119 results on '"Namrata D Udeshi"'
Search Results
2. Dynamic regulation of inter-organelle communication by ubiquitylation controls skeletal muscle development and disease onset
- Author
-
Arian Mansur, Remi Joseph, Euri S Kim, Pierre M Jean-Beltran, Namrata D Udeshi, Cadence Pearce, Hanjie Jiang, Reina Iwase, Miroslav P Milev, Hashem A Almousa, Elyshia McNamara, Jeffrey Widrick, Claudio Perez, Gianina Ravenscroft, Michael Sacher, Philip A Cole, Steven A Carr, and Vandana A Gupta
- Subjects
myopathy ,ubiquitylation ,skeletal muscle ,genetic disease ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Ubiquitin-proteasome system (UPS) dysfunction is associated with the pathology of a wide range of human diseases, including myopathies and muscular atrophy. However, the mechanistic understanding of specific components of the regulation of protein turnover during development and disease progression in skeletal muscle is unclear. Mutations in KLHL40, an E3 ubiquitin ligase cullin3 (CUL3) substrate-specific adapter protein, result in severe congenital nemaline myopathy, but the events that initiate the pathology and the mechanism through which it becomes pervasive remain poorly understood. To characterize the KLHL40-regulated ubiquitin-modified proteome during skeletal muscle development and disease onset, we used global, quantitative mass spectrometry-based ubiquitylome and global proteome analyses of klhl40a mutant zebrafish during disease progression. Global proteomics during skeletal muscle development revealed extensive remodeling of functional modules linked with sarcomere formation, energy, biosynthetic metabolic processes, and vesicle trafficking. Combined analysis of klh40 mutant muscle proteome and ubiquitylome identified thin filament proteins, metabolic enzymes, and ER-Golgi vesicle trafficking pathway proteins regulated by ubiquitylation during muscle development. Our studies identified a role for KLHL40 as a regulator of ER-Golgi anterograde trafficking through ubiquitin-mediated protein degradation of secretion-associated Ras-related GTPase1a (Sar1a). In KLHL40-deficient muscle, defects in ER exit site vesicle formation and downstream transport of extracellular cargo proteins result in structural and functional abnormalities. Our work reveals that the muscle proteome is dynamically fine-tuned by ubiquitylation to regulate skeletal muscle development and uncovers new disease mechanisms for therapeutic development in patients.
- Published
- 2023
- Full Text
- View/download PDF
3. An engineered transcriptional reporter of protein localization identifies regulators of mitochondrial and ER membrane protein trafficking in high-throughput CRISPRi screens
- Author
-
Robert Coukos, David Yao, Mateo I Sanchez, Eric T Strand, Meagan E Olive, Namrata D Udeshi, Jonathan S Weissman, Steven A Carr, Michael C Bassik, and Alice Y Ting
- Subjects
protein engineering ,CRISPR ,high-throughput screens ,tail-anchored proteins ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The trafficking of specific protein cohorts to correct subcellular locations at correct times is essential for every signaling and regulatory process in biology. Gene perturbation screens could provide a powerful approach to probe the molecular mechanisms of protein trafficking, but only if protein localization or mislocalization can be tied to a simple and robust phenotype for cell selection, such as cell proliferation or fluorescence-activated cell sorting (FACS). To empower the study of protein trafficking processes with gene perturbation, we developed a genetically encoded molecular tool named HiLITR (High-throughput Localization Indicator with Transcriptional Readout). HiLITR converts protein colocalization into proteolytic release of a membrane-anchored transcription factor, which drives the expression of a chosen reporter gene. Using HiLITR in combination with FACS-based CRISPRi screening in human cell lines, we identified genes that influence the trafficking of mitochondrial and ER tail-anchored proteins. We show that loss of the SUMO E1 component SAE1 results in mislocalization and destabilization of many mitochondrial tail-anchored proteins. We also demonstrate a distinct regulatory role for EMC10 in the ER membrane complex, opposing the transmembrane-domain insertion activity of the complex. Through transcriptional integration of complex cellular functions, HiLITR expands the scope of biological processes that can be studied by genetic perturbation screening technologies.
- Published
- 2021
- Full Text
- View/download PDF
4. Correction: Proteomic mapping of cytosol-facing outer mitochondrial and ER membranes in living human cells by proximity biotinylation
- Author
-
Victoria Hung, Stephanie S Lam, Namrata D Udeshi, Tanya Svinkina, Gaelen Guzman, Vamsi K Mootha, Steven A Carr, and Alice Y Ting
- Subjects
Medicine ,Science ,Biology (General) ,QH301-705.5 - Published
- 2019
- Full Text
- View/download PDF
5. Lso2 is a conserved ribosome-bound protein required for translational recovery in yeast.
- Author
-
Yinuo J Wang, Pavanapuresan P Vaidyanathan, Maria F Rojas-Duran, Namrata D Udeshi, Kristen M Bartoli, Steven A Carr, and Wendy V Gilbert
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Ribosome-binding proteins function broadly in protein synthesis, gene regulation, and cellular homeostasis, but the complete complement of functional ribosome-bound proteins remains unknown. Using quantitative mass spectrometry, we identified late-annotated short open reading frame 2 (Lso2) as a ribosome-associated protein that is broadly conserved in eukaryotes. Genome-wide crosslinking and immunoprecipitation of Lso2 and its human ortholog coiled-coil domain containing 124 (CCDC124) recovered 25S ribosomal RNA in a region near the A site that overlaps the GTPase activation center. Consistent with this location, Lso2 also crosslinked to most tRNAs. Ribosome profiling of yeast lacking LSO2 (lso2Δ) revealed global translation defects during recovery from stationary phase with translation of most genes reduced more than 4-fold. Ribosomes accumulated at start codons, were depleted from stop codons, and showed codon-specific changes in occupancy in lso2Δ. These defects, and the conservation of the specific ribosome-binding activity of Lso2/CCDC124, indicate broadly important functions in translation and physiology.
- Published
- 2018
- Full Text
- View/download PDF
6. Proteomic mapping of cytosol-facing outer mitochondrial and ER membranes in living human cells by proximity biotinylation
- Author
-
Victoria Hung, Stephanie S Lam, Namrata D Udeshi, Tanya Svinkina, Gaelen Guzman, Vamsi K Mootha, Steven A Carr, and Alice Y Ting
- Subjects
microscopy ,promiscuous enzymatic labeling ,subcellular regions ,APEX2 ,mitochondria-ER junctions ,mitochondria-associated membrane ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The cytosol-facing membranes of cellular organelles contain proteins that enable signal transduction, regulation of morphology and trafficking, protein import and export, and other specialized processes. Discovery of these proteins by traditional biochemical fractionation can be plagued with contaminants and loss of key components. Using peroxidase-mediated proximity biotinylation, we captured and identified endogenous proteins on the outer mitochondrial membrane (OMM) and endoplasmic reticulum membrane (ERM) of living human fibroblasts. The proteomes of 137 and 634 proteins, respectively, are highly specific and highlight 94 potentially novel mitochondrial or ER proteins. Dataset intersection identified protein candidates potentially localized to mitochondria-ER contact sites. We found that one candidate, the tail-anchored, PDZ-domain-containing OMM protein SYNJ2BP, dramatically increases mitochondrial contacts with rough ER when overexpressed. Immunoprecipitation-mass spectrometry identified ribosome-binding protein 1 (RRBP1) as SYNJ2BP’s ERM binding partner. Our results highlight the power of proximity biotinylation to yield insights into the molecular composition and function of intracellular membranes.
- Published
- 2017
- Full Text
- View/download PDF
7. Quantitative-proteomic comparison of alpha and Beta cells to uncover novel targets for lineage reprogramming.
- Author
-
Amit Choudhary, Kaihui Hu He, Philipp Mertins, Namrata D Udeshi, Vlado Dančík, Dina Fomina-Yadlin, Stefan Kubicek, Paul A Clemons, Stuart L Schreiber, Steven A Carr, and Bridget K Wagner
- Subjects
Medicine ,Science - Abstract
Type-1 diabetes (T1D) is an autoimmune disease in which insulin-secreting pancreatic beta cells are destroyed by the immune system. An emerging strategy to regenerate beta-cell mass is through transdifferentiation of pancreatic alpha cells to beta cells. We previously reported two small molecules, BRD7389 and GW8510, that induce insulin expression in a mouse alpha cell line and provide a glimpse into potential intermediate cell states in beta-cell reprogramming from alpha cells. These small-molecule studies suggested that inhibition of kinases in particular may induce the expression of several beta-cell markers in alpha cells. To identify potential lineage reprogramming protein targets, we compared the transcriptome, proteome, and phosphoproteome of alpha cells, beta cells, and compound-treated alpha cells. Our phosphoproteomic analysis indicated that two kinases, BRSK1 and CAMKK2, exhibit decreased phosphorylation in beta cells compared to alpha cells, and in compound-treated alpha cells compared to DMSO-treated alpha cells. Knock-down of these kinases in alpha cells resulted in expression of key beta-cell markers. These results provide evidence that perturbation of the kinome may be important for lineage reprogramming of alpha cells to beta cells.
- Published
- 2014
- Full Text
- View/download PDF
8. Engineered allostery in light-regulated LOV-Turbo enables precise spatiotemporal control of proximity labeling in living cells
- Author
-
Song-Yi Lee, Joleen S. Cheah, Boxuan Zhao, Charles Xu, Heegwang Roh, Christina K. Kim, Kelvin F. Cho, Namrata D. Udeshi, Steven A. Carr, and Alice Y. Ting
- Subjects
Cell Biology ,Molecular Biology ,Biochemistry ,Article ,Biotechnology - Abstract
The incorporation of light-responsive domains into engineered proteins has enabled control of protein localization, interactions, and function with light. We integrated optogenetic control into proximity labeling (PL), a cornerstone technique for high-resolution proteomic mapping of organelles and interactomes in living cells. Through structure-guided screening and directed evolution, we installed the light-sensitive LOV domain into the PL enzyme TurboID to rapidly and reversibly control its labeling activity with low-power blue light. “LOV-Turbo” works in multiple contexts and dramatically reduces background in biotin-rich environments such as neurons. We used LOV-Turbo for pulse-chase labeling to discover proteins that traffick between endoplasmic reticulum, nuclear, and mitochondrial compartments under cellular stress. We also showed that instead of external light, LOV-Turbo can be activated by BRET from luciferase, enabling interaction-dependent PL. Overall, LOV-Turbo increases the spatial and temporal precision of PL, expanding the scope of experimental questions that can be addressed with PL.
- Published
- 2023
9. Supplementary Data from Avadomide Induces Degradation of ZMYM2 Fusion Oncoproteins in Hematologic Malignancies
- Author
-
Benjamin L. Ebert, Steven A. Carr, Philip P. Chamberlain, Mark Rolfe, Jean-Michel Cayuela, Jean-Jacques Kiladjian, Stéphane De Botton, Véronique Saada, Christophe Marzac, Sophie Cotteret, Rob S. Sellar, Andrew A. Guirguis, Alexander Tepper, Kaushik Viswanathan, Marie McConkey, Thomas Clayton, Mary E. Matyskiela, Namrata D. Udeshi, Pierre M. Jean Beltran, Daniel E. Grinshpun, Jessica A. Gasser, and Aline Renneville
- Abstract
Supplementary Methods, Figures, and Tables
- Published
- 2023
10. Data from Avadomide Induces Degradation of ZMYM2 Fusion Oncoproteins in Hematologic Malignancies
- Author
-
Benjamin L. Ebert, Steven A. Carr, Philip P. Chamberlain, Mark Rolfe, Jean-Michel Cayuela, Jean-Jacques Kiladjian, Stéphane De Botton, Véronique Saada, Christophe Marzac, Sophie Cotteret, Rob S. Sellar, Andrew A. Guirguis, Alexander Tepper, Kaushik Viswanathan, Marie McConkey, Thomas Clayton, Mary E. Matyskiela, Namrata D. Udeshi, Pierre M. Jean Beltran, Daniel E. Grinshpun, Jessica A. Gasser, and Aline Renneville
- Abstract
Thalidomide analogues exert their therapeutic effects by binding to the CRL4CRBN E3 ubiquitin ligase, promoting ubiquitination and subsequent proteasomal degradation of specific protein substrates. Drug-induced degradation of IKZF1 and IKZF3 in B-cell malignancies demonstrates the clinical utility of targeting disease-relevant transcription factors for degradation. Here, we found that avadomide (CC-122) induces CRBN-dependent ubiquitination and proteasomal degradation of ZMYM2 (ZNF198), a transcription factor involved in balanced chromosomal rearrangements with FGFR1 and FLT3 in aggressive forms of hematologic malignancies. The minimal drug-responsive element of ZMYM2 is a zinc-chelating MYM domain and is contained in the N-terminal portion of ZMYM2 that is universally included in the derived fusion proteins. We demonstrate that avadomide has the ability to induce proteasomal degradation of ZMYM2–FGFR1 and ZMYM2–FLT3 chimeric oncoproteins, both in vitro and in vivo. Our findings suggest that patients with hematologic malignancies harboring these ZMYM2 fusion proteins may benefit from avadomide treatment.Significance:We extend the potential clinical scope of thalidomide analogues by the identification of a novel avadomide-dependent CRL4CRBN substrate, ZMYM2. Avadomide induces ubiquitination and degradation of ZMYM2–FGFR1 and ZMYM2–FLT3, two chimeric oncoproteins involved in hematologic malignancies, providing a proof of concept for drug-induced degradation of transcription factor fusion proteins by thalidomide analogues.
