Dorr Stobbe Professor of Bioinformatics in the Departments of Pathology and Computational Medicine & Bioinformatics at the University of Michigan, Ann Arbor. Prof. Nesvizhskii earned his Ph.D. in Physics from the University of Washington in Seattle in 2001, followed by postdoctoral research in proteomics with Prof. Ruedi Aebersold at the Institute for Systems Biology. He has been on the faculty at the University of Michigan since 2005. His research focuses on mass spectrometry-based proteomics and proteogenomics, bioinformatics, and multi-omics data integration. The computational algorithms and software tools developed by Prof. Nesvizhskii and his group are utilized by thousands of scientists worldwide.
|
|
Bing Zhang
(Bio)
Dr. Bing Zhang is a Cancer Prevention and Research Institute of Texas (CPRIT) Scholar, McNair Medical Institute Scholar, and Professor of Molecular and Human Genetics in Baylor College of Medicine. He is an internationally recognized leader in computational cancer proteogenomics, with a focus on developing informatics solutions that help translate cancer genomic and proteomic data into biological and clinical insights.
|
|
Natalie Clark, Integration of metabolomics analyses with PANOPLY (15mins)
(Bio)
Natalie Clark is a Computational Scientist in the Proteomics Platform at the Broad Institute of MIT and Harvard. She received her Ph.D. in Biomathematics from North Carolina State University where she worked on spatial transcriptomics in the Arabidopsis root. She then completed a post-doc focused on maize proteomics at Iowa State University prior to joining the Broad Institute. Natalie's main focus is developing new methods for integrative, multi-omics analyses of large-scale data such as those produced by CPTAC.
|
|
Pei Wang
(Bio)
Dr. Pei Wang is a Professor of Genetic and Genomic Sciences at Icahn School of Medicine at Mount Sinai. She obtained her B.S. in Mathematics from Peking University, China, in 2000; and her Ph.D. in Statistics from Stanford University in 2004. Between 2004-2013, Dr. Wang served as a faculty in Program of Biostatistics, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center and Department of Biostatistics, University of Washington, Seattle, WA. In Oct 2013, Dr. Wang joint Icahn Medical School at Mount Sinai, New York to lead an integrative proteogenomic research program. Dr. Wang's research interest has focused on developing statistical and computational methods for analyzing high-throughput omics data including mass-spectrometry based proteomics data, as well as high-dimensional network inference. She is a Principle Investigator of the NCI CPTAC (Clinical Proteomic Tumor Analysis Consortium) that aims to understand the molecular basis of cancer through large-scale proteome and genome analysis.
|
|
Ratna Thangudu
(Bio)
Dr. Ratna Rajesh Thangudu is the Director of Bioinformatics at ICF, where he leads data commons initiatives that align with the National Cancer Institute's (NCI) mission to accelerate cancer research. He currently manages the Proteomic Data Commons (PDC), a key component of the Cancer Research Data Commons (CRDC), and works closely with other CRDC teams to ensure seamless integration of imaging, genomic, and proteomic data resources. With deep expertise in developing and deploying cloud-based platforms, Dr. Thangudu specializes in large-scale, data-driven solutions that optimize data reuse and enable high-throughput analysis of multi-omic datasets.
|
|
|
Evening Workshop: Business of Starting a Lab
|
|
Evening Workshop: Ed&Out Committee - Video Competition
|
7:30 PM - 8:30 PM - ECR Event
|
|
ECR Event: Elevator Pitches and Speed Dating
|