28 results on '"Shishkova E"'
Search Results
2. Проблемы и пути развития малого бизнеса в условиях инновационной экономики
- Author
-
Шишкова, Е. Е., Shishkova, E. E., Шишкова, Е. Е., and Shishkova, E. E.
- Published
- 2011
3. Повышение эффективности инвестиций в основной капитал в условиях инновационной экономики
- Author
-
Шишкова, Е. Е., Shishkova, E. E., Шишкова, Е. Е., and Shishkova, E. E.
- Published
- 2010
4. Повышение роли экономико-математических методов в планировании показателей деятельности организаций
- Author
-
Шишкова, Е. Е., Shishkova, E. E., Шишкова, Е. Е., and Shishkova, E. E.
- Published
- 2005
5. Fast and deep phosphoproteome analysis with the Orbitrap Astral mass spectrometer.
- Author
-
Lancaster NM, Sinitcyn P, Forny P, Peters-Clarke TM, Fecher C, Smith AJ, Shishkova E, Arrey TN, Pashkova A, Robinson ML, Arp N, Fan J, Hansen J, Galmozzi A, Serrano LR, Rojas J, Gasch AP, Westphall MS, Stewart H, Hock C, Damoc E, Pagliarini DJ, Zabrouskov V, and Coon JJ
- Subjects
- Humans, Animals, HeLa Cells, Phosphorylation, Mice, Phosphoproteins metabolism, Phosphoproteins analysis, Proteome metabolism, Mass Spectrometry methods, Proteomics methods
- Abstract
Owing to its roles in cellular signal transduction, protein phosphorylation plays critical roles in myriad cell processes. That said, detecting and quantifying protein phosphorylation has remained a challenge. We describe the use of a novel mass spectrometer (Orbitrap Astral) coupled with data-independent acquisition (DIA) to achieve rapid and deep analysis of human and mouse phosphoproteomes. With this method, we map approximately 30,000 unique human phosphorylation sites within a half-hour of data collection. The technology is benchmarked to other state-of-the-art MS platforms using both synthetic peptide standards and with EGF-stimulated HeLa cells. We apply this approach to generate a phosphoproteome multi-tissue atlas of the mouse. Altogether, we detect 81,120 unique phosphorylation sites within 12 hours of measurement. With this unique dataset, we examine the sequence, structural, and kinase specificity context of protein phosphorylation. Finally, we highlight the discovery potential of this resource with multiple examples of phosphorylation events relevant to mitochondrial and brain biology., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
6. The One Hour Human Proteome.
- Author
-
Serrano LR, Peters-Clarke TM, Arrey TN, Damoc E, Robinson ML, Lancaster NM, Shishkova E, Moss C, Pashkova A, Sinitcyn P, Brademan DR, Quarmby ST, Peterson AC, Zeller M, Hermanson D, Stewart H, Hock C, Makarov A, Zabrouskov V, and Coon JJ
- Subjects
- Humans, Time Factors, Proteome analysis, Tandem Mass Spectrometry, Proteomics methods
- Abstract
We describe deep analysis of the human proteome in less than 1 h. We achieve this expedited proteome characterization by leveraging state-of-the-art sample preparation, chromatographic separations, and data analysis tools, and by using the new Orbitrap Astral mass spectrometer equipped with a quadrupole mass filter, a high-field Orbitrap mass analyzer, and an asymmetric track lossless (Astral) mass analyzer. The system offers high tandem mass spectrometry acquisition speed of 200 Hz and detects hundreds of peptide sequences per second within data-independent acquisition or data-dependent acquisition modes of operation. The fast-switching capabilities of the new quadrupole complement the sensitivity and fast ion scanning of the Astral analyzer to enable narrow-bin data-independent analysis methods. Over a 30-min active chromatographic method consuming a total analysis time of 56 min, the Q-Orbitrap-Astral hybrid MS collects an average of 4319 MS
1 scans and 438,062 tandem mass spectrometry scans per run, producing 235,916 peptide sequences (1% false discovery rate). On average, each 30-min analysis achieved detection of 10,411 protein groups (1% false discovery rate). We conclude, with these results and alongside other recent reports, that the 1-h human proteome is within reach., Competing Interests: Conflict of interest The authors declare the following competing financial interest(s): J. J. C. is a consultant for Thermo Fisher Scientific, Seer, and 908 Devices. T. N. A., E. D., A. P., M. Z., D. H., H. S., C. H., A. M., and V. Z. are employees of Thermo Fisher Scientific., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
7. MmuPV1 E6 induces cell proliferation and other hallmarks of cancer.
- Author
-
Romero-Masters JC, Muehlbauer LK, Hayes M, Grace M, Shishkova E, Coon JJ, Munger K, and Lambert PF
- Subjects
- Animals, Mice, Keratinocytes virology, Keratinocytes metabolism, Proteomics, Papillomavirus Infections virology, Neoplasms pathology, Neoplasms metabolism, Humans, Cell Differentiation, Cell Proliferation, Oncogene Proteins, Viral genetics, Oncogene Proteins, Viral metabolism
- Abstract
Importance: The Mus musculus papillomavirus 1 (MmuPV1) E6 and E7 proteins are required for MmuPV1-induced disease. Our understanding of the activities of MmuPV1 E6 has been based on affinity purification/mass spectrometry studies where cellular interacting partners of MmuPV1 E6 were identified, and these studies revealed that MmuPV1 E6 can inhibit keratinocyte differentiation through multiple mechanisms. We report that MmuPV1 E6 encodes additional activities including the induction of proliferation, resistance to density-mediated growth arrest, and decreased dependence on exogenous growth factors. Proteomic and transcriptomic analyses provided evidence that MmuPV1 E6 increases the expression and steady state levels of a number of cellular proteins that promote cellular proliferation and other hallmarks of cancer. These results indicate that MmuPV1 E6 is a major driver of MmuPV1-induced pathogenesis., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