- Published
- 2023
11. Data from A Ubiquitination Cascade Regulating the Integrated Stress Response and Survival in Carcinomas
- Author
-
William C. Hahn, Francisca Vazquez, James M. McFarland, David E. Root, Steven A. Carr, Meagan E. Olive, Namrata D. Udeshi, Federica Piccioni, John Michael Krill-Burger, Joshua M. Dempster, Mariya Kazachkova, Nolan R. Bick, Alfredo Gonzalez, Nancy Dumont, Brian H. Shim, Sydney M. Moyer, Naomi Li, Lisa Leung, Linh He, Benjamin Gaeta, Ashir A. Borah, Tsukasa Shibue, and Lisa D. Cervia
- Abstract
Systematic identification of signaling pathways required for the fitness of cancer cells will facilitate the development of new cancer therapies. We used gene essentiality measurements in 1,086 cancer cell lines to identify selective coessentiality modules and found that a ubiquitin ligase complex composed of UBA6, BIRC6, KCMF1, and UBR4 is required for the survival of a subset of epithelial tumors that exhibit a high degree of aneuploidy. Suppressing BIRC6 in cell lines that are dependent on this complex led to a substantial reduction in cell fitness in vitro and potent tumor regression in vivo. Mechanistically, BIRC6 suppression resulted in selective activation of the integrated stress response (ISR) by stabilization of the heme-regulated inhibitor, a direct ubiquitination target of the UBA6/BIRC6/KCMF1/UBR4 complex. These observations uncover a novel ubiquitination cascade that regulates ISR and highlight the potential of ISR activation as a new therapeutic strategy.Significance:We describe the identification of a heretofore unrecognized ubiquitin ligase complex that prevents the aberrant activation of the ISR in a subset of cancer cells. This provides a novel insight on the regulation of ISR and exposes a therapeutic opportunity to selectively eliminate these cancer cells.See related commentary Leli and Koumenis, p. 535.This article is highlighted in the In This Issue feature, p. 517
- Published
- 2023
12. Supplementary Figures and Tables from A Ubiquitination Cascade Regulating the Integrated Stress Response and Survival in Carcinomas
- Author
-
William C. Hahn, Francisca Vazquez, James M. McFarland, David E. Root, Steven A. Carr, Meagan E. Olive, Namrata D. Udeshi, Federica Piccioni, John Michael Krill-Burger, Joshua M. Dempster, Mariya Kazachkova, Nolan R. Bick, Alfredo Gonzalez, Nancy Dumont, Brian H. Shim, Sydney M. Moyer, Naomi Li, Lisa Leung, Linh He, Benjamin Gaeta, Ashir A. Borah, Tsukasa Shibue, and Lisa D. Cervia
- Abstract
Supplementary Figure S1 shows the strategy and result of co-essentiality module identificationfrom the DepMap CRISPR screen dataset and further characterization of the BIRC6 module. Supplementary Figure S2 shows the viability effect of BIRC6 depletion in BIRC6-dependent and -nondependent cancer cells lines as well asin nontransformed cells. Supplementary Figure S3 shows in vitro confirmations of the inducible BIRC6 depletion (by RNAi and CRISPR) systems as well as the effect of BIRC6 depletion on metastasis in xenograft models. Supplementary Figure S4 shows detailed results for the allele competition assay to evaluate the role of the BIR and UBC domains of BIRC6 and further evidence for the physical assembly of BIRC6 ubiquitin ligase complex. Supplementary Figure S5 shows evidence for the selective activation of the p-eIF2alpha/ATF4 arm of UPR (or ISR) upon BIRC6 depletion. Supplementary Figure S6 shows evidence supporting the importance of HRI-mediated ISR activation as a mechanism underlying the loss of viability caused by BIRC6 complex suppression. Supplementary Figure S7 shows evidence for the direct regulation of HRI ubiquitination and degradation by the BIRC6 complex Supplementary Figure S8 shows elevated expression of HRI mRNA in the tumor samples compared to the normal samples in large-scale datasets (TCGA, TARGET and GTEx). Supplementary Figure S9 shows the inefficiency of the gene expression and copy number of the functionally related genes in predicting BIRC6 dependency. Supplementary Table S1 lists the top 50 co-essentiality modules identified in the analysis explained in Supplementary Figure S1A. Supplementary Table S2 lists the key DNA and RNA sequences used in this study.
- Published
- 2023
13. Supplementary Table S1 from Avadomide Induces Degradation of ZMYM2 Fusion Oncoproteins in Hematologic Malignancies
- Author
-
Benjamin L. Ebert, Steven A. Carr, Philip P. Chamberlain, Mark Rolfe, Jean-Michel Cayuela, Jean-Jacques Kiladjian, Stéphane De Botton, Véronique Saada, Christophe Marzac, Sophie Cotteret, Rob S. Sellar, Andrew A. Guirguis, Alexander Tepper, Kaushik Viswanathan, Marie McConkey, Thomas Clayton, Mary E. Matyskiela, Namrata D. Udeshi, Pierre M. Jean Beltran, Daniel E. Grinshpun, Jessica A. Gasser, and Aline Renneville
- Abstract
Supplementary Table 1. Proteomic data
- Published
- 2023
14. Workflow enabling deepscale immunopeptidome, proteome, ubiquitylome, phosphoproteome, and acetylome analyses of sample-limited tissues
- Author
-
Jennifer G. Abelin, Erik J. Bergstrom, Keith D. Rivera, Hannah B. Taylor, Susan Klaeger, Charles Xu, Eva K. Verzani, C. Jackson White, Hilina B. Woldemichael, Maya Virshup, Meagan E. Olive, Myranda Maynard, Stephanie A. Vartany, Joseph D. Allen, Kshiti Phulphagar, M. Harry Kane, Suzanna Rachimi, D. R. Mani, Michael A. Gillette, Shankha Satpathy, Karl R. Clauser, Namrata D. Udeshi, and Steven A. Carr
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
Serial multi-omic analysis of proteome, phosphoproteome, and acetylome provides insights into changes in protein expression, cell signaling, cross-talk and epigenetic pathways involved in disease pathology and treatment. However, ubiquitylome and HLA peptidome data collection used to understand protein degradation and antigen presentation have not together been serialized, and instead require separate samples for parallel processing using distinct protocols. Here we present MONTE, a highly sensitive multi-omic native tissue enrichment workflow, that enables serial, deep-scale analysis of HLA-I and HLA-II immunopeptidome, ubiquitylome, proteome, phosphoproteome, and acetylome from the same tissue sample. We demonstrate that the depth of coverage and quantitative precision of each ‘ome is not compromised by serialization, and the addition of HLA immunopeptidomics enables the identification of peptides derived from cancer/testis antigens and patient specific neoantigens. We evaluate the technical feasibility of the MONTE workflow using a small cohort of patient lung adenocarcinoma tumors.
- Published
- 2023
15. Sensitive, high-throughput HLA-I and HLA-II immunopeptidomics using parallel accumulation-serial fragmentation mass spectrometry
- Author
-
Kshiti Meera Phulphagar, Claudia Ctortecka, Alvaro Sebastian Vaca Jacome, Susan Klaeger, Eva K. Verzani, Gabrielle M. Hernandez, Namrata D. Udeshi, Karl R. Clauser, Jennifer G. Abelin, and Steven A. Carr
- Subjects
Molecular Biology ,Biochemistry ,Article ,Analytical Chemistry - Abstract
Comprehensive, in-depth identification of the human leukocyte antigen HLA-I and HLA-II tumor immunopeptidome can inform the development of cancer immunotherapies. Mass spectrometry (MS) is powerful technology for direct identification of HLA peptides from patient derived tumor samples or cell lines. However, achieving sufficient coverage to detect rare, clinically relevant antigens requires highly sensitive MS-based acquisition methods and large amounts of sample. While immunopeptidome depth can be increased by off-line fractionation prior to MS, its use is impractical when analyzing limited amounts of primary tissue biopsies. To address this challenge, we developed and applied a high throughput, sensitive, single-shot MS-based immunopeptidomics workflow that leverages trapped ion mobility time-of-flight mass spectrometry on the Bruker timsTOF SCP. We demonstrate >2-fold improved coverage of HLA immunopeptidomes relative to prior methods with up to 15,000 distinct HLA-I and HLA-II peptides from 4e7 cells. Our optimized single-shot MS acquisition method on the timsTOF SCP maintains high coverage, eliminates the need for off-line fractionation and reduces input requirements to as few as 1e6 A375 cells for > 800 distinct HLA-I peptides. This depth is sufficient to identify HLA-I peptides derived from cancer-testis antigen, and novel/unannotated open reading frames. We also apply our optimized single-shot SCP acquisition methods to tumor derived samples, enabling sensitive, high throughput and reproducible immunopeptidome profiling with detection of clinically relevant peptides from less than 4e7 cells or 15 mg wet weight tissue.
- Published
- 2023
16. Abstract P3-09-01: A ubiquitination cascade regulating the integrated stress response and survival in carcinomas
- Author
-
Lisa D Cervia, Tsukasa Shibue, Benjamin Gaeta, Ashir A Borah, Lisa Leung, Naomi Li, Nancy Dumont, Alfredo Gonzalez, Nolan Bick, Mariya Kazachkova, Joshua M Dempster, John M Krill-Burger, Federica Piccioni, Namrata D Udeshi, Meagan E Olive, Steven A Carr, David E Root, James M McFarland, Francisca Vazquez, and William C Hahn
- Subjects
Cancer Research ,Oncology - Abstract
Targeting of mutated oncogenes has led to the identification of new targeted therapies. However, druggable oncogenes do not occur in most cancers. Systematic identification of signaling pathways required for the fitness of cancer cells will facilitate the development of new cancer therapies. We used gene essentiality measurements in 793 cancer cell lines to identify selective co-essentiality modules and found that a ubiquitination ligase complex composed of UBA6, BIRC6, KCMF1 and UBR4, which encode an E1, E2 and two heterodimeric E3 subunits, respectively, is required for the survival of a subset of epithelial tumors, particularly subtypes of breast cancer. Suppressing BIRC6 in cell lines that are dependent on this complex led to a substantial reduction in cell fitness in vitro and potent tumor regression in vivo. Mechanistically, BIRC6 suppression resulted in selective activation of the integrated stress response (ISR) by stabilization and upregulation of the heme-regulated inhibitor (HRI), a direct ubiquitination target of the UBA6/BIRC6/KCMF1/UBR4 complex. These observations uncover a novel ubiquitination cascade that regulates ISR and highlight the potential of ISR activation as a new therapeutic strategy. Citation Format: Lisa D Cervia, Tsukasa Shibue, Benjamin Gaeta, Ashir A Borah, Lisa Leung, Naomi Li, Nancy Dumont, Alfredo Gonzalez, Nolan Bick, Mariya Kazachkova, Joshua M Dempster, John M Krill-Burger, Federica Piccioni, Namrata D Udeshi, Meagan E Olive, Steven A Carr, David E Root, James M McFarland, Francisca Vazquez, William C Hahn. A ubiquitination cascade regulating the integrated stress response and survival in carcinomas [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P3-09-01.
