Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Mol Cell ; 82(2): 435-446.e7, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34847359

ABSTRACT

Most mitochondrial proteins are translated in the cytosol and imported into mitochondria. Mutations in the mitochondrial protein import machinery cause human pathologies. However, a lack of suitable tools to measure protein uptake across the mitochondrial proteome has prevented the identification of specific proteins affected by import perturbation. Here, we introduce mePRODmt, a pulsed-SILAC based proteomics approach that includes a booster signal to increase the sensitivity for mitochondrial proteins selectively, enabling global dynamic analysis of endogenous mitochondrial protein uptake in cells. We applied mePRODmt to determine protein uptake kinetics and examined how inhibitors of mitochondrial import machineries affect protein uptake. Monitoring changes in translation and uptake upon mitochondrial membrane depolarization revealed that protein uptake was extensively modulated by the import and translation machineries via activation of the integrated stress response. Strikingly, uptake changes were not uniform, with subsets of proteins being unaffected or decreased due to changes in translation or import capacity.


Subject(s)
Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Biosynthesis , Proteome , Proteomics , Carbonyl Cyanide m-Chlorophenyl Hydrazone/pharmacology , Electron Transport Complex I/metabolism , Female , HeLa Cells , Humans , Kinetics , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Protein Biosynthesis/drug effects , Protein Transport , Uncoupling Agents/pharmacology
2.
Nature ; 607(7919): 593-603, 2022 07.
Article in English | MEDLINE | ID: mdl-35768510

ABSTRACT

Aggressive and metastatic cancers show enhanced metabolic plasticity1, but the precise underlying mechanisms of this remain unclear. Here we show how two NOP2/Sun RNA methyltransferase 3 (NSUN3)-dependent RNA modifications-5-methylcytosine (m5C) and its derivative 5-formylcytosine (f5C) (refs.2-4)-drive the translation of mitochondrial mRNA to power metastasis. Translation of mitochondrially encoded subunits of the oxidative phosphorylation complex depends on the formation of m5C at position 34 in mitochondrial tRNAMet. m5C-deficient human oral cancer cells exhibit increased levels of glycolysis and changes in their mitochondrial function that do not affect cell viability or primary tumour growth in vivo; however, metabolic plasticity is severely impaired as mitochondrial m5C-deficient tumours do not metastasize efficiently. We discovered that CD36-dependent non-dividing, metastasis-initiating tumour cells require mitochondrial m5C to activate invasion and dissemination. Moreover, a mitochondria-driven gene signature in patients with head and neck cancer is predictive for metastasis and disease progression. Finally, we confirm that this metabolic switch that allows the metastasis of tumour cells can be pharmacologically targeted through the inhibition of mitochondrial mRNA translation in vivo. Together, our results reveal that site-specific mitochondrial RNA modifications could be therapeutic targets to combat metastasis.


Subject(s)
5-Methylcytosine , Cytosine/analogs & derivatives , Glycolysis , Mitochondria , Neoplasm Metastasis , Oxidative Phosphorylation , RNA, Mitochondrial , 5-Methylcytosine/biosynthesis , 5-Methylcytosine/metabolism , CD36 Antigens , Cell Survival , Cytosine/metabolism , Disease Progression , Glycolysis/drug effects , Humans , Methylation/drug effects , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Oxidative Phosphorylation/drug effects , Protein Biosynthesis/drug effects , RNA, Mitochondrial/genetics , RNA, Mitochondrial/metabolism , RNA, Transfer, Met/genetics , RNA, Transfer, Met/metabolism
3.
Mol Cell ; 77(4): 913-925.e4, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31812349

ABSTRACT

Regulation of translation is essential during stress. However, the precise sets of proteins regulated by the key translational stress responses-the integrated stress response (ISR) and mTORC1-remain elusive. We developed multiplexed enhanced protein dynamics (mePROD) proteomics, adding signal amplification to dynamic-SILAC and multiplexing, to enable measuring acute changes in protein synthesis. Treating cells with ISR/mTORC1-modulating stressors, we showed extensive translatome modulation with ∼20% of proteins synthesized at highly reduced rates. Comparing translation-deficient sub-proteomes revealed an extensive overlap demonstrating that target specificity is achieved on protein level and not by pathway activation. Titrating cap-dependent translation inhibition confirmed that synthesis of individual proteins is controlled by intrinsic properties responding to global translation attenuation. This study reports a highly sensitive method to measure relative translation at the nascent chain level and provides insight into how the ISR and mTORC1, two key cellular pathways, regulate the translatome to guide cellular survival upon stress.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Protein Biosynthesis , Proteomics/methods , HeLa Cells , Humans , Stress, Physiological/genetics , Unfolded Protein Response
4.
Mol Cell ; 80(1): 164-174.e4, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32877642

ABSTRACT

SARS-CoV-2 infections are rapidly spreading around the globe. The rapid development of therapies is of major importance. However, our lack of understanding of the molecular processes and host cell signaling events underlying SARS-CoV-2 infection hinders therapy development. We use a SARS-CoV-2 infection system in permissible human cells to study signaling changes by phosphoproteomics. We identify viral protein phosphorylation and define phosphorylation-driven host cell signaling changes upon infection. Growth factor receptor (GFR) signaling and downstream pathways are activated. Drug-protein network analyses revealed GFR signaling as key pathways targetable by approved drugs. The inhibition of GFR downstream signaling by five compounds prevents SARS-CoV-2 replication in cells, assessed by cytopathic effect, viral dsRNA production, and viral RNA release into the supernatant. This study describes host cell signaling events upon SARS-CoV-2 infection and reveals GFR signaling as a central pathway essential for SARS-CoV-2 replication. It provides novel strategies for COVID-19 treatment.