8. Global detection of human variants and isoforms by deep proteome sequencing.
- Author
-
Sinitcyn P, Richards AL, Weatheritt RJ, Brademan DR, Marx H, Shishkova E, Meyer JG, Hebert AS, Westphall MS, Blencowe BJ, Cox J, and Coon JJ
- Subjects
- Humans, Protein Isoforms genetics, Peptides chemistry, Amino Acid Sequence, Proteome genetics, Proteome metabolism, Alternative Splicing genetics
- Abstract
An average shotgun proteomics experiment detects approximately 10,000 human proteins from a single sample. However, individual proteins are typically identified by peptide sequences representing a small fraction of their total amino acids. Hence, an average shotgun experiment fails to distinguish different protein variants and isoforms. Deeper proteome sequencing is therefore required for the global discovery of protein isoforms. Using six different human cell lines, six proteases, deep fractionation and three tandem mass spectrometry fragmentation methods, we identify a million unique peptides from 17,717 protein groups, with a median sequence coverage of approximately 80%. Direct comparison with RNA expression data provides evidence for the translation of most nonsynonymous variants. We have also hypothesized that undetected variants likely arise from mutation-induced protein instability. We further observe comparable detection rates for exon-exon junction peptides representing constitutive and alternative splicing events. Our dataset represents a resource for proteoform discovery and provides direct evidence that most frame-preserving alternatively spliced isoforms are translated., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
9. PPTC7 maintains mitochondrial protein content by suppressing receptor-mediated mitophagy.
- Author
-
Niemi NM, Serrano LR, Muehlbauer LK, Balnis CE, Wei L, Smith AJ, Kozul KL, Forny M, Connor OM, Rashan EH, Shishkova E, Schueler KL, Keller MP, Attie AD, Friedman JR, Pagan JK, Coon JJ, and Pagliarini DJ
- Subjects
- Animals, Mice, Fibroblasts metabolism, Mitochondria genetics, Mitochondria metabolism, Phosphoric Monoester Hydrolases metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Mitophagy genetics
- Abstract
PPTC7 is a resident mitochondrial phosphatase essential for maintaining proper mitochondrial content and function. Newborn mice lacking Pptc7 exhibit aberrant mitochondrial protein phosphorylation, suffer from a range of metabolic defects, and fail to survive beyond one day after birth. Using an inducible knockout model, we reveal that loss of Pptc7 in adult mice causes marked reduction in mitochondrial mass and metabolic capacity with elevated hepatic triglyceride accumulation. Pptc7 knockout animals exhibit increased expression of the mitophagy receptors BNIP3 and NIX, and Pptc7
-/- mouse embryonic fibroblasts (MEFs) display a major increase in mitophagy that is reversed upon deletion of these receptors. Our phosphoproteomics analyses reveal a common set of elevated phosphosites between perinatal tissues, adult liver, and MEFs, including multiple sites on BNIP3 and NIX, and our molecular studies demonstrate that PPTC7 can directly interact with and dephosphorylate these proteins. These data suggest that Pptc7 deletion causes mitochondrial dysfunction via dysregulation of several metabolic pathways and that PPTC7 may directly regulate mitophagy receptor function or stability. Overall, our work reveals a significant role for PPTC7 in the mitophagic response and furthers the growing notion that management of mitochondrial protein phosphorylation is essential for ensuring proper organelle content and function., (© 2023. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
10. Regulation of DNA damage and transcriptional output in the vasculature through a cytoglobin-HMGB2 axis.
- Author
-
Mathai C, Jourd'heuil F, Pham LGC, Gilliard K, Howard D, Balnis J, Jaitovich A, Chittur SV, Rilley M, Peredo-Wende R, Ammoura I, Shin SJ, Barroso M, Barra J, Shishkova E, Coon JJ, Lopez-Soler RI, and Jourd'heuil D
- Subjects
- Animals, Mice, Cytoglobin genetics, DNA Damage, Transcription Factors genetics, Globins genetics, Globins metabolism, HMGB2 Protein genetics, HMGB2 Protein metabolism
- Abstract
Identifying novel regulators of vascular smooth muscle cell function is necessary to further understand cardiovascular diseases. We previously identified cytoglobin, a hemoglobin homolog, with myogenic and cytoprotective roles in the vasculature. The specific mechanism of action of cytoglobin is unclear but does not seem to be related to oxygen transport or storage like hemoglobin. Herein, transcriptomic profiling of injured carotid arteries in cytoglobin global knockout mice revealed that cytoglobin deletion accelerated the loss of contractile genes and increased DNA damage. Overall, we show that cytoglobin is actively translocated into the nucleus of vascular smooth muscle cells through a redox signal driven by NOX4. We demonstrate that nuclear cytoglobin heterodimerizes with the non-histone chromatin structural protein HMGB2. Our results are consistent with a previously unknown function by which a non-erythrocytic hemoglobin inhibits DNA damage and regulates gene programs in the vasculature by modulating the genome-wide binding of HMGB2., Competing Interests: Declaration of competing interest JJC is a consultant for Thermo Fisher Scientific, 908 Devices, and Seer., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