- Published
- 2022
17. Dynamic mapping of proteome trafficking within and between living cells by TransitID
- Author
-
Wei Qin Xu, Joleen S. Cheah, Charles Xu, James Messing, Brian D. Freibaum, Steven Boeynaems, J. Paul Taylor, Namrata D. Udeshi, Steven A. Carr, and Alice Y. Ting
- Subjects
Article - Abstract
SummaryThe ability to map trafficking for thousands of endogenous proteins at once in living cells would reveal biology currently invisible to both microscopy and mass spectrometry. Here we report TransitID, a method for unbiased mapping of endogenous proteome trafficking with nanometer spatial resolution in living cells. Two proximity labeling (PL) enzymes, TurboID and APEX, are targeted to source and destination compartments, and PL with each enzyme is performed in tandem via sequential addition of their small-molecule substrates. Mass spectrometry identifies the proteins tagged by both enzymes. Using TransitID, we mapped proteome trafficking between cytosol and mitochondria, cytosol and nucleus, and nucleolus and stress granules, uncovering a role for stress granules in protecting the transcription factor JUN from oxidative stress. TransitID also identifies proteins that signal intercellularly between macrophages and cancer cells. TransitID introduces a powerful approach for distinguishing protein populations based on compartment or cell type of origin.
- Published
- 2023
18. Arginine metabolism regulates human erythroid differentiation through hypusination of eIF5A
- Author
-
Pedro Gonzalez-Menendez, Ira Phadke, Meagan E Olive, Axel Joly, Julien Papoin, Hongxia Yan, Jérémy Galtier, Jessica Platon, Sun Woo Sophie Kang, Kathy L. McGraw, Marie Daumur, Marie Pouzolles, Taisuke Kondo, Stéphanie Boireau, Franciane Paul, David J Young, Sylvain Lamure, Raghavendra G Mirmira, Anu Narla, Guillaume Cartron, Cynthia E. Dunbar, Myriam Boyer-Clavel, Natalie Porat-Shliom, Valerie Dardalhon, Valerie S Zimmermann, Marc Sitbon, Thomas Dever, Narla Mohandas, Lydie M Da Costa, Namrata D. Udeshi, Lionel Blanc, Sandrina Kinet, and Naomi Taylor
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Abstract
Metabolic programs contribute to hematopoietic stem and progenitor cell (HSPC) fate, but it is not known whether the metabolic regulation of protein synthesis controls HSPC differentiation. Here, we show that SLC7A1/CAT1-dependent arginine uptake and its catabolism to the polyamine spermidine control human erythroid specification of HSPCs via activation of the eukaryotic translation initiation factor 5A (eIF5A). eIF5A activity is dependent on its hypusination, a post-translational modification resulting from the conjugation of the aminobutyl moiety of spermidine to lysine. Notably, attenuation of hypusine synthesis in erythroid progenitors--by inhibition of deoxyhypusine synthase--abrogates erythropoiesis but not myeloid cell differentiation. Proteomic profiling reveals mitochondrial translation to be a critical target of hypusinated eIF5A and accordingly, progenitors with decreased hypusine activity exhibit diminished oxidative phosphorylation. This impacted pathway is critical for eIF5A-regulated erythropoiesis as interventions augmenting mitochondrial function partially rescue human erythropoiesis under conditions of attenuated hypusination. Levels of mitochondrial ribosomal proteins were especially sensitive to the loss of hypusine and we find that the ineffective erythropoiesis linked to haploinsufficiency of RPS14 in del(5q) myelodysplastic syndrome is associated with a diminished pool of hypusinated eIF5A. Moreover, patients with RPL11-haploinsufficient Diamond-Blackfan anemia as well as CD34+ progenitors with downregulated RPL11 exhibit a markedly decreased hypusination in erythroid progenitors, concomitant with a loss of mitochondrial metabolism. Thus, eIF5A-dependent protein synthesis regulates human erythropoiesis and our data reveal a novel role for RPs in controlling eIF5A hypusination in HSPC, synchronizing mitochondrial metabolism with erythroid differentiation.
- Published
- 2023
19. A ubiquitination cascade regulating the integrated stress response and survival in carcinomas
- Author
-
Lisa D. Cervia, Tsukasa Shibue, Ashir A. Borah, Benjamin Gaeta, Linh He, Lisa Leung, Naomi Li, Sydney M. Moyer, Brian H. Shim, Nancy Dumont, Alfredo Gonzalez, Nolan R. Bick, Mariya Kazachkova, Joshua M. Dempster, John Michael Krill-Burger, Federica Piccioni, Namrata D. Udeshi, Meagan E. Olive, Steven A. Carr, David E. Root, James M. McFarland, Francisca Vazquez, and William C. Hahn
- Subjects
Oncology - Abstract
Systematic identification of signaling pathways required for the fitness of cancer cells will facilitate the development of new cancer therapies. We used gene essentiality measurements in 1,086 cancer cell lines to identify selective coessentiality modules and found that a ubiquitin ligase complex composed of UBA6, BIRC6, KCMF1, and UBR4 is required for the survival of a subset of epithelial tumors that exhibit a high degree of aneuploidy. Suppressing BIRC6 in cell lines that are dependent on this complex led to a substantial reduction in cell fitness in vitro and potent tumor regression in vivo. Mechanistically, BIRC6 suppression resulted in selective activation of the integrated stress response (ISR) by stabilization of the heme-regulated inhibitor, a direct ubiquitination target of the UBA6/BIRC6/KCMF1/UBR4 complex. These observations uncover a novel ubiquitination cascade that regulates ISR and highlight the potential of ISR activation as a new therapeutic strategy.Significance:We describe the identification of a heretofore unrecognized ubiquitin ligase complex that prevents the aberrant activation of the ISR in a subset of cancer cells. This provides a novel insight on the regulation of ISR and exposes a therapeutic opportunity to selectively eliminate these cancer cells.See related commentary Leli and Koumenis, p. 535.This article is highlighted in the In This Issue feature, p. 517
- Published
- 2022
20. Degradation of GSPT1 causes TP53-independent cell death in leukemia while sparing normal hematopoietic stem cells
- Author
-
Rob S. Sellar, Adam S. Sperling, Mikołaj Słabicki, Jessica A. Gasser, Marie E. McConkey, Katherine A. Donovan, Nada Mageed, Dylan N. Adams, Charles Zou, Peter G. Miller, Ravi K. Dutta, Steffen Boettcher, Amy E. Lin, Brittany Sandoval, Vanessa A. Quevedo Barrios, Veronica Kovalcik, Jonas Koeppel, Elizabeth K. Henderson, Emma C. Fink, Lu Yang, Anthony Chan, Sheela Pangeni Pokharel, Erik J. Bergstrom, Rajan Burt, Namrata D. Udeshi, Steven A. Carr, Eric S. Fischer, Chun-Wei Chen, and Benjamin L. Ebert
- Subjects
Mice ,Leukemia ,Cell Death ,Proteolysis ,Animals ,General Medicine ,Hematopoietic Stem Cells ,Peptide Termination Factors - Abstract
Targeted protein degradation is a rapidly advancing and expanding therapeutic approach. Drugs that degrade GSPT1 via the CRL4CRBN ubiquitin ligase are a new class of cancer therapy in active clinical development with evidence of activity against acute myeloid leukemia in early-phase trials. However, other than activation of the integrated stress response, the downstream effects of GSPT1 degradation leading to cell death are largely undefined, and no murine models are available to study these agents. We identified the domains of GSPT1 essential for cell survival and show that GSPT1 degradation leads to impaired translation termination, activation of the integrated stress response pathway, and TP53-independent cell death. CRISPR/Cas9 screens implicated decreased translation initiation as protective following GSPT1 degradation, suggesting that cells with higher levels of translation are more susceptible to the effects of GSPT1 degradation. We defined 2 Crbn amino acids that prevent Gspt1 degradation in mice, generated a knockin mouse with alteration of these residues, and demonstrated the efficacy of GSPT1-degrading drugs in vivo with relative sparing of numbers and function of long-term hematopoietic stem cells. Our results provide a mechanistic basis for the use of GSPT1 degraders for the treatment of cancer, including TP53-mutant acute myeloid leukemia.
- Published
- 2022
21. A genetic model for
- Author
-
Rui, Yang, Amanda S, Meyer, Ilia A, Droujinine, Namrata D, Udeshi, Yanhui, Hu, Jinjin, Guo, Jill A, McMahon, Dominique K, Carey, Charles, Xu, Qiao, Fang, Jihui, Sha, Shishang, Qin, David, Rocco, James, Wohlschlegel, Alice Y, Ting, Steven A, Carr, Norbert, Perrimon, and Andrew P, McMahon
- Subjects
Mammals ,Proteomics ,Mice ,Models, Genetic ,Animals ,Biotinylation ,Mass Spectrometry ,Secretome - Abstract
Organ functions are highly specialized and interdependent. Secreted factors regulate organ development and mediate homeostasis through serum trafficking and inter-organ communication. Enzyme-catalysed proximity labelling enables the identification of proteins within a specific cellular compartment. Here, we report a
- Published
- 2022
22. Dynamic regulation of inter-organelle communication by ubiquitylation controls skeletal muscle development and disease onset
- Author
-
Arian Mansur, Remi Joseph, Pierre Michael Jean-Beltran, Namrata D. Udeshi, Cadence Pearce, Hanjie Jiang, Reina Iwase, Elyshia McNamara, Jeff Widrick, Claudio Perez, Gianina Ravenscroft, Philip A Cole, Steven A Carr, and Vandana A Gupta
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Abstract
Ubiquitin-proteasome system (UPS) dysfunction is associated with the pathology of a wide range of human diseases including myopathies and muscular atrophy. However, the mechanistic understanding of specific components on the regulation of protein turnover during development and disease progression in skeletal muscle is unclear. Mutations in KLHL40, an E3 ubiquitin ligase cullin3 (CUL3) substrate-specific adapter protein result in a severe form of congenital nemaline myopathy, but the events that initiate the pathology and the mechanism through which it becomes pervasive, remains poorly understood. To characterize the KLHL40-regulated ubiquitin modified proteome during skeletal muscle development and disease onset, we used global, quantitative mass spectrometry-based ubiquitylome and global proteome analyses of klhl40 mutant zebrafish during disease progression. Global proteomics during skeletal muscle development revealed extensive remodeling of functional modules linked with sarcomere formation, energy and biosynthetic metabolic processes and vesicle trafficking. Combined analysis of klh40 mutant muscle proteome and ubiquitylome identified thin filament proteins, metabolic enzymes and ER-Golgi vesicle trafficking pathway proteins regulated by ubiquitylation during muscle development. Our studies identified a role for KLHL40 as a negative regulator of ER-Golgi anterograde trafficking through ubiquitin-mediated protein degradation of secretion associated Ras related GTPase1a (Sar1a). In KLHL40 deficient muscle, defects in ER exit site vesicle formation alter Golgi compartment and downstream transport of extracellular cargo proteins, resulting in structural and functional abnormalities. Our work reveals that the muscle proteome is dynamically fine-tuned by ubiquitylation to regulate skeletal muscle development and uncovers new disease mechanisms for therapeutic development in patients.Graphical Abstract
- Published
- 2022
23. CBL mutations drive PI3K/AKT signaling via increased interaction with LYN and PIK3R1
- Author
-
Christina R. Hartigan, Benjamin L. Ebert, Caroline Stanclift, Namrata D. Udeshi, Monica Schenone, Amanuel Bizuayehu, Veronica Kovalcik, Tanya Svinkina, Alexis Vedder, Marie McConkey, Eric Padron, Sebastian Koochaki, Roger Belizaire, Steven A. Carr, and Lei Sun
- Subjects
Immunology ,medicine.disease_cause ,environment and public health ,Biochemistry ,Phosphatidylinositol 3-Kinases ,LYN ,PIK3R1 ,hemic and lymphatic diseases ,medicine ,Humans ,Protein Interaction Maps ,Proto-Oncogene Proteins c-cbl ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Mutation ,Myeloid Neoplasia ,biology ,Chemistry ,fungi ,Cell Biology ,Hematology ,Ubiquitin ligase ,Class Ia Phosphatidylinositol 3-Kinase ,enzymes and coenzymes (carbohydrates) ,src-Family Kinases ,Hematologic Neoplasms ,biology.protein ,Cancer research ,Phosphorylation ,biological phenomena, cell phenomena, and immunity ,Proto-Oncogene Proteins c-akt ,Tyrosine kinase ,Signal Transduction - Abstract
Casitas B-lineage lymphoma (CBL) encodes an E3 ubiquitin ligase and signaling adaptor that regulates receptor and nonreceptor tyrosine kinases. Recurrent CBL mutations occur in myeloid neoplasms, including 10% to 20% of chronic myelomonocytic leukemia (CMML) cases, and selectively disrupt the protein’s E3 ubiquitin ligase activity. CBL mutations have been associated with poor prognosis, but the oncogenic mechanisms and therapeutic implications of CBL mutations remain incompletely understood. We combined functional assays and global mass spectrometry to define the phosphoproteome, CBL interactome, and mechanism of signaling activation in a panel of cell lines expressing an allelic series of CBL mutations. Our analyses revealed that increased LYN activation and interaction with mutant CBL are key drivers of enhanced CBL phosphorylation, phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1) recruitment, and downstream phosphatidylinositol 3-kinase (PI3K)/AKT signaling in CBL-mutant cells. Signaling adaptor domains of CBL, including the tyrosine kinase–binding domain, proline-rich region, and C-terminal phosphotyrosine sites, were all required for the oncogenic function of CBL mutants. Genetic ablation or dasatinib-mediated inhibition of LYN reduced CBL phosphorylation, CBL-PIK3R1 interaction, and PI3K/AKT signaling. Furthermore, we demonstrated in vitro and in vivo antiproliferative efficacy of dasatinib in CBL-mutant cell lines and primary CMML. Overall, these mechanistic insights into the molecular function of CBL mutations provide rationale to explore the therapeutic potential of LYN inhibition in CBL-mutant myeloid malignancies.