Subject(s)
Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Mitogen-Activated Protein Kinases/genetics , Phosphatidylinositol 3-Kinase/genetics , Receptors, Growth Factor/genetics , Viral Proteins/genetics , Adrenal Cortex Hormones/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Antibodies, Neutralizing/therapeutic use , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , Caco-2 Cells , Gene Expression Regulation , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/metabolism , SARS-CoV-2 , Signal Transduction , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Virus Replication/drug effects
5.
Nature ; 583(7816): 469-472, 2020 07.
Article in English | MEDLINE | ID: mdl-32408336

ABSTRACT

A new coronavirus was recently discovered and named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Infection with SARS-CoV-2 in humans causes coronavirus disease 2019 (COVID-19) and has been rapidly spreading around the globe1,2. SARS-CoV-2 shows some similarities to other coronaviruses; however, treatment options and an understanding of how SARS-CoV-2 infects cells are lacking. Here we identify the host cell pathways that are modulated by SARS-CoV-2 and show that inhibition of these pathways prevents viral replication in human cells. We established a human cell-culture model for infection with a clinical isolate of SARS-CoV-2. Using this cell-culture system, we determined the infection profile of SARS-CoV-2 by translatome3 and proteome proteomics at different times after infection. These analyses revealed that SARS-CoV-2 reshapes central cellular pathways such as translation, splicing, carbon metabolism, protein homeostasis (proteostasis) and nucleic acid metabolism. Small-molecule inhibitors that target these pathways prevented viral replication in cells. Our results reveal the cellular infection profile of SARS-CoV-2 and have enabled the identification of drugs that inhibit viral replication. We anticipate that our results will guide efforts to understand the molecular mechanisms that underlie the modulation of host cells after infection with SARS-CoV-2. Furthermore, our findings provide insights for the development of therapies for the treatment of COVID-19.


Subject(s)
Betacoronavirus/drug effects , Betacoronavirus/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/metabolism , Molecular Targeted Therapy , Pneumonia, Viral/drug therapy , Pneumonia, Viral/metabolism , Proteomics , Betacoronavirus/genetics , Betacoronavirus/growth & development , COVID-19 , Caco-2 Cells , Carbon/metabolism , Coronavirus Infections/genetics , Coronavirus Infections/virology , Drug Evaluation, Preclinical , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , In Vitro Techniques , Kinetics , Pandemics , Pneumonia, Viral/genetics , Pneumonia, Viral/virology , Protein Biosynthesis/drug effects , Proteome/metabolism , Proteostasis , RNA Splicing , SARS-CoV-2 , Time Factors , Virus Replication/drug effects , COVID-19 Drug Treatment
6.
Mol Cell Proteomics ; 22(5): 100537, 2023 05.
Article in English | MEDLINE | ID: mdl-37001587

ABSTRACT

The ancestral SARS-CoV-2 strain that initiated the Covid-19 pandemic at the end of 2019 has rapidly mutated into multiple variants of concern with variable pathogenicity and increasing immune escape strategies. However, differences in host cellular antiviral responses upon infection with SARS-CoV-2 variants remain elusive. Leveraging whole-cell proteomics, we determined host signaling pathways that are differentially modulated upon infection with the clinical isolates of the ancestral SARS-CoV-2 B.1 and the variants of concern Delta and Omicron BA.1. Our findings illustrate alterations in the global host proteome landscape upon infection with SARS-CoV-2 variants and the resulting host immune responses. Additionally, viral proteome kinetics reveal declining levels of viral protein expression during Omicron BA.1 infection when compared to ancestral B.1 and Delta variants, consistent with its reduced replication rates. Moreover, molecular assays reveal deferral activation of specific host antiviral signaling upon Omicron BA.1 and BA.2 infections. Our study provides an overview of host proteome profile of multiple SARS-CoV-2 variants and brings forth a better understanding of the instigation of key immune signaling pathways causative for the differential pathogenicity of SARS-CoV-2 variants.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Proteome , Pandemics , Antiviral Agents , Antibodies, Neutralizing
7.
Nature ; 561(7722): 268-272, 2018 09.
Article in English | MEDLINE | ID: mdl-30158700