11. N-glycoproteomics of brain synapses and synaptic vesicles.
- Author
-
Bradberry MM, Peters-Clarke TM, Shishkova E, Chapman ER, and Coon JJ
- Subjects
- Mice, Animals, Glycoproteins metabolism, Brain metabolism, Polysaccharides metabolism, Mammals metabolism, Synaptic Vesicles metabolism, Synapses metabolism
- Abstract
At mammalian neuronal synapses, synaptic vesicle (SV) glycoproteins are essential for robust neurotransmission. Asparagine (N)-linked glycosylation is required for delivery of the major SV glycoproteins synaptophysin and SV2A to SVs. Despite this key role for N-glycosylation, the molecular compositions of SV N-glycans are largely unknown. In this study, we combined organelle isolation techniques and high-resolution mass spectrometry to characterize N-glycosylation at synapses and SVs from mouse brain. Detecting over 2,500 unique glycopeptides, we found that SVs harbor a distinct population of oligomannose and highly fucosylated N-glycans. Using complementary fluorescence methods, we identify at least one highly fucosylated N-glycan enriched in SVs compared with synaptosomes. High fucosylation was characteristic of SV proteins, plasma membrane proteins, and cell adhesion molecules with key roles in synaptic function and development. Our results define the N-glycoproteome of a specialized neuronal organelle and inform timely questions in the glycobiology of synaptic pruning and neuroinflammation., Competing Interests: Declaration of interests J.J.C. is a consultant for Thermo Fisher Scientific, Seer, and 908 Devices., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
12. Defining mitochondrial protein functions through deep multiomic profiling.
- Author
-
Rensvold JW, Shishkova E, Sverchkov Y, Miller IJ, Cetinkaya A, Pyle A, Manicki M, Brademan DR, Alanay Y, Raiman J, Jochem A, Hutchins PD, Peters SR, Linke V, Overmyer KA, Salome AZ, Hebert AS, Vincent CE, Kwiecien NW, Rush MJP, Westphall MS, Craven M, Akarsu NA, Taylor RW, Coon JJ, and Pagliarini DJ
- Subjects
- Cation Transport Proteins, Cell Cycle Proteins, Energy Metabolism, Humans, Mass Spectrometry, Mitochondrial Diseases genetics, Mitochondrial Diseases metabolism, Transcription Factors, rab5 GTP-Binding Proteins, Mitochondria genetics, Mitochondria metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism
- Abstract
Mitochondria are epicentres of eukaryotic metabolism and bioenergetics. Pioneering efforts in recent decades have established the core protein componentry of these organelles
1 and have linked their dysfunction to more than 150 distinct disorders2,3 . Still, hundreds of mitochondrial proteins lack clear functions4 , and the underlying genetic basis for approximately 40% of mitochondrial disorders remains unresolved5 . Here, to establish a more complete functional compendium of human mitochondrial proteins, we profiled more than 200 CRISPR-mediated HAP1 cell knockout lines using mass spectrometry-based multiomics analyses. This effort generated approximately 8.3 million distinct biomolecule measurements, providing a deep survey of the cellular responses to mitochondrial perturbations and laying a foundation for mechanistic investigations into protein function. Guided by these data, we discovered that PIGY upstream open reading frame (PYURF) is an S-adenosylmethionine-dependent methyltransferase chaperone that supports both complex I assembly and coenzyme Q biosynthesis and is disrupted in a previously unresolved multisystemic mitochondrial disorder. We further linked the putative zinc transporter SLC30A9 to mitochondrial ribosomes and OxPhos integrity and established RAB5IF as the second gene harbouring pathogenic variants that cause cerebrofaciothoracic dysplasia. Our data, which can be explored through the interactive online MITOMICS.app resource, suggest biological roles for many other orphan mitochondrial proteins that still lack robust functional characterization and define a rich cell signature of mitochondrial dysfunction that can support the genetic diagnosis of mitochondrial diseases., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
13. Multi-omics analysis identifies essential regulators of mitochondrial stress response in two wild-type C. elegans strains.
- Author
-
Gao AW, El Alam G, Lalou A, Li TY, Molenaars M, Zhu Y, Overmyer KA, Shishkova E, Hof K, Bou Sleiman M, Houtkooper RH, Coon JJ, and Auwerx J
- Abstract
The mitochondrial unfolded protein response (UPRmt) is a promising pharmacological target for aging and age-related diseases. However, the integrative analysis of the impact of UPRmt activation on different signaling layers in animals with different genetic backgrounds is lacking. Here, we applied systems approaches to investigate the effect of UPRmt induced by doxycycline (Dox) on transcriptome, proteome, and lipidome in two genetically divergent worm strains, named N2 and CB4856. From the integrated omics datasets, we found that Dox prolongs lifespan of both worm strains through shared and strain-specific mechanisms. Specifically, Dox strongly impacts mitochondria, upregulates defense response, and lipid metabolism, while decreasing triglycerides. We further validated that lipid genes acs-2/20 and fat-7/6 were required for Dox-induced UPRmt and longevity in N2 and CB4856 worms, respectively. Our data have translational value as they indicate that the beneficial effects of Dox-induced UPRmt on lifespan are consistent across different genetic backgrounds through different regulators., Competing Interests: The authors declare no competing interests., (© 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