- Published
- 2021
24. Proteomics of protein trafficking by in vivo tissue-specific labeling
- Author
-
Jinjin Guo, Tanya Svinkina, Alice Y. Ting, Namrata D. Udeshi, Luye Mu, Dan Wang, John M. Asara, David Rocco, Steven A. Carr, Dominique K. Carey, Amanda S. Meyer, Yanhui Hu, Rui Yang, Jill A. McMahon, Tess C. Branon, Andrew P. McMahon, Ilia A. Droujinine, Norbert Perrimon, Areya Tabatabai, Rebecca Zeng, and Justin A. Bosch
- Subjects
0301 basic medicine ,Male ,Proteomics ,Science ,General Physics and Astronomy ,Biotin ,Proteomic analysis ,Biology ,Protein Engineering ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,Animals, Genetically Modified ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Tandem Mass Spectrometry ,Compartment (development) ,Animals ,Humans ,Biotinylation ,Carbon-Nitrogen Ligases ,Embryonic Stem Cells ,chemistry.chemical_classification ,DNA ligase ,Multidisciplinary ,Staining and Labeling ,Escherichia coli Proteins ,Teratoma ,Embryo ,General Chemistry ,Blood proteins ,Cell biology ,Repressor Proteins ,Disease Models, Animal ,Protein Transport ,Circulation ,030104 developmental biology ,Secretory protein ,chemistry ,Drosophila ,Female ,Extracellular signalling molecules ,030217 neurology & neurosurgery - Abstract
Conventional approaches to identify secreted factors that regulate homeostasis are limited in their abilities to identify the tissues/cells of origin and destination. We established a platform to identify secreted protein trafficking between organs using an engineered biotin ligase (BirA*G3) that biotinylates, promiscuously, proteins in a subcellular compartment of one tissue. Subsequently, biotinylated proteins are affinity-enriched and identified from distal organs using quantitative mass spectrometry. Applying this approach in Drosophila, we identify 51 muscle-secreted proteins from heads and 269 fat body-secreted proteins from legs/muscles, including CG2145 (human ortholog ENDOU) that binds directly to muscles and promotes activity. In addition, in mice, we identify 291 serum proteins secreted from conditional BirA*G3 embryo stem cell-derived teratomas, including low-abundance proteins with hormonal properties. Our findings indicate that the communication network of secreted proteins is vast. This approach has broad potential across different model systems to identify cell-specific secretomes and mediators of interorgan communication in health or disease., The network of proteins secreted for interorgan communication is poorly understood. Here, the authors develop a method, based on protein labeling, to study cell-specific secretomes and interorgan protein trafficking, and demonstrate their approach in Drosophila and mouse models.
- Published
- 2021
25. In situ cell-type-specific cell-surface proteomic profiling in mice
- Author
-
S. Andrew Shuster, Jiefu Li, URee Chon, Miley C. Sinantha-Hu, David J. Luginbuhl, Namrata D. Udeshi, Dominique Kiki Carey, Yukari H. Takeo, Qijing Xie, Chuanyun Xu, D.R. Mani, Shuo Han, Alice Y. Ting, Steven A. Carr, and Liqun Luo
- Subjects
Proteomics ,Mammals ,Mice ,General Neuroscience ,Animals - Abstract
Cell-surface proteins (CSPs) mediate intercellular communication throughout the lives of multicellular organisms. However, there are no generalizable methods for quantitative CSP profiling in specific cell types in vertebrate tissues. Here, we present in situ cell-surface proteome extraction by extracellular labeling (iPEEL), a proximity labeling method in mice that enables spatiotemporally precise labeling of cell-surface proteomes in a cell-type-specific environment in native tissues for discovery proteomics. Applying iPEEL to developing and mature cerebellar Purkinje cells revealed differential enrichment in CSPs with post-translational protein processing and synaptic functions in the developing and mature cell-surface proteomes, respectively. A proteome-instructed in vivo loss-of-function screen identified a critical, multifaceted role for Armh4 in Purkinje cell dendrite morphogenesis. Armh4 overexpression also disrupts dendrite morphogenesis; this effect requires its conserved cytoplasmic domain and is augmented by disrupting its endocytosis. Our results highlight the utility of CSP profiling in native mammalian tissues for identifying regulators of cell-surface signaling.
- Published
- 2022
26. A genetic model for in vivo proximity labeling of the mammalian secretome
- Author
-
Rui Yang, Amanda S. Meyer, Ilia A. Droujinine, Namrata D. Udeshi, Yanhui Hu, Jinjin Guo, Jill A. McMahon, Dominique K. Carey, Charles Xu, Qiao Fang, Jihui Sha, Shishang Qin, David Rocco, James Wohlschlegel, Alice Y. Ting, Steven A. Carr, Norbert Perrimon, and Andrew P. McMahon
- Abstract
Organ functions are highly specialized and interdependent. Secreted factors regulate organ development and mediate homeostasis through serum trafficking and inter-organ communication. Enzyme-catalyzed proximity labeling enables the identification of proteins within a specific cellular compartment. Here, we report a BirA*G3 mouse strain that enables CRE-dependent promiscuous biotinylation of proteins trafficking through the endoplasmic reticulum. When broadly activated throughout the mouse, widespread labeling of proteins was observed within the secretory pathway. Streptavidin affinity purification and peptide mapping by quantitative mass spectrometry (MS) proteomics revealed organ-specific secretory profiles and serum trafficking. As expected, secretory proteomes were highly enriched for signal peptide-containing proteins, highlighting both conventional and non-conventional secretory processes, and ectodomain shedding. Lower-abundance proteins with hormone-like properties were recovered and validated using orthogonal approaches. Hepatocyte-specific activation of BirA*G3 highlighted liver-specific biotinylated secretome profiles. The BirA*G3 mouse model demonstrates enhanced labeling efficiency and tissue specificity over viral transduction approaches and will facilitate a deeper understanding of secretory protein interplay in development, and healthy and diseased adult states.
- Published
- 2022
27. Arginine-dependent hypusination of the eukaryotic translation initiation factor (eIF)5A drives erythroid lineage differentiation
- Author
-
Pedro Gonzalez-Menendez, Ira Phadke, Meagan E. Olive, Kathy L. McGraw, Jessica Platon, Julien Papoin, Hongxia Yan, Marie Daumur, Franciane Paul, Raghavendra Mirmira, Axel Joly, Jeremy Galtier, Anupama Narla, Guillaume Cartron, Valérie Dardalhon, Valérie S. Zimmermann, Marc Sitbon, Thomas E. Dever, Narla Mohandas, Lydie da Costa, Namrata D. Udeshi, Lionel Blanc, Sandrina Kinet, and Naomi Taylor
- Abstract
Metabolic programs contribute to hematopoietic stem and progenitor cell (HSPC) fate but it is not known whether the metabolic regulation of protein synthesis controls HSPC differentiation. We discovered that SLC7A1/CAT1-dependent arginine uptake and its catabolism to spermidine control the erythroid specification of HSPCs via activation of eukaryotic translation initiation factor 5A (eIF5A). eIF5A activity is dependent on the metabolism of spermidine to hypusine and inhibiting hypusine synthesis abrogates erythropoiesis and diverts EPO-stimulated HSPCs to a myeloid fate. Proteomic profiling reveals mitochondrial translation to be a critical target of hypusinated eIF5A and induction of mitochondrial function partially rescues erythropoiesis in the absence of hypusine. Within the hypusine network, ribosomal proteins are highly enriched and we identify defective eIF5A hypusination in erythroid pathologies caused by abnormal ribosome biogenesis. Thus, eIF5A-dependent protein synthesis is critical in the branching of erythro-myeloid differentiation and attenuated eIF5A activity characterizes ribosomal protein-linked disorders of ineffective erythropoiesis.
- Published
- 2022
28. Proximity labeling in mammalian cells with TurboID and split-TurboID
- Author
-
Kelvin F. Cho, Namrata D. Udeshi, Alice Y. Ting, Steven A. Carr, Tess C. Branon, and Samuel A. Myers
- Subjects
chemistry.chemical_classification ,Streptavidin ,0303 health sciences ,DNA ligase ,Computational biology ,Directed evolution ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Biotin ,chemistry ,In vivo ,Biotinylation ,Proteome ,Organelle ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
This protocol describes the use of TurboID and split-TurboID in proximity labeling applications for mapping protein–protein interactions and subcellular proteomes in live mammalian cells. TurboID is an engineered biotin ligase that uses ATP to convert biotin into biotin–AMP, a reactive intermediate that covalently labels proximal proteins. Optimized using directed evolution, TurboID has substantially higher activity than previously described biotin ligase–related proximity labeling methods, such as BioID, enabling higher temporal resolution and broader application in vivo. Split-TurboID consists of two inactive fragments of TurboID that can be reconstituted through protein–protein interactions or organelle–organelle interactions, which can facilitate greater targeting specificity than full-length enzymes alone. Proteins biotinylated by TurboID or split-TurboID are then enriched with streptavidin beads and identified by mass spectrometry. Here, we describe fusion construct design and characterization (variable timing), proteomic sample preparation (5–7 d), mass spectrometric data acquisition (2 d), and proteomic data analysis (1 week). This protocol describes proximity labeling approaches using TurboID and split-TurboID, which can be used for mapping protein–protein interactions and organelle proteomes in live mammalian cells with nanometer spatial resolution.
- Published
- 2020
29. Multi-Omics Analysis Identifies MGA as a Negative Regulator of the MYC Pathway in Lung Adenocarcinoma
- Author
-
Jin Zhou, Joshua M. Francis, Montse Sanchez-Cespedes, Xiaoyang Zhang, Matthew Meyerson, Steven A. Carr, Tanya Svinkina, Estrella Aguilera-Jimenez, Namrata D. Udeshi, Manuel Torres-Diz, Diana Cai, Paula Llabata, Yoichiro Mitsuishi, Zhong Wu, Peter S. Choi, Lior Golomb, and Yanli Liu
- Subjects
Cancer Research ,Lung Neoplasms ,Adenocarcinoma of Lung ,Protein Max ,Biology ,Article ,Receptor tyrosine kinase ,law.invention ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,0302 clinical medicine ,law ,Cell Line, Tumor ,Basic Helix-Loop-Helix Transcription Factors ,Transcriptional regulation ,Humans ,Molecular Biology ,Gene ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,RNA ,Chromatin ,HEK293 Cells ,Oncology ,A549 Cells ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,biology.protein ,Suppressor ,Sequence motif - Abstract
Genomic analysis of lung adenocarcinomas has revealed that the MGA gene, which encodes a heterodimeric partner of the MYC-interacting protein MAX, is significantly mutated or deleted in lung adenocarcinomas. Most of the mutations are loss of function for MGA, suggesting that MGA may act as a tumor suppressor. Here, we characterize both the molecular and cellular role of MGA in lung adenocarcinomas and illustrate its functional relevance in the MYC pathway. Although MGA and MYC interact with the same binding partner, MAX, and recognize the same E-box DNA motif, we show that the molecular function of MGA appears to be antagonistic to that of MYC. Using mass spectrometry–based affinity proteomics, we demonstrate that MGA interacts with a noncanonical PCGF6-PRC1 complex containing MAX and E2F6 that is involved in gene repression, while MYC is not part of this MGA complex, in agreement with previous studies describing the interactomes of E2F6 and PCGF6. Chromatin immunoprecipitation-sequencing and RNA sequencing assays show that MGA binds to and represses genes that are bound and activated by MYC. In addition, we show that, as opposed to the MYC oncoprotein, MGA acts as a negative regulator for cancer cell proliferation. Our study defines a novel MYC/MAX/MGA pathway, in which MYC and MGA play opposite roles in protein interaction, transcriptional regulation, and cellular proliferation. Implications: This study expands the range of key cancer-associated genes whose dysregulation is functionally equivalent to MYC activation and places MYC within a linear pathway analogous to cell-cycle or receptor tyrosine kinase/RAS/RAF pathways in lung adenocarcinomas.