ABSTRACT

The folding of newly synthesized proteins to the native state is a major challenge within the crowded cellular environment, as non-productive interactions can lead to misfolding, aggregation and degradation1. Cells cope with this challenge by coupling synthesis with polypeptide folding and by using molecular chaperones to safeguard folding cotranslationally2. However, although most of the cellular proteome forms oligomeric assemblies3, little is known about the final step of folding: the assembly of polypeptides into complexes. In prokaryotes, a proof-of-concept study showed that the assembly of heterodimeric luciferase is an organized cotranslational process that is facilitated by spatially confined translation of the subunits encoded on a polycistronic mRNA4. In eukaryotes, however, fundamental differences-such as the rarity of polycistronic mRNAs and different chaperone constellations-raise the question of whether assembly is also coordinated with translation. Here we provide a systematic and mechanistic analysis of the assembly of protein complexes in eukaryotes using ribosome profiling. We determined the in vivo interactions of the nascent subunits from twelve hetero-oligomeric protein complexes of Saccharomyces cerevisiae at near-residue resolution. We find nine complexes assemble cotranslationally; the three complexes that do not show cotranslational interactions are regulated by dedicated assembly chaperones5-7. Cotranslational assembly often occurs uni-directionally, with one fully synthesized subunit engaging its nascent partner subunit, thereby counteracting its propensity for aggregation. The onset of cotranslational subunit association coincides directly with the full exposure of the nascent interaction domain at the ribosomal tunnel exit. The action of the ribosome-associated Hsp70 chaperone Ssb8 is coordinated with assembly. Ssb transiently engages partially synthesized interaction domains and then dissociates before the onset of partner subunit association, presumably to prevent premature assembly interactions. Our study shows that cotranslational subunit association is a prevalent mechanism for the assembly of hetero-oligomers in yeast and indicates that translation, folding and the assembly of protein complexes are integrated processes in eukaryotes.


Subject(s)
Amino Acyl-tRNA Synthetases/biosynthesis , Fatty Acid Synthases/biosynthesis , Multiprotein Complexes/biosynthesis , Multiprotein Complexes/chemistry , Protein Biosynthesis , Ribosomes/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , Fatty Acid Synthases/chemistry , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , HSP70 Heat-Shock Proteins/metabolism , Models, Molecular , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Protein Binding , Protein Subunits/biosynthesis , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Article in English | MEDLINE | ID: mdl-33947818

ABSTRACT

Salmonella is an intracellular pathogen of a substantial global health concern. In order to identify key players involved in Salmonella infection, we performed a global host phosphoproteome analysis subsequent to bacterial infection. Thereby, we identified the kinase SIK2 as a central component of the host defense machinery upon Salmonella infection. SIK2 depletion favors the escape of bacteria from the Salmonella-containing vacuole (SCV) and impairs Xenophagy, resulting in a hyperproliferative phenotype. Mechanistically, SIK2 associates with actin filaments under basal conditions; however, during bacterial infection, SIK2 is recruited to the SCV together with the elements of the actin polymerization machinery (Arp2/3 complex and Formins). Notably, SIK2 depletion results in a severe pathological cellular actin nucleation and polymerization defect upon Salmonella infection. We propose that SIK2 controls the formation of a protective SCV actin shield shortly after invasion and orchestrates the actin cytoskeleton architecture in its entirety to control an acute Salmonella infection after bacterial invasion.


Subject(s)
Actins/metabolism , Epithelial Cells/metabolism , Protein Interaction Maps , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Animals , Cells, Cultured , Epithelial Cells/microbiology , HCT116 Cells , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Immunoblotting , Mice , Phosphoproteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proteomics/methods , RNA Interference , Salmonella/physiology
9.
Int J Mol Sci ; 24(23)2023 Nov 30.
Article in English | MEDLINE | ID: mdl-38069304

ABSTRACT

Despite the importance of rapid adaptive responses in the course of inflammation and the notion that post-transcriptional regulation plays an important role herein, relevant translational alterations, especially during the resolution phase, remain largely elusive. In the present study, we analyzed translational changes in inflammatory bone marrow-derived macrophages upon resolution-promoting efferocytosis. Total RNA-sequencing confirmed that apoptotic cell phagocytosis induced a pro-resolution signature in LPS/IFNγ-stimulated macrophages (Mϕ). While inflammation-dependent transcriptional changes were relatively small between efferocytic and non-efferocytic Mϕ; considerable differences were observed at the level of de novo synthesized proteins. Interestingly, translationally regulated targets in response to inflammatory stimuli were mostly downregulated, with only minimal impact of efferocytosis. Amongst these targets, pro-resolving matrix metallopeptidase 12 (Mmp12) was identified as a translationally repressed candidate during early inflammation that recovered during the resolution phase. Functionally, reduced MMP12 production enhanced matrix-dependent migration of Mϕ. Conclusively, translational control of MMP12 emerged as an efficient strategy to alter the migratory properties of Mϕ throughout the inflammatory response, enabling Mϕ migration within the early inflammatory phase while restricting migration during the resolution phase.


Subject(s)
Matrix Metalloproteinase 12 , Phagocytosis , Humans , Matrix Metalloproteinase 12/genetics , Matrix Metalloproteinase 12/metabolism , Phagocytosis/physiology