14. Rapid preparation of human blood plasma for bottom-up proteomics analysis.
- Author
-
Shishkova E and Coon JJ
- Subjects
- Chromatography, Liquid methods, Humans, Peptide Fragments analysis, Tandem Mass Spectrometry methods, Blood Proteins analysis, Proteome analysis, Proteomics methods
- Abstract
This protocol offers step-by-step instructions for preparation of raw blood plasma for liquid chromatography - tandem mass spectrometry (LC-MS/MS) analysis in clinical proteomics studies. The technique is simple, robust, and reproducible, and the entire transformation from plasma proteins to desalted tryptic peptides takes only 3-4 h. The protocol ensures efficient denaturation of native proteases that, in combination with the speediness of the procedure, prevents non-specific and irreproducible cleavage of digested peptides. The protocol can be adopted for large-scale studies and automation. For complete details on the use and execution of this protocol, please refer to Overmyer et al. (2020)., Competing Interests: E.S. declares no competing interest. J.J.C. is a consultant for Thermo Fisher Scientific., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
15. Loss of C2orf69 defines a fatal autoinflammatory syndrome in humans and zebrafish that evokes a glycogen-storage-associated mitochondriopathy.
- Author
-
Wong HH, Seet SH, Maier M, Gurel A, Traspas RM, Lee C, Zhang S, Talim B, Loh AYT, Chia CY, Teoh TS, Sng D, Rensvold J, Unal S, Shishkova E, Cepni E, Nathan FM, Sirota FL, Liang C, Yarali N, Simsek-Kiper PO, Mitani T, Ceylaner S, Arman-Bilir O, Mbarek H, Gumruk F, Efthymiou S, Çïmen DU, Georgiadou D, Sotiropoulou K, Houlden H, Paul F, Pehlivan D, Lainé C, Chai G, Ali NA, Choo SC, Keng SS, Boisson B, Yılmaz E, Xue S, Coon JJ, Nguyen Ly TT, Gilani N, Hasbini D, Kayserili H, Zaki MS, Isfort RJ, Ordonez N, Tripolszki K, Bauer P, Rezaei N, Seyedpour S, Khotaei GT, Bascom CC, Maroofian R, Chaabouni M, Alsubhi A, Eyaid W, Işıkay S, Gleeson JG, Lupski JR, Casanova JL, Pagliarini DJ, Akarsu NA, Maurer-Stroh S, Cetinkaya A, Bertoli-Avella A, Mathuru AS, Ho L, Bard FA, and Reversade B
- Published
- 2021
- Full Text
- View/download PDF
16. Unique inflammatory profile is associated with higher SARS-CoV-2 acute respiratory distress syndrome (ARDS) mortality.
- Author
-
Balnis J, Adam AP, Chopra A, Chieng HC, Drake LA, Martino N, Bossardi Ramos R, Feustel PJ, Overmyer KA, Shishkova E, Coon JJ, Singer HA, Judson MA, and Jaitovich A
- Subjects
- Aged, COVID-19 mortality, Cohort Studies, Cytokines genetics, Cytokines metabolism, Female, Gene Expression Regulation, Humans, Male, Middle Aged, COVID-19 metabolism, COVID-19 pathology, Inflammation metabolism, Respiratory Distress Syndrome mortality, SARS-CoV-2
- Abstract
The COVID19 pandemic has caused more than a million of deaths worldwide, primarily due to complications from COVID19-associated acute respiratory distress syndrome (ARDS). Controversy surrounds the circulating cytokine/chemokine profile of COVID19-associated ARDS, with some groups suggesting that it is similar to patients without COVID19 ARDS and others observing substantial differences. Moreover, although a hyperinflammatory phenotype associates with higher mortality in non-COVID19 ARDS, there is little information on the inflammatory landscape's association with mortality in patients with COVID19 ARDS. Even though the circulating leukocytes' transcriptomic signature has been associated with distinct phenotypes and outcomes in critical illness including ARDS, it is unclear whether the mortality-associated inflammatory mediators from patients with COVID19 are transcriptionally regulated in the leukocyte compartment. Here, we conducted a prospective cohort study of 41 mechanically ventilated patients with COVID19 infection using highly calibrated methods to define the levels of plasma cytokines/chemokines and their gene expressions in circulating leukocytes. Plasma IL1RA and IL8 were found positively associated with mortality, whereas RANTES and EGF negatively associated with that outcome. However, the leukocyte gene expression of these proteins had no statistically significant correlation with mortality. These data suggest a unique inflammatory signature associated with severe COVID19.
- Published
- 2021
- Full Text
- View/download PDF
17. Large-Scale Multi-omic Analysis of COVID-19 Severity.
- Author
-
Overmyer KA, Shishkova E, Miller IJ, Balnis J, Bernstein MN, Peters-Clarke TM, Meyer JG, Quan Q, Muehlbauer LK, Trujillo EA, He Y, Chopra A, Chieng HC, Tiwari A, Judson MA, Paulson B, Brademan DR, Zhu Y, Serrano LR, Linke V, Drake LA, Adam AP, Schwartz BS, Singer HA, Swanson S, Mosher DF, Stewart R, Coon JJ, and Jaitovich A
- Subjects
- Aged, Aged, 80 and over, COVID-19 therapy, Cohort Studies, Female, Gelsolin blood, Gelsolin genetics, Humans, Inflammation Mediators blood, Male, Middle Aged, Neutrophils metabolism, Principal Component Analysis methods, COVID-19 blood, COVID-19 genetics, Machine Learning, Sequence Analysis, RNA methods, Severity of Illness Index
- Abstract
We performed RNA-seq and high-resolution mass spectrometry on 128 blood samples from COVID-19-positive and COVID-19-negative patients with diverse disease severities and outcomes. Quantified transcripts, proteins, metabolites, and lipids were associated with clinical outcomes in a curated relational database, uniquely enabling systems analysis and cross-ome correlations to molecules and patient prognoses. We mapped 219 molecular features with high significance to COVID-19 status and severity, many of which were involved in complement activation, dysregulated lipid transport, and neutrophil activation. We identified sets of covarying molecules, e.g., protein gelsolin and metabolite citrate or plasmalogens and apolipoproteins, offering pathophysiological insights and therapeutic suggestions. The observed dysregulation of platelet function, blood coagulation, acute phase response, and endotheliopathy further illuminated the unique COVID-19 phenotype. We present a web-based tool (covid-omics.app) enabling interactive exploration of our compendium and illustrate its utility through a machine learning approach for prediction of COVID-19 severity., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. Survival Following Traumatic Brain Injury in Drosophila Is Increased by Heterozygosity for a Mutation of the NF-κB Innate Immune Response Transcription Factor Relish.