- Published
- 2020
30. Transcription Factor Control of Dendrite Targeting via Combinatorial Cell-Surface Codes
- Author
-
Hongjie Li, S Shuster, Tongchao Li, Qijing Xie, Ricardo Guajardo, David Luginbuhl, Chuanyun Xu, Liqun Luo, Tanya Svinkina, Steven A. Carr, Namrata D. Udeshi, Alice Y. Ting, D. R. Mani, Shuo Han, Wei Wei, Sayeh Kohani, and Jiefu Li
- Subjects
Mechanosensitive ion channel ,Mutant ,Cell fate determination ,Biology ,Cell adhesion ,Phenotype ,Transcription factor ,Function (biology) ,Cell biology ,Genetic screen - Abstract
Transcription factors are central commanders specifying cell fate, morphology, and physiology while cell-surface proteins execute these commands through interaction with cellular environment. In developing neurons, it is presumed that transcription factors control wiring specificity through regulation of cell-surface protein expression. However, the number and identity of cell-surface protein(s) a transcription factor regulates remain largely unclear1,2. Also unknown is whether a transcription factor regulates the same or different cell-surface proteins in different neuron types to specify their connectivity. Here we use a lineage-defining transcription factor, Acj6 (ref. 3), to investigate how it controls precise dendrite targeting of Drosophila olfactory projection neurons (PNs). Quantitative cell-surface proteomic profiling of wild-type and acj6 mutant PNs in intact developing brains and a proteome-informed genetic screen identified PN surface proteins that execute Acj6-regulated wiring decisions. These include canonical cell adhesion proteins and proteins previously not associated with wiring, such as the mechanosensitive ion channel Piezo—whose channel activity is dispensable for its wiring function. Comprehensive genetic analyses revealed that Acj6 employs unique sets of cell-surface proteins in different PN types for dendrite targeting. Combinatorial expression of Acj6 wiring executors rescued acj6 mutant phenotypes with higher efficacy and breadth than expression of individual executors. Thus, a key transcription factor controls wiring specificity of different neuron types by specifying distinct combinatorial expression of cell-surface executors.
- Published
- 2021
31. Transcription Factor Acj6 Controls Dendrite Targeting via Combinatorial Cell-Surface Codes
- Author
-
Qijing Xie, Jiefu Li, Hongjie Li, Namrata D Udeshi, Tanya Svinkina, Daniel Orlin, Sayeh Kohani, Ricardo Guajardo, DR Mani, Chuanyun Xu, Tongchao Li, Shuo Han, Wei Wei, S Andrew Shuster, David J Luginbuhl, Stephen R. Quake, Swetha E. Murthy, Alice Y Ting, Steven A Carr, and Liqun Luo
- Abstract
SUMMARYTranscription factors specify the fate and connectivity of developing neurons. We investigate how a lineage-specific transcription factor, Acj6, controls the precise dendrite targeting of Drosophila olfactory projection neurons (PNs) by regulating the expression of cell-surface proteins. Quantitative cell-surface proteomic profiling of wild-type and acj6 mutant PNs in intact developing brains and a proteome-informed genetic screen identified PN surface proteins that execute Acj6-regulated wiring decisions. These include canonical cell adhesion molecules and proteins previously not associated with wiring, such as Piezo, whose mechanosensitive ion channel activity is dispensable for its function in PN dendrite targeting. Comprehensive genetic analyses revealed that Acj6 employs unique sets of cell-surface proteins in different PN types for dendrite targeting. Combinatorial expression of Acj6 wiring executors rescued acj6 mutant phenotypes with higher efficacy and breadth than expression of individual executors. Thus, Acj6 controls wiring specificity of different neuron types by specifying distinct combinatorial expression of cell- surface executors.
- Published
- 2021
32. An engineered transcriptional reporter of protein localization identifies regulators of mitochondrial and ER membrane protein trafficking in high-throughput CRISPRi screens
- Author
-
Jonathan S. Weissman, Eric T. Strand, Robert Coukos, Michael C. Bassik, Meagan E. Olive, Mateo Lopez Sanchez, David Yao, Namrata D. Udeshi, Alice Y. Ting, and Steven A. Carr
- Subjects
QH301-705.5 ,Science ,Chemical biology ,Ubiquitin-Activating Enzymes ,Biology ,Endoplasmic Reticulum ,General Biochemistry, Genetics and Molecular Biology ,Biochemistry and Chemical Biology ,Humans ,Clustered Regularly Interspaced Short Palindromic Repeats ,tail-anchored proteins ,Biology (General) ,high-throughput screens ,Gene ,Transcription factor ,Reporter gene ,General Immunology and Microbiology ,General Neuroscience ,Membrane Proteins ,protein engineering ,Cell Biology ,General Medicine ,Protein engineering ,Cell sorting ,Flow Cytometry ,Phenotype ,Protein subcellular localization prediction ,Mitochondria ,Cell biology ,Protein Transport ,HEK293 Cells ,CRISPR ,Medicine ,K562 Cells ,Research Article ,Human ,HeLa Cells ,Signal Transduction - Abstract
The trafficking of specific protein cohorts to correct subcellular locations at correct times is essential for every signaling and regulatory process in biology. Gene perturbation screens could provide a powerful approach to probe the molecular mechanisms of protein trafficking, but only if protein localization or mislocalization can be tied to a simple and robust phenotype for cell selection, such as cell proliferation or fluorescence-activated cell sorting (FACS). To empower the study of protein trafficking processes with gene perturbation, we developed a genetically encoded molecular tool named HiLITR (High-throughput Localization Indicator with Transcriptional Readout). HiLITR converts protein colocalization into proteolytic release of a membrane-anchored transcription factor, which drives the expression of a chosen reporter gene. Using HiLITR in combination with FACS-based CRISPRi screening in human cell lines, we identified genes that influence the trafficking of mitochondrial and ER tail-anchored proteins. We show that loss of the SUMO E1 component SAE1 results in mislocalization and destabilization of many mitochondrial tail-anchored proteins. We also demonstrate a distinct regulatory role for EMC10 in the ER membrane complex, opposing the transmembrane-domain insertion activity of the complex. Through transcriptional integration of complex cellular functions, HiLITR expands the scope of biological processes that can be studied by genetic perturbation screening technologies.
- Published
- 2021
33. Author response: An engineered transcriptional reporter of protein localization identifies regulators of mitochondrial and ER membrane protein trafficking in high-throughput CRISPRi screens
- Author
-
David Yao, Robert Coukos, Mateo I Sanchez, Eric T Strand, Meagan E Olive, Namrata D Udeshi, Jonathan S Weissman, Steven A Carr, Michael C Bassik, and Alice Y Ting
- Published
- 2021
34. RNF43 G659fs is an oncogenic colorectal cancer mutation and sensitizes tumor cells to PI3K/mTOR inhibition
- Author
-
Lishan Fang, Dane Ford-Roshon, Max Russo, Casey O’Brien, Xiaozhe Xiong, Carino Gurjao, Maximilien Grandclaudon, Srivatsan Raghavan, Steven M. Corsello, Steven A. Carr, Namrata D. Udeshi, James Berstler, Ewa Sicinska, Kimmie Ng, and Marios Giannakis
- Subjects
Phosphatidylinositol 3-Kinases ,Multidisciplinary ,Cell Line, Tumor ,TOR Serine-Threonine Kinases ,Ubiquitin-Protein Ligases ,Mutation ,General Physics and Astronomy ,Humans ,General Chemistry ,Colorectal Neoplasms ,General Biochemistry, Genetics and Molecular Biology - Abstract
The RNF43_p.G659fs mutation occurs frequently in colorectal cancer, but its function remains poorly understood and there are no specific therapies directed against this alteration. In this study, we find that RNF43_p.G659fs promotes cell growth independent of Wnt signaling. We perform a drug repurposing library screen and discover that cells with RNF43_p.G659 mutations are selectively killed by inhibition of PI3K signaling. PI3K/mTOR inhibitors yield promising antitumor activity in RNF43659mut isogenic cell lines and xenograft models, as well as in patient-derived organoids harboring RNF43_p.G659fs mutations. We find that RNF43659mut binds p85 leading to increased PI3K signaling through p85 ubiquitination and degradation. Additionally, RNA-sequencing of RNF43659mut isogenic cells reveals decreased interferon response gene expression, that is reversed by PI3K/mTOR inhibition, suggesting that RNF43659mut may alter tumor immunity. Our findings suggest a therapeutic application for PI3K/mTOR inhibitors in treating RNF43_p.G659fs mutant cancers.
- Published
- 2021
35. MONTE enables serial immunopeptidome, ubiquitylome, proteome, phosphoproteome, acetylome analyses of sample-limited tissues
- Author
-
Jennifer G. Abelin, White Cj, Namrata D. Udeshi, Erik J. Bergstrom, Karl R. Clauser, Pearce C, Meagan E. Olive, Rivera Kd, Shankha Satpathy, Steven A. Carr, Michael A. Gillette, Maynard M, Susan Klaeger, Kane Mh, Rachimi S, and Taylor Hb
- Subjects
Biological pathway ,Cell signaling ,Proteome ,Context (language use) ,Human leukocyte antigen ,Computational biology ,Epigenetics ,Biology ,Protein degradation ,Function (biology) - Abstract
Multiomic characterization of patient tissues provides insights into the function of different biological pathways in the context of disease. Much work has been done to serialize proteome and post-translational modification (PTM) analyses to conserve precious patient samples. However, characterizing clinically relevant tissues with multi-ome workflows that have distinct sample processing requirements remains challenging. To overcome the obstacles of combining enrichment workflows that have unique input amounts and utilize both label free and chemical labeling strategies, we developed a highly-sensitive multi-omic networked tissue enrichment (MONTE) workflow for the full analysis of HLA-I and HLA-II immunopeptidome, ubiquitylome, proteome, phosphoproteome and acetylome all from the same tissue sample. The MONTE workflow enables identification of a median of 9,000 HLA-I peptides, 6,000 HLA-II peptides, 10,000 Ub sites, 12,000 proteins, 20,000 phosphorylation sites and 15,000 acetylation sites from patient LUAD tumors. Because all omes are generated from the exact same tissue sample, there is less biological variability in the data enabling more robust integration. The information available in MONTE datasets facilitates the identification of putative immunotherapeutic targets, such as CT antigens and neoantigens presented by HLA complexes, as well as reveal insights into how disease-specific changes in protein expression, protein degradation, cell signaling, metabolic, and epigenetic pathways are involved in disease pathology and treatment.
- Published
- 2021
36. Transcription factor Acj6 controls dendrite targeting via a combinatorial cell-surface code
- Author
-
Qijing Xie, Jiefu Li, Hongjie Li, Namrata D. Udeshi, Tanya Svinkina, Daniel Orlin, Sayeh Kohani, Ricardo Guajardo, D.R. Mani, Chuanyun Xu, Tongchao Li, Shuo Han, Wei Wei, S. Andrew Shuster, David J. Luginbuhl, Stephen R. Quake, Swetha E. Murthy, Alice Y. Ting, Steven A. Carr, and Liqun Luo
- Subjects
Proteomics ,General Neuroscience ,POU Domain Factors ,Animals ,Drosophila Proteins ,Membrane Proteins ,Drosophila ,Nerve Tissue Proteins ,Dendrites ,Olfactory Pathways ,Ion Channels ,Olfactory Receptor Neurons ,Transcription Factors - Abstract
Transcription factors specify the fate and connectivity of developing neurons. We investigate how a lineage-specific transcription factor, Acj6, controls the precise dendrite targeting of Drosophila olfactory projection neurons (PNs) by regulating the expression of cell-surface proteins. Quantitative cell-surface proteomic profiling of wild-type and acj6 mutant PNs in intact developing brains, and a proteome-informed genetic screen identified PN surface proteins that execute Acj6-regulated wiring decisions. These include canonical cell adhesion molecules and proteins previously not associated with wiring, such as Piezo, whose mechanosensitive ion channel activity is dispensable for its function in PN dendrite targeting. Comprehensive genetic analyses revealed that Acj6 employs unique sets of cell-surface proteins in different PN types for dendrite targeting. Combined expression of Acj6 wiring executors rescued acj6 mutant phenotypes with higher efficacy and breadth than expression of individual executors. Thus, Acj6 controls wiring specificity of different neuron types by specifying distinct combinatorial expression of cell-surface executors.