- Author
-
Swanson LC, Trujillo EA, Thiede GH, Katzenberger RJ, Shishkova E, Coon JJ, Ganetzky B, and Wassarman DA
- Subjects
- Animals, Brain Injuries, Traumatic immunology, Drosophila melanogaster, Genetic Background, Heterozygote, Immunity, Innate, Mutation, Transcriptome, Brain Injuries, Traumatic genetics, Drosophila Proteins genetics, Transcription Factors genetics
- Abstract
Traumatic brain injury (TBI) pathologies are caused by primary and secondary injuries. Primary injuries result from physical damage to the brain, and secondary injuries arise from cellular responses to primary injuries. A characteristic cellular response is sustained activation of inflammatory pathways commonly mediated by nuclear factor-κB (NF-κB) transcription factors. Using a Drosophila melanogaster TBI model, we previously found that the main proximal transcriptional response to primary injuries is triggered by activation of Toll and Imd innate immune response pathways that engage NF-κB factors Dif and Relish (Rel), respectively. Here, we found by mass spectrometry that Rel protein level increased in fly heads at 4-8 hr after TBI. To investigate the necessity of Rel for secondary injuries, we generated a null allele, Rel
del , by CRISPR/Cas9 editing. When heterozygous but not homozygous, the Reldel mutation reduced mortality at 24 hr after TBI and increased the lifespan of injured flies. Additionally, the effect of heterozygosity for Reldel on mortality was modulated by genetic background and diet. To identify genes that facilitate effects of Reldel on TBI outcomes, we compared genome-wide mRNA expression profiles of uninjured and injured +/+, +/ Reldel , and Reldel / Reldel flies at 4 hr following TBI. Only a few genes changed expression more than twofold in +/ Reldel flies relative to +/+ and Reldel / Reldel flies, and they were not canonical innate immune response genes. Therefore, Rel is necessary for TBI-induced secondary injuries but in complex ways involving Rel gene dose, genetic background, diet, and possibly small changes in expression of innate immune response genes., (Copyright © 2020 by the Genetics Society of America.)- Published
- 2020
- Full Text
- View/download PDF
19. Mass spectrometry proteomics reveals a function for mammalian CALCOCO1 in MTOR-regulated selective autophagy.
- Author
-
Stefely JA, Zhang Y, Freiberger EC, Kwiecien NW, Thomas HE, Davis AM, Lowry ND, Vincent CE, Shishkova E, Clark NA, Medvedovic M, Coon JJ, Pagliarini DJ, and Mercer CA
- Subjects
- Amino Acid Sequence, Animals, Calcium-Binding Proteins chemistry, Conserved Sequence, Embryo, Mammalian cytology, Fibroblasts metabolism, HEK293 Cells, Humans, MCF-7 Cells, Mice, Microtubule-Associated Proteins metabolism, Protein Binding, Saccharomyces cerevisiae metabolism, Transcription Factors chemistry, Autophagy, Calcium-Binding Proteins metabolism, Mammals metabolism, Mass Spectrometry, Proteomics, TOR Serine-Threonine Kinases metabolism, Transcription Factors metabolism
- Abstract
Macroautophagy/autophagy is suppressed by MTOR (mechanistic target of rapamycin kinase) and is an anticancer target under active investigation. Yet, MTOR-regulated autophagy remains incompletely mapped. We used proteomic profiling to identify proteins in the MTOR-autophagy axis. Wild-type (WT) mouse cell lines and cell lines lacking individual autophagy genes ( Atg5 or Ulk1/Ulk2 ) were treated with an MTOR inhibitor to induce autophagy and cultured in media with either glucose or galactose. Mass spectrometry proteome profiling revealed an elevation of known autophagy proteins and candidates for new autophagy components, including CALCOCO1 (calcium binding and coiled-coil domain protein 1). We show that CALCOCO1 physically interacts with MAP1LC3C, a key protein in the machinery of autophagy. Genetic deletion of CALCOCO1 disrupted autophagy of the endoplasmic reticulum (reticulophagy). Together, these results reveal a role for CALCOCO1 in MTOR-regulated selective autophagy. More generally, the resource generated by this work provides a foundation for establishing links between the MTOR-autophagy axis and proteins not previously linked to this pathway. Abbreviations: ATG: autophagy-related; CALCOCO1: calcium binding and coiled-coil domain protein 1; CALCOCO2/NDP52: calcium binding and coiled-coil domain protein 2; CLIR: MAP1LC3C-interacting region; CQ: chloroquine; KO: knockout; LIR: MAP1LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; MLN: MLN0128 ATP-competitive MTOR kinase inhibitor; MTOR: mechanistic target of rapamycin kinase; reticulophagy: selective autophagy of the endoplasmic reticulum; TAX1BP1/CALCOCO3: TAX1 binding protein 1; ULK: unc 51-like autophagy activating kinase; WT: wild-type.
- Published
- 2020
- Full Text
- View/download PDF
20. Constructing and deconstructing GATA2-regulated cell fate programs to establish developmental trajectories.
- Author
-
Johnson KD, Conn DJ, Shishkova E, Katsumura KR, Liu P, Shen S, Ranheim EA, Kraus SG, Wang W, Calvo KR, Hsu AP, Holland SM, Coon JJ, Keles S, and Bresnick EH
- Subjects
- Adolescent, Adult, Animals, Basophils physiology, Cells, Cultured, Enhancer Elements, Genetic genetics, Erythrocytes physiology, Female, Hematopoiesis genetics, Humans, Macrophages physiology, Megakaryocytes physiology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Single-Cell Analysis, Cell Differentiation genetics, GATA2 Transcription Factor genetics, Gene Deletion, Germ-Line Mutation, Stem Cells physiology
- Abstract
Stem and progenitor cell fate transitions constitute key decision points in organismal development that enable access to a developmental path or actively preclude others. Using the hematopoietic system, we analyzed the relative importance of cell fate-promoting mechanisms versus negating fate-suppressing mechanisms to engineer progenitor cells with multilineage differentiation potential. Deletion of the murine Gata2-77 enhancer, with a human equivalent that causes leukemia, downregulates the transcription factor GATA2 and blocks progenitor differentiation into erythrocytes, megakaryocytes, basophils, and granulocytes, but not macrophages. Using multiomics and single-cell analyses, we demonstrated that the enhancer orchestrates a balance between pro- and anti-fate circuitry in single cells. By increasing GATA2 expression, the enhancer instigates a fate-promoting mechanism while abrogating an innate immunity-linked, fate-suppressing mechanism. During embryogenesis, the suppressing mechanism dominated in enhancer mutant progenitors, thus yielding progenitors with a predominant monocytic differentiation potential. Coordinating fate-promoting and -suppressing circuits therefore averts deconstruction of a multifate system into a monopotent system and maintains critical progenitor heterogeneity and functionality., Competing Interests: Disclosures: The authors declare no competing interests exist., (© 2020 Johnson et al.)
- Published
- 2020
- Full Text
- View/download PDF
21. Argonaut: A Web Platform for Collaborative Multi-omic Data Visualization and Exploration.
- Author
-
Brademan DR, Miller IJ, Kwiecien NW, Pagliarini DJ, Westphall MS, Coon JJ, and Shishkova E
- Abstract
Researchers now generate large multi-omic datasets using increasingly mature mass spectrometry techniques at an astounding pace, facing new challenges of "Big Data" dissemination, visualization, and exploration. Conveniently, web-based data portals accommodate the complexity of multi-omic experiments and the many experts involved. However, developing these tailored companion resources requires programming expertise and knowledge of web server architecture-a substantial burden for most. Here, we describe Argonaut, a simple, code-free, and user-friendly platform for creating customizable, interactive data-hosting websites. Argonaut carries out real-time statistical analyses of the data, which it organizes into easily sharable projects. Collaborating researchers worldwide can explore the results, visualized through popular plots, and modify them to streamline data interpretation. Increasing the pace and ease of access to multi-omic data, Argonaut aims to propel discovery of new biological insights. We showcase the capabilities of this tool using a published multi-omics dataset on the large mitochondrial protease deletion collection., Competing Interests: DECLARATION OF INTERESTS N.W.K., M.S.W., and J.J.C. filed a patent, entitled “Web-Based Data Upload and Visualization Platform Enabling Creation of Code-Free Exploration of MS-Based Omics Data” (US20190034047A1; status 9.6.2020 “Pending”), related to the work described in this manuscript. The other authors declare no competing financial interest. SUPPLEMENTAL INFORMATION Supplemental Information can be found online at https://doi.org/10.1016/j.patter.2020.100122.
- Published
- 2020
- Full Text
- View/download PDF
22. Mapping Physiological ADP-Ribosylation Using Activated Ion Electron Transfer Dissociation.
- Author
-
Buch-Larsen SC, Hendriks IA, Lodge JM, Rykær M, Furtwängler B, Shishkova E, Westphall MS, Coon JJ, and Nielsen ML
- Subjects
- Adenosine Diphosphate Ribose metabolism, HeLa Cells, Humans, Ions, Poly (ADP-Ribose) Polymerase-1 metabolism, ADP-Ribosylation physiology, Electrons
- Abstract
ADP-ribosylation (ADPr) is a post-translational modification that plays pivotal roles in a wide range of cellular processes. Mass spectrometry (MS)-based analysis of ADPr under physiological conditions, without relying on genetic or chemical perturbation, has been hindered by technical limitations. Here, we describe the applicability of activated ion electron transfer dissociation (AI-ETD) for MS-based proteomics analysis of physiological ADPr using our unbiased Af1521 enrichment strategy. To benchmark AI-ETD, we profile 9,000 ADPr peptides mapping to >5,000 unique ADPr sites from a limited number of cells exposed to oxidative stress and identify 120% and 28% more ADPr peptides compared to contemporary strategies using ETD and electron-transfer higher-energy collisional dissociation (EThcD), respectively. Under physiological conditions, AI-ETD identifies 450 ADPr sites on low-abundant proteins, including in vivo cysteine modifications on poly(ADP-ribosyl)polymerase (PARP) 8 and tyrosine modifications on PARP14, hinting at specialist enzymatic functions for these enzymes. Collectively, our data provide insights into the physiological regulation of ADPr., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