- Published
- 2022
37. Abstract 73: A ubiquitination cascade regulating the integrated stress response and survival in carcinomas
- Author
-
Lisa D. Cervia, Tsukasa Shibue, Benjamin Gaeta, Ashir A. Borah, Lisa Leung, Naomi Li, Nancy Dumont, Alfredo Gonzalez, Nolan Bick, Mariya Kazachkova, Joshua M. Dempster, John M. Krill-Burger, Federica Piccioni, Namrata D. Udeshi, Meagan E. Olive, Steven A. Carr, David E. Root, James M. McFarland, Francisca Vazquez, and William C. Hahn
- Subjects
Cancer Research ,Oncology - Abstract
Targeting of mutated oncogenes has led to the identification of new targeted therapies. However, druggable oncogenes do not occur in most cancers. Systematic identification of signaling pathways required for the fitness of cancer cells will facilitate the development of new cancer therapies. We used gene essentiality measurements in 793 cancer cell lines to identify selective co-essentiality modules and found that a ubiquitination ligase complex composed of UBA6, BIRC6, KCMF1 and UBR4, which encode an E1, E2 and two heterodimeric E3 subunits, respectively, is required for the survival of a subset of epithelial tumors. Suppressing BIRC6 in cell lines that are dependent on this complex led to a substantial reduction in cell fitness in vitro and potent tumor regression in vivo. Mechanistically, BIRC6 suppression resulted in selective activation of the integrated stress response (ISR) by stabilization and upregulation of the heme-regulated inhibitor (HRI), a direct ubiquitination target of the UBA6/BIRC6/KCMF1/UBR4 complex. These observations uncover a novel ubiquitination cascade that regulates ISR and highlight the potential of ISR activation as a new therapeutic strategy. Citation Format: Lisa D. Cervia, Tsukasa Shibue, Benjamin Gaeta, Ashir A. Borah, Lisa Leung, Naomi Li, Nancy Dumont, Alfredo Gonzalez, Nolan Bick, Mariya Kazachkova, Joshua M. Dempster, John M. Krill-Burger, Federica Piccioni, Namrata D. Udeshi, Meagan E. Olive, Steven A. Carr, David E. Root, James M. McFarland, Francisca Vazquez, William C. Hahn. A ubiquitination cascade regulating the integrated stress response and survival in carcinomas [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 73.
- Published
- 2022
38. Avadomide induces degradation of ZMYM2 fusion oncoproteins in hematologic malignancies
- Author
-
Andrew A Guirguis, Benjamin L. Ebert, Sophie Cotteret, Marie McConkey, Rob S. Sellar, Mary E Matyskiela, Jessica A. Gasser, Christophe Marzac, Aline Renneville, Steven A. Carr, Alexander Tepper, Mark Rolfe, Namrata D. Udeshi, Kaushik Viswanathan, Stéphane de Botton, Thomas Clayton, Jean-Michel Cayuela, Philip P Chamberlain, Véronique Saada, Jean-Jacques Kiladjian, Pierre M. Jean Beltran, and Daniel E Grinshpun
- Subjects
Zinc finger ,Oncogene Proteins ,biology ,Chemistry ,General Medicine ,Fusion protein ,IKZF3 ,In vitro ,Article ,Ubiquitin ligase ,Thalidomide ,DNA-Binding Proteins ,Ubiquitin ,In vivo ,Hematologic Neoplasms ,biology.protein ,Cancer research ,Humans ,Transcription factor ,Lenalidomide ,Transcription Factors - Abstract
Thalidomide analogues exert their therapeutic effects by binding to the CRL4CRBN E3 ubiquitin ligase, promoting ubiquitination and subsequent proteasomal degradation of specific protein substrates. Drug-induced degradation of IKZF1 and IKZF3 in B-cell malignancies demonstrates the clinical utility of targeting disease-relevant transcription factors for degradation. Here, we found that avadomide (CC-122) induces CRBN-dependent ubiquitination and proteasomal degradation of ZMYM2 (ZNF198), a transcription factor involved in balanced chromosomal rearrangements with FGFR1 and FLT3 in aggressive forms of hematologic malignancies. The minimal drug-responsive element of ZMYM2 is a zinc-chelating MYM domain and is contained in the N-terminal portion of ZMYM2 that is universally included in the derived fusion proteins. We demonstrate that avadomide has the ability to induce proteasomal degradation of ZMYM2–FGFR1 and ZMYM2–FLT3 chimeric oncoproteins, both in vitro and in vivo. Our findings suggest that patients with hematologic malignancies harboring these ZMYM2 fusion proteins may benefit from avadomide treatment. Significance: We extend the potential clinical scope of thalidomide analogues by the identification of a novel avadomide-dependent CRL4CRBN substrate, ZMYM2. Avadomide induces ubiquitination and degradation of ZMYM2–FGFR1 and ZMYM2–FLT3, two chimeric oncoproteins involved in hematologic malignancies, providing a proof of concept for drug-induced degradation of transcription factor fusion proteins by thalidomide analogues.
- Published
- 2021
39. Dual functions of SPOP and ERG dictate androgen therapy responses in prostate cancer
- Author
-
Francesco Bertoni, Steven A. Carr, Manuela Cavalli, Holger Moch, Anna Rinaldi, Arianna Vallerga, Azzurra Mutti, Tanya Svinkina, Ze Dong, Hana Janouskova, Geniver El Tekle, Tiziano Bernasocchi, Simon Linder, Giuseppina M. Carbone, Daniela Bossi, Marianna Kruithof-de Julio, Roger Geiger, Laura P. Brandt, Wilbert Zwart, Andrea Rinaldi, Simone Mosole, Marita Zoma, Marco Bolis, Jean-Philippe Theurillat, Filippo Spriano, Andrea Alimonti, Peter Schraml, Cai-Guang Yang, Domenico Albino, Valentina Ceserani, Namrata D. Udeshi, and Mark A. Rubin
- Subjects
Proteomics ,Male ,Cancer therapy ,medicine.drug_class ,Science ,Mice, Nude ,610 Medicine & health ,Cell Cycle Proteins ,SPOP ,medicine.disease_cause ,Article ,Tumour biomarkers ,Prostate cancer ,Mice ,Transcriptional Regulator ERG ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Immunoprecipitation ,Transcription factor ,Cancer genetics ,Oncogene Proteins ,biology ,Nuclear Proteins ,Prostatic Neoplasms ,Ubiquitin-Protein Ligase Complexes ,medicine.disease ,Androgen ,Immunohistochemistry ,Ubiquitin ligase ,Androgen receptor ,DNA-Binding Proteins ,Repressor Proteins ,HEK293 Cells ,Androgen Therapy ,Receptors, Androgen ,Mutation ,biology.protein ,Cancer research ,Carcinogenesis ,610 Medizin und Gesundheit ,Co-Repressor Proteins ,Protein Binding ,Signal Transduction - Abstract
Driver genes with a mutually exclusive mutation pattern across tumor genomes are thought to have overlapping roles in tumorigenesis. In contrast, we show here that mutually exclusive prostate cancer driver alterations involving the ERG transcription factor and the ubiquitin ligase adaptor SPOP are synthetic sick. At the molecular level, the incompatible cancer pathways are driven by opposing functions in SPOP. ERG upregulates wild type SPOP to dampen androgen receptor (AR) signaling and sustain ERG activity through degradation of the bromodomain histone reader ZMYND11. Conversely, SPOP-mutant tumors stabilize ZMYND11 to repress ERG-function and enable oncogenic androgen receptor signaling. This dichotomy regulates the response to therapeutic interventions in the AR pathway. While mutant SPOP renders tumor cells susceptible to androgen deprivation therapies, ERG promotes sensitivity to high-dose androgen therapy and pharmacological inhibition of wild type SPOP. More generally, these results define a distinct class of antagonistic cancer drivers and a blueprint toward their therapeutic exploitation., Nature Communications, 12, ISSN:2041-1723
- Published
- 2021
- Full Text
- View/download PDF
40. A proteogenomic portrait of lung squamous cell carcinoma
- Author
-
Shuang Cai, Elizabeth R. Duffy, Felipe da Veiga Leprevost, D. R. Mani, Antonio Colaprico, Jiayi Ji, Mehdi Mesri, Alicia Francis, Peter B. McGarvey, Myvizhi Esai Selvan, Corbin D. Jones, Michael J. Birrer, Robert J. Welsh, Lori Bernard, Shankha Satpathy, Li Ding, Sara R. Savage, Eugene S. Fedorov, Fernanda Martins Rodrigues, Marcin J. Domagalski, Jennifer M. Eschbacher, Shayan C. Avanessian, Boris Reva, Harsh Batra, Suhas Vasaikar, Nathan Edwards, Michael A. Gillette, Chet Birger, Scott D. Jewell, Kei Suzuki, William Bocik, Shilpi Singh, Meenakshi Anurag, Karen E. Christianson, Namrata D. Udeshi, Vasileios Stathias, Warren G. Tourtellotte, Karl R. Clauser, Shutack, Andrii Karnuta, Dana R. Valley, Kelly V. Ruggles, Qing Kay Li, Amanda G. Paulovich, MacIntosh Cornwell, Shankara Anand, Bartosz Kubisa, Pierre M. Jean Beltran, James Suh, Gilbert S. Omenn, Azra Krek, Wohaib Hasan, Yongchao Dou, David Fenyö, Henry Rodriguez, Samuel H. Payne, Małgorzata Wojtyś, Daniel W. Chan, Bo Wen, Nicollette Maunganidze, Özgün Babur, Renganayaki Pandurengan, Karen A. Ketchum, Nikolay Gabrovski, Pankaj Vats, Saravana M. Dhanasekaran, Richard D. Smith, Gad Getz, Sailaja Mareedu, Yuxing Liao, Mikhail Krotevich, Hui Zhang, Eric J. Jaehnig, Charles A. Goldthwaite, Alexey I. Nesvizhskii, Katherine A. Hoadley, Alexander A. Green, Francesca Petralia, Chandan Kumar-Sinha, Karsten Krug, Eunkyung An, Elena V. Ponomareva, Ximing Tang, Nancy Roche, Daniel C. Rohrer, David I. Heiman, Arul M. Chinnaiyan, Pamela Grady, Rebecca I. Montgomery, Galen Hostetter, Liqun Qi, Stephan C. Schürer, George D. Wilson, Pushpa Hariharan, Zhen Zhang, Yvonne, David Chesla, Chia-Kuei Mo, Maria Gabriela Raso, Negin Vatanian, Paul K. Paik, Fei Ding, Thomas L. Bauer, Barbara Hindenach, Matthew J. Ellis, Chen Huang, Karin D. Rodland, Oluwole Fadare, Ramaswamy Govindan, Eric E. Schadt, Sandra Cottingham, Barbara Pruetz, Sendurai A. Mani, Shirley Tsang, Carissa Huynh, Weiping Ma, Jennifer E. Maas, Tobias Schraink, Stacey Gabriel, Bing Zhang, Tara Hiltke, Rama Soundararajan, Tatiana Omelchenko, Brian J. Druker, Matthew A. Wyczalkowski, Neil R. Mucci, Ziad Hanhan, Donna E. Hansel, Yifat Geffen, Mathangi Thiagarajan, Xiaojun Jing, Pei Wang, Alfredo Molinolo, Tanmayi Vashist, Ratna R. Thangudu, Maciej Wiznerowicz, Edwin R. Parra, Tanvi H. Visal, Maureen Dyer, Melissa Borucki, Ki Sung Um, Jonathan T. Lei, Marcin Cieslik, Christopher R. Kinsinger, M. Harry Kane, Houxiang Zhu, Chelsea J. Newton, Steven A. Carr, Tao Liu, Wenke Liu, Volodymyr Sovenko, Olga Potapova, Eric J. Burks, Song Cao, Ana I. Robles, Yuping Zhang, Yize Li, Midie Xu, Erik J. Bergstrom, Zeynep H. Gümüş, Kai Li, and Xiaoyu Song
- Subjects
Adult ,Male ,Epithelial-Mesenchymal Transition ,Lung Neoplasms ,Biology ,Proteomics ,Receptor Tyrosine Kinase-like Orphan Receptors ,General Biochemistry, Genetics and Molecular Biology ,Article ,SOX2 ,CDKN2A ,Survivin ,medicine ,Cluster Analysis ,Humans ,Receptors, Platelet-Derived Growth Factor ,Phosphorylation ,Lung cancer ,Aged ,Proteogenomics ,Aged, 80 and over ,EZH2 ,Ubiquitination ,Cyclin-Dependent Kinase 4 ,Acetylation ,Cyclin-Dependent Kinase 6 ,Middle Aged ,medicine.disease ,Chromatin ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Mutation ,Cancer research ,Carcinoma, Squamous Cell ,Female ,Protein Binding ,Signal Transduction - Abstract
Lung squamous cell carcinoma (LSCC) remains a leading cause of cancer death with few therapeutic options. We characterized the proteogenomic landscape of LSCC, providing a deeper exposition of LSCC biology with potential therapeutic implications. We identify NSD3 as an alternative driver in FGFR1-amplified tumors and low-p63 tumors overexpressing the therapeutic target survivin. SOX2 is considered undruggable, but our analyses provide rationale for exploring chromatin modifiers such as LSD1 and EZH2 to target SOX2-overexpressing tumors. Our data support complex regulation of metabolic pathways by crosstalk between post-translational modifications including ubiquitylation. Numerous immune-related proteogenomic observations suggest directions for further investigation. Proteogenomic dissection of CDKN2A mutations argue for more nuanced assessment of RB1 protein expression and phosphorylation before declaring CDK4/6 inhibition unsuccessful. Finally, triangulation between LSCC, LUAD, and HNSCC identified both unique and common therapeutic vulnerabilities. These observations and proteogenomics data resources may guide research into the biology and treatment of LSCC.