23. The genetic basis of aneuploidy tolerance in wild yeast.
- Author
-
Hose J, Escalante LE, Clowers KJ, Dutcher HA, Robinson D, Bouriakov V, Coon JJ, Shishkova E, and Gasch AP
- Subjects
- Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism, Aneuploidy, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics
- Abstract
Aneuploidy is highly detrimental during development yet common in cancers and pathogenic fungi - what gives rise to differences in aneuploidy tolerance remains unclear. We previously showed that wild isolates of Saccharomyces cerevisiae tolerate chromosome amplification while laboratory strains used as a model for aneuploid syndromes do not. Here, we mapped the genetic basis to Ssd1, an RNA-binding translational regulator that is functional in wild aneuploids but defective in laboratory strain W303. Loss of SSD1 recapitulates myriad aneuploidy signatures previously taken as eukaryotic responses. We show that aneuploidy tolerance is enabled via a role for Ssd1 in mitochondrial physiology, including binding and regulating nuclear-encoded mitochondrial mRNAs, coupled with a role in mitigating proteostasis stress. Recapitulating ssd1Δ defects with combinatorial drug treatment selectively blocked proliferation of wild-type aneuploids compared to euploids. Our work adds to elegant studies in the sensitized laboratory strain to present a mechanistic understanding of eukaryotic aneuploidy tolerance., Competing Interests: JH, LE, KC, HD, DR, VB, JC, ES, AG No competing interests declared, (© 2020, Hose et al.)
- Published
- 2020
- Full Text
- View/download PDF
24. Vitamin B 12b Enhances the Cytotoxicity of Diethyldithiocarbamate in a Synergistic Manner, Inducing the Paraptosis-Like Death of Human Larynx Carcinoma Cells.
- Author
-
Solovieva M, Shatalin Y, Fadeev R, Krestinina O, Baburina Y, Kruglov A, Kharechkina E, Kobyakova M, Rogachevsky V, Shishkova E, and Akatov AV
- Subjects
- Apoptosis drug effects, Autophagy drug effects, Carcinoma metabolism, Cell Death drug effects, Cell Line, Tumor, Cell Survival drug effects, Ditiocarb metabolism, Drug Synergism, Endoplasmic Reticulum drug effects, Endoplasmic Reticulum Stress drug effects, Humans, Hydroxocobalamin metabolism, Laryngeal Neoplasms metabolism, Larynx metabolism, Mitochondria metabolism, Oxidative Stress drug effects, Vacuoles drug effects, Vitamin B 12 metabolism, Vitamin B 12 pharmacology, Vitamins metabolism, Vitamins pharmacology, Ditiocarb pharmacology, Hydroxocobalamin pharmacology, Laryngeal Neoplasms drug therapy
- Abstract
We have shown that hydroxycobalamin (vitamin В
12b ) increases the toxicity of diethyldithiocarbamate (DDC) to tumor cells by catalyzing the formation of disulfiram (DSF) oxi-derivatives. The purpose of this study was to elucidate the mechanism of tumor cell death induced by the combination DDC + В12b . It was found that cell death induced by DDC + B12b differed from apoptosis, autophagy, and necrosis. During the initiation of cell death, numerous vacuoles formed from ER cisterns in the cytoplasm, and cell death was partially suppressed by the inhibitors of protein synthesis and folding, the IP3 receptor inhibitor as well as by thiols. At this time, a short-term rise in the expression of ER-stress markers BiP and PERK with a steady increase in the expression of CHOP were detected. After the vacuolization of the cytoplasm, functional disorders of mitochondria and an increase in the generation of superoxide anion in them occurred. Taken together, the results obtained indicate that DDC and B12b used in combination exert a synergistic toxic effect on tumor cells by causing severe ER stress, extensive ER vacuolization, and inhibition of apoptosis, which ultimately leads to the induction of paraptosis-like cell death., Competing Interests: The authors declare no conflict of interest.- Published
- 2020
- Full Text
- View/download PDF
25. Decoupling Yeast Cell Division and Stress Defense Implicates mRNA Repression in Translational Reallocation during Stress.
- Author
-
Ho YH, Shishkova E, Hose J, Coon JJ, and Gasch AP
- Subjects
- Polyribosomes genetics, Polyribosomes metabolism, Proteome genetics, Proteome metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Schizosaccharomyces genetics, Schizosaccharomyces pombe Proteins genetics, Schizosaccharomyces pombe Proteins metabolism, Transcriptome genetics, Cell Division genetics, RNA, Fungal metabolism, RNA, Messenger metabolism, Saccharomyces cerevisiae physiology, Schizosaccharomyces physiology, Stress, Physiological genetics
- Abstract
Stress tolerance and rapid growth are often competing interests in cells. Upon severe environmental stress, many organisms activate defense systems concurrent with growth arrest. There has been debate as to whether aspects of the stress-activated transcriptome are regulated by stress or an indirect byproduct of reduced proliferation. For example, stressed Saccharomyces cerevisiae cells mount a common gene expression program called the environmental stress response (ESR) [1] comprised of ∼300 induced (iESR) transcripts involved in stress defense and ∼600 reduced (rESR) mRNAs encoding ribosomal proteins (RPs) and ribosome biogenesis factors (RiBi) important for division. Because ESR activation also correlates with reduced growth rate in nutrient-restricted chemostats and prolonged G1 in slow-growing mutants, an alternate proposal is that the ESR is simply a consequence of reduced division [2-5]. A major challenge is that past studies did not separate effects of division arrest and stress defense; thus, the true responsiveness of the ESR-and the purpose of stress-dependent rESR repression in particular-remains unclear. Here, we decoupled cell division from the stress response by following transcriptome, proteome, and polysome changes in arrested cells responding to acute stress. We show that the ESR cannot be explained by changes in growth rate or cell-cycle phase during stress acclimation. Instead, failure to repress rESR transcripts reduces polysome association of induced transcripts, delaying production of their proteins. Our results suggest that stressed cells alleviate competition for translation factors by removing mRNAs and ribosomes from the translating pool, directing translational capacity toward induced transcripts to accelerate protein production., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
26. Network inference reveals novel connections in pathways regulating growth and defense in the yeast salt response.
- Author
-
MacGilvray ME, Shishkova E, Chasman D, Place M, Gitter A, Coon JJ, and Gasch AP
- Subjects
- Cell Cycle, Computational Biology, Computer Simulation, Cyclic AMP-Dependent Protein Kinases metabolism, Immunoprecipitation, Mass Spectrometry, Models, Biological, Osmotic Pressure, Phosphorylation, Protein Interaction Mapping, Protein Serine-Threonine Kinases metabolism, Proteome, Signal Transduction, Transcription Factors metabolism, Proteomics methods, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae Proteins metabolism, Sodium Chloride chemistry
- Abstract
Cells respond to stressful conditions by coordinating a complex, multi-faceted response that spans many levels of physiology. Much of the response is coordinated by changes in protein phosphorylation. Although the regulators of transcriptome changes during stress are well characterized in Saccharomyces cerevisiae, the upstream regulatory network controlling protein phosphorylation is less well dissected. Here, we developed a computational approach to infer the signaling network that regulates phosphorylation changes in response to salt stress. We developed an approach to link predicted regulators to groups of likely co-regulated phospho-peptides responding to stress, thereby creating new edges in a background protein interaction network. We then use integer linear programming (ILP) to integrate wild type and mutant phospho-proteomic data and predict the network controlling stress-activated phospho-proteomic changes. The network we inferred predicted new regulatory connections between stress-activated and growth-regulating pathways and suggested mechanisms coordinating metabolism, cell-cycle progression, and growth during stress. We confirmed several network predictions with co-immunoprecipitations coupled with mass-spectrometry protein identification and mutant phospho-proteomic analysis. Results show that the cAMP-phosphodiesterase Pde2 physically interacts with many stress-regulated transcription factors targeted by PKA, and that reduced phosphorylation of those factors during stress requires the Rck2 kinase that we show physically interacts with Pde2. Together, our work shows how a high-quality computational network model can facilitate discovery of new pathway interactions during osmotic stress.
- Published
- 2018
- Full Text
- View/download PDF
27. Global mapping of CARM1 substrates defines enzyme specificity and substrate recognition.
- Author
-
Shishkova E, Zeng H, Liu F, Kwiecien NW, Hebert AS, Coon JJ, and Xu W
- Subjects
- Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Arginine metabolism, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Chromatography, High Pressure Liquid methods, Computational Biology, Enzyme Inhibitors, Female, Gene Knockout Techniques, HEK293 Cells, Humans, MCF-7 Cells, Methylation, Molecular Targeted Therapy methods, Mutation, Proline chemistry, Proline metabolism, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Protein-Arginine N-Methyltransferases chemistry, Protein-Arginine N-Methyltransferases genetics, Substrate Specificity physiology, Tandem Mass Spectrometry methods, Amino Acid Motifs physiology, Protein Processing, Post-Translational physiology, Protein-Arginine N-Methyltransferases metabolism
- Abstract
Protein arginine methyltransferases (PRMTs) introduce arginine methylation, a post-translational modification with the increasingly eminent role in normal physiology and disease. PRMT4 or coactivator-associated arginine methyltransferase 1 (CARM1) is a propitious target for cancer therapy; however, few CARM1 substrates are known, and its mechanism of substrate recognition is poorly understood. Here we employed a quantitative mass spectrometry approach to globally profile CARM1 substrates in breast cancer cell lines. We identified >130 CARM1 protein substrates and validated in vitro >90% of sites they encompass. Bioinformatics analyses reveal enrichment of proline-containing motifs, in which both methylation sites and their proximal sequences are frequently targeted by somatic mutations in cancer. Finally, we demonstrate that the N-terminus of CARM1 is involved in substrate recognition and nearly indispensable for substrate methylation. We propose that development of CARM1-specific inhibitors should focus on its N-terminus and predict that other PRMTs may employ similar mechanism for substrate recognition.
- Published
- 2017
- Full Text
- View/download PDF
28. Now, More Than Ever, Proteomics Needs Better Chromatography.
- Author
-
Shishkova E, Hebert AS, and Coon JJ
- Subjects
- Animals, Chromatography, Mass Spectrometry, Peptides, Proteome, Proteomics
- Abstract
From plant research to biomedicine, proteome analysis plays a critical role in many areas of biological inquiry. Steady improvement in mass spectrometer (MS) technology has transformed the speed and depth of proteome analysis. Proteomes of simple organisms can now be sequenced to near completion in just over an hour. Comparable coverage of mammalian proteomes, however, still requires hours or even days of analysis. Here we ask why current technology fails to achieve comprehensive and rapid analysis of the more complex mammalian proteomes. We propose that further advancements in MS technology alone are unlikely to solve this problem and suggest that concomitant improvements in peptide separation technology will be critical., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.