- Published
- 2020
41. Split-TurboID enables contact-dependent proximity labeling in cells
- Author
-
Sanjana Rajeev, Chulhwan Kwak, Tanya Svinkina, Alice Y. Ting, Kelvin F. Cho, Themis Thoudam, Hyun-Woo Rhee, Steven A. Carr, Inkyu Lee, Namrata D. Udeshi, and Tess C. Branon
- Subjects
chemistry.chemical_classification ,0303 health sciences ,Biochemical fractionation ,Conventional fractionation ,Multidisciplinary ,Proteome ,Staining and Labeling ,Chemistry ,Endoplasmic reticulum ,Membrane Proteins ,Biological Sciences ,Endoplasmic Reticulum ,Mitochondria ,03 medical and health sciences ,0302 clinical medicine ,Enzyme ,HEK293 Cells ,Biotinylation ,Organelle ,Mitochondrial Membranes ,Biophysics ,Humans ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Proximity labeling (PL) catalyzed by promiscuous enzymes such as TurboID have enabled the proteomic analysis of subcellular regions difficult or impossible to access by conventional fractionation-based approaches. Yet some cellular regions, such as organelle contact sites, remain out of reach for current PL methods. To address this limitation, we split the enzyme TurboID into two inactive fragments that recombine when driven together by a protein-protein interaction or membrane-membrane apposition. At endoplasmic reticulum (ER)-mitochondria contact sites, reconstituted TurboID catalyzed spatially-restricted biotinylation, enabling the enrichment and identification of >100 endogenous proteins, including many not previously linked to ER-mitochondria contacts. We validated eight novel candidates by biochemical fractionation and overexpression imaging. Overall, split-TurboID is a versatile tool for conditional and spatially-specific proximity labeling in cells.
- Published
- 2020
42. Proteomics of protein trafficking by in vivo tissue-specific labeling
- Author
-
Rebecca Zeng, Luye Mu, Tanya Svinkina, Alice Y. Ting, Tess C. Branon, Norbert Perrimon, Ilia A. Droujinine, Areya Tabatabai, Steven A. Carr, Yanhui Hu, Namrata D. Udeshi, Dan Wang, Justin A. Bosch, and John M. Asara
- Subjects
chemistry.chemical_classification ,0303 health sciences ,DNA ligase ,Biology ,Proteomics ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Secretory protein ,chemistry ,Biotin ,In vivo ,030220 oncology & carcinogenesis ,Biotinylation ,Compartment (development) ,Secretion ,030304 developmental biology - Abstract
Secreted interorgan communication factors encode key regulators of homeostasis. However, long-standing questions surround their origins/destinations, mechanisms of interactions, and the number of proteins involved. Progress has been hindered by the lack of methodologies for these factors’ large-scale identification and characterization, as conventional approaches cannot identify low-abundance factors and the origins and destinations of secreted proteins. We established an in vivo platform to investigate secreted protein trafficking between organs proteome-wide, whereby engineered promiscuous biotin ligase BirA*G3 (a relative of TurboID) biotinylates all proteins in a subcellular compartment of one tissue, and biotinylated proteins are affinity-enriched and identified from distal organs using quantitative mass spectrometry. Using this platform, we identified 51 putative muscle-secreted proteins from heads and 269 fat body-secreted proteins from legs/muscles, of which 60-70% have human orthologs. We demonstrate, in particular, that conserved fat body-derived novel interorgan communication factors CG31326, CG2145, and CG4332 promote muscle activity. Our results indicate that the communication network of secreted proteins is vast, and we identified systemic functions for a number of these factors. This approach is widely applicable to studies in interorgan, local and intracellular protein trafficking networks, non-conventional secretion, and to mammalian systems, under healthy or diseased states.One Sentence SummaryWe developed an in vivo platform to investigate protein trafficking between organs proteome-wide, provide a resource for interorgan communication factors, and determined conserved adipokines that affect muscles.
- Published
- 2020
- Full Text
- View/download PDF
43. CBL mutations promote activation of PI3K/AKT signaling via LYN kinase
- Author
-
Caroline Stanclift, Roger Belizaire, Tanya Svinkina, Alexis Vedder, Christina R. Hartigan, Steven A. Carr, Sebastian Koochaki, Benjamin L. Ebert, Lei Sun, Monica Schenone, Eric Padron, and Namrata D. Udeshi
- Subjects
Myeloid ,Biology ,medicine.disease_cause ,environment and public health ,Interactome ,03 medical and health sciences ,0302 clinical medicine ,LYN ,hemic and lymphatic diseases ,medicine ,Protein kinase B ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,0303 health sciences ,fungi ,3. Good health ,Ubiquitin ligase ,Dasatinib ,enzymes and coenzymes (carbohydrates) ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Carcinogenesis ,medicine.drug - Abstract
CBL encodes an E3 ubiquitin ligase and signaling adaptor that acts downstream of cytokine receptors. Recurrent CBL mutations occur in myeloid malignancies, but the mechanism by which these mutations drive oncogenesis remains incompletely understood. Here we performed a series of studies to define the phosphoproteome, CBL interactome and molecular mechanisms of signaling activation in cells expressing an allelic series of CBL mutants. Our analyses revealed that increased LYN activation and interaction with mutant CBL are key drivers of enhanced PIK3R1 recruitment and downstream PI3K/AKT signaling in CBL-mutant cells. Furthermore, we demonstrated in vitro and in vivo efficacy of LYN inhibition by dasatinib in CBL-mutant cell lines and primary chronic myelomonocytic leukemia cells. Overall, our data provide rationale for exploring the therapeutic potential of LYN inhibition in patients with CBL-mutated myeloid malignancies.Statement of SignificanceWe investigated the oncogenic mechanisms of myeloid malignancy-associated CBL mutations by mass spectrometry-based proteomics and interactomics. Our findings indicate that increased LYN kinase activity in CBL-mutant cells stimulates PI3K/AKT signaling, revealing opportunities for the use of targeted inhibitors in CBL-mutated myeloid malignancies.
- Published
- 2020
44. Steroid resistance in Diamond Blackfan anemia associates with p57Kip2 dysregulation in erythroid progenitors
- Author
-
Julien Papoin, Brian M. Dulmovits, Jeffrey M. Lipton, Hongxia Yan, Narla Mohandas, Nan Wang, Christopher D. Hillyer, Steven A. Carr, Lydie Da Costa, Ryan Ashley, Meagan E. Olive, Naomi Taylor, Adrianna Vlachos, Sandrina Kinet, Namrata D. Udeshi, Lionel Blanc, Anupama Narla, John Hale, Institut de Génétique Moléculaire de Montpellier (IGMM), Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM), Cornell University [New York], Dynamique des interactions membranaires normales et pathologiques (DIMNP), Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Université Montpellier 1 (UM1), The Broad Institute [Cambridge, MA, USA], Harvard University [Cambridge]-Massachusetts Institute of Technology (MIT), and The Feinstein Institute for Medical Research
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,[SDV]Life Sciences [q-bio] ,Population ,Drug Resistance ,Context (language use) ,Biology ,Dexamethasone ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Antigens, CD ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Progenitor cell ,Diamond–Blackfan anemia ,education ,Cyclin-Dependent Kinase Inhibitor p57 ,ComputingMilieux_MISCELLANEOUS ,Anemia, Diamond-Blackfan ,Erythroid Precursor Cells ,education.field_of_study ,Hematology ,General Medicine ,medicine.disease ,Up-Regulation ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer research ,Erythropoiesis ,Female ,Cyclin-Dependent Kinase Inhibitor p27 ,Research Article ,medicine.drug - Abstract
Despite the effective clinical use of steroids for the treatment of Diamond Blackfan anemia (DBA), the mechanisms through which glucocorticoids regulate human erythropoiesis remain poorly understood. We report that the sensitivity of erythroid differentiation to dexamethasone is dependent on the developmental origin of human CD34(+) progenitor cells, specifically increasing the expansion of CD34(+) progenitors from peripheral blood (PB) but not cord blood (CB). Dexamethasone treatment of erythroid-differentiated PB, but not CB, CD34(+) progenitors resulted in the expansion of a newly defined CD34(+)CD36(+)CD71(hi)CD105(med) immature colony-forming unit–erythroid (CFU-E) population. Furthermore, proteomics analyses revealed the induction of distinct proteins in dexamethasone-treated PB and CB erythroid progenitors. Dexamethasone treatment of PB progenitors resulted in the specific upregulation of p57(Kip2), a Cip/Kip cyclin–dependent kinase inhibitor, and we identified this induction as critical; shRNA-mediated downregulation of p57(Kip2), but not the related p27(Kip1), significantly attenuated the impact of dexamethasone on erythroid differentiation and inhibited the expansion of the immature CFU-E subset. Notably, in the context of DBA, we found that steroid resistance was associated with dysregulated p57(Kip2) expression. Altogether, these data identify a unique glucocorticoid-responsive human erythroid progenitor and provide new insights into glucocorticoid-based therapeutic strategies for the treatment of patients with DBA.
- Published
- 2020
45. Discovery of suppressors of CRMP2 phosphorylation reveals compounds that mimic the behavioral effects of lithium on amphetamine-induced hyperlocomotion
- Author
-
Deepak Mani, Cameron D. Pernia, Steven D. Sheridan, Jasmin Lalonde, Joshua A. Bishop, Brian T. D. Tobe, Debasis Patnaik, Namrata D. Udeshi, Stephen J. Haggarty, Steven A. Carr, Lucius L Xuan, Irina N. Gaisina, Evan Y. Snyder, Sarah R. Blumenthal, Daniel P Zolg, Wen-Ning Zhao, Amanda J. Roberts, and Iren Kurtser
- Subjects
0301 basic medicine ,Bipolar Disorder ,Lithium (medication) ,Induced Pluripotent Stem Cells ,Lithium ,Predictive markers ,Molecular neuroscience ,Article ,lcsh:RC321-571 ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Stable isotope labeling by amino acids in cell culture ,medicine ,Animals ,Humans ,Phosphorylation ,Kinase activity ,Protein kinase A ,Induced pluripotent stem cell ,Amphetamine ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Biological Psychiatry ,Chemistry ,3. Good health ,Cell biology ,Psychiatry and Mental health ,030104 developmental biology ,Lithium Compounds ,Collapsin response mediator protein family ,030217 neurology & neurosurgery ,medicine.drug - Abstract
The effective treatment of bipolar disorder (BD) represents a significant unmet medical need. Although lithium remains a mainstay of treatment for BD, limited knowledge regarding how it modulates affective behavior has proven an obstacle to discovering more effective mood stabilizers with fewer adverse side effects. One potential mechanism of action of lithium is through inhibition of the serine/threonine protein kinase GSK3β, however, relevant substrates whose change in phosphorylation may mediate downstream changes in neuroplasticity remain poorly understood. Here, we used human induced pluripotent stem cell (hiPSC)-derived neuronal cells and stable isotope labeling by amino acids in cell culture (SILAC) along with quantitative mass spectrometry to identify global changes in the phosphoproteome upon inhibition of GSK3α/β with the highly selective, ATP-competitive inhibitor CHIR-99021. Comparison of phosphorylation changes to those induced by therapeutically relevant doses of lithium treatment led to the identification of collapsin response mediator protein 2 (CRMP2) as being highly sensitive to both treatments as well as an extended panel of structurally distinct GSK3α/β inhibitors. On this basis, a high-content image-based assay in hiPSC-derived neurons was developed to screen diverse compounds, including FDA-approved drugs, for their ability to mimic lithium’s suppression of CRMP2 phosphorylation without directly inhibiting GSK3β kinase activity. Systemic administration of a subset of these CRMP2-phosphorylation suppressors were found to mimic lithium’s attenuation of amphetamine-induced hyperlocomotion in mice. Taken together, these studies not only provide insights into the neural substrates regulated by lithium, but also provide novel human neuronal assays for supporting the development of mechanism-based therapeutics for BD and related neuropsychiatric disorders.
- Published
- 2020
46. Rapid and deep-scale ubiquitylation profiling for biology and translational research
- Author
-
Jessica A. Gasser, Shankha Satpathy, Tanya Svinkina, Shaunt Fereshetian, Steven A. Carr, Philipp Mertins, Benjamin L. Ebert, Deepak Mani, Namrata D. Udeshi, and Meagan E. Olive
- Subjects
Proteomics ,0301 basic medicine ,Ubiquitylation ,Proteome ,Ion-mobility spectrometry ,Science ,Ubiquitin-Protein Ligases ,General Physics and Astronomy ,Breast Neoplasms ,Peptide ,Sensitivity and Specificity ,Quantitative accuracy ,Article ,General Biochemistry, Genetics and Molecular Biology ,Translational Research, Biomedical ,Ikaros Transcription Factor ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Protein ubiquitylation ,Ubiquitin ,Animals ,Humans ,lcsh:Science ,Casein Kinase Ialpha ,chemistry.chemical_classification ,Multidisciplinary ,Mass spectrometry ,Staining and Labeling ,biology ,Ubiquitination ,Proteins ,General Chemistry ,Ubiquitin ligase ,Cell biology ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Female ,lcsh:Q ,Asymmetric waveform ,Multiple Myeloma ,Protein Processing, Post-Translational ,HeLa Cells - Abstract
Protein ubiquitylation is involved in a plethora of cellular processes. While antibodies directed at ubiquitin remnants (K-ɛ-GG) have improved the ability to monitor ubiquitylation using mass spectrometry, methods for highly multiplexed measurement of ubiquitylation in tissues and primary cells using sub-milligram amounts of sample remains a challenge. Here, we present a highly sensitive, rapid and multiplexed protocol termed UbiFast for quantifying ~10,000 ubiquitylation sites from as little as 500 μg peptide per sample from cells or tissue in a TMT10plex in ca. 5 h. High-field Asymmetric Waveform Ion Mobility Spectrometry (FAIMS) is used to improve quantitative accuracy for posttranslational modification analysis. We use the approach to rediscover substrates of the E3 ligase targeting drug lenalidomide and to identify proteins modulated by ubiquitylation in models of basal and luminal human breast cancer. The sensitivity and speed of the UbiFast method makes it suitable for large-scale studies in primary tissue samples., Comprehensive protein ubiquitylation profiling by mass spectrometry typically requires large sample amounts, limiting its applicability to tissue samples. Here, the authors present an optimized proteomics method that enables multiplexed ubiquitylome analysis of cells and tumor tissue samples.
- Published
- 2020
47. Crbn I391V is sufficient to confer in vivo sensitivity to thalidomide and its derivatives in mice
- Author
-
Tanya Svinkina, James A. Kennedy, Emma C. Fink, D. R. Mani, Benjamin L. Ebert, Dylan N. Adams, Steven A. Carr, Saurav D. Haldar, Michelle C. Chen, Andrew A Guirguis, Namrata D. Udeshi, Brian J. Liddicoat, Marie McConkey, and Andrew T. Nguyen
- Subjects
0301 basic medicine ,biology ,Chemistry ,Cereblon ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Pomalidomide ,Biochemistry ,IKZF3 ,Ubiquitin ligase ,Thalidomide ,03 medical and health sciences ,030104 developmental biology ,In vivo ,medicine ,biology.protein ,Cancer research ,Multiple myeloma ,Lenalidomide ,medicine.drug - Abstract
Thalidomide and its derivatives, lenalidomide and pomalidomide, are clinically effective treatments for multiple myeloma and myelodysplastic syndrome with del(5q). These molecules lack activity in murine models, limiting investigation of their therapeutic activity or toxicity in vivo. Here, we report the development of a mouse model that is sensitive to thalidomide derivatives because of a single amino acid change in the direct target of thalidomide derivatives, cereblon (Crbn). In human cells, thalidomide and its analogs bind CRBN and recruit protein targets to the CRL4CRBN E3 ubiquitin ligase, resulting in their ubiquitination and subsequent degradation by the proteasome. We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1α), both in vitro and in vivo. We use the CrbnI391V model to demonstrate that the in vivo therapeutic activity of lenalidomide in del(5q) myelodysplastic syndrome can be explained by heterozygous expression of Ck1α in del(5q) cells. We found that lenalidomide acts on hematopoietic stem cells with heterozygous expression of Ck1α and inactivation of Trp53 causes lenalidomide resistance. We further demonstrate that CrbnI391V is sufficient to confer thalidomide-induced fetal loss in mice, capturing a major toxicity of this class of drugs. Further study of the CrbnI391V model will provide valuable insights into the in vivo efficacy and toxicity of this class of drugs.
- Published
- 2018
48. Homo-PROTACs for the Chemical Knockdown of Cereblon
- Author
-
Steven A. Carr, Simon Köpff, Hannes Kehm, Michael Gütschow, Deepak Mani, Jan Krönke, Namrata D. Udeshi, Christian Steinebach, and Stefanie Lindner
- Subjects
0301 basic medicine ,Ubiquitin-Protein Ligases ,Biochemistry ,Cell Line ,03 medical and health sciences ,Ubiquitin ,Cell Line, Tumor ,medicine ,Humans ,Immunologic Factors ,Adaptor Proteins, Signal Transducing ,Gene knockdown ,biology ,Chemistry ,Cereblon ,Ubiquitination ,Signal transducing adaptor protein ,General Medicine ,Pomalidomide ,IKZF3 ,Thalidomide ,Cell biology ,Ubiquitin ligase ,030104 developmental biology ,Proteolysis ,Proteome ,biology.protein ,Molecular Medicine ,Dimerization ,Peptide Hydrolases ,medicine.drug - Abstract
The immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide, all approved for the treatment of multiple myeloma, induce targeted ubiquitination and degradation of Ikaros (IKZF1) and Aiolos (IKZF3) via the cereblon (CRBN) E3 ubiquitin ligase. IMiD-based proteolysis-targeting chimeras (PROTACs) can efficiently recruit CRBN to a protein of interest, leading to its ubiquitination and proteasomal degradation. By linking two pomalidomide molecules, we designed homobifunctional, so-called homo-PROTACs and investigated their ability to induce self-directed ubiquitination and degradation. The homodimerized compound 15a was characterized as a highly potent and efficient CRBN degrader with only minimal effects on IKZF1 and IKZF3. The cellular selectivity of 15a for CRBN degradation was confirmed at the proteome level by quantitative mass spectrometry. Inactivation by compound 15a did not affect proliferation of different cell lines, prevented pomalidomide-induced degradation of IKZF1 and IKZF3, and antagonized the effects of pomalidomide on multiple myeloma cells. Homobifunctional CRBN degraders will be useful tools for future biomedical investigations of CRBN-related signaling and may help to further elucidate the molecular mechanism of thalidomide analogues.
- Published
- 2018
49. Efficient proximity labeling in living cells and organisms with TurboID
- Author
-
Jessica L. Feldman, Justin A. Bosch, Steven A. Carr, Namrata D. Udeshi, Norbert Perrimon, Ariana D. Sanchez, Tess C. Branon, Tanya Svinkina, and Alice Y. Ting
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Mutation ,DNA ligase ,Staining and Labeling ,Mutant ,Biomedical Engineering ,Bioengineering ,Yeast display ,Compartmentalization (psychology) ,Directed evolution ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Article ,Cell biology ,Cell Physiological Phenomena ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Biotin ,chemistry ,Protein Interaction Networks ,medicine ,Molecular Medicine ,Biotechnology - Abstract
Protein interaction networks and protein compartmentalization underlie all signaling and regulatory processes in cells. Enzyme-catalyzed proximity labeling (PL) has emerged as a new approach to study the spatial and interaction characteristics of proteins in living cells. However, current PL methods require over 18 hour labeling times or utilize chemicals with limited cell permeability or high toxicity. We used yeast display-based directed evolution to engineer two promiscuous mutants of biotin ligase, TurboID and miniTurbo, which catalyze PL with much greater efficiency than BioID or BioID2, and enable 10-minute PL in cells with non-toxic and easily deliverable biotin. Furthermore, TurboID extends biotin-based PL to flies and worms.
- Published
- 2018
50. Reproducible workflow for multiplexed deep-scale proteome and phosphoproteome analysis of tumor tissues by liquid chromatography–mass spectrometry
- Author
-
Philipp Mertins, Lijun Chen, Daniel W. Chan, Tao Liu, Deepak Mani, Hui Zhang, Karsten Krug, David J. Clark, Michael A. Gillette, Matthew E. Monroe, D. R. Mani, Steven A. Carr, Therese R. W. Clauss, Richard D. Smith, Marina A. Gritsenko, Filip Mundt, Sherri R. Davies, Karl R. Clauser, Hasmik Keshishian, Vladislav A. Petyuk, Namrata D. Udeshi, Yingwei Hu, Ronald J. Moore, Zhen Zhang, Punit Shah, Rui Zhao, Lauren C. Tang, Raymond R. Townsend, Michael Schnaubelt, and Stefani N. Thomas
- Subjects
Proteomics ,0301 basic medicine ,Proteome ,Breast Neoplasms ,Tandem mass tag ,Mass spectrometry ,Mass Spectrometry ,Article ,General Biochemistry, Genetics and Molecular Biology ,Workflow ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Liquid chromatography–mass spectrometry ,Animals ,Humans ,Reproducibility ,Chromatography ,Chemistry ,Phosphoproteins ,Tumor tissue ,High-Throughput Screening Assays ,Benchmarking ,Disease Models, Animal ,030104 developmental biology ,030220 oncology & carcinogenesis ,Heterografts ,Female ,Neoplasm Transplantation ,Chromatography, Liquid - Abstract
Here we present an optimized workflow for global proteome and phosphoproteome analysis of tissues or cell lines that uses isobaric tags (TMT (tandem mass tags)-10) for multiplexed analysis and relative quantification, and provides 3× higher throughput than iTRAQ (isobaric tags for absolute and relative quantification)-4-based methods with high intra- and inter-laboratory reproducibility. The workflow was systematically characterized and benchmarked across three independent laboratories using two distinct breast cancer subtypes from patient-derived xenograft models to enable assessment of proteome and phosphoproteome depth and quantitative reproducibility. Each plex consisted of ten samples, each being 300 μg of peptide derived from 0.88. The maximum deviation for the phosphoproteome coverage was 37,000 quantified phosphosites per sample and differential quantification correlations of r > 0.72. The full procedure, including sample processing and data generation, can be completed within 10 d for ten tissue samples, and 100 samples can be analyzed in −4 months using a single LC-MS/MS instrument. The high quality, depth, and reproducibility of the data obtained both within and across laboratories should enable new biological insights to be obtained from mass spectrometry-based proteomics analyses of cells and tissues together with proteogenomic data integration.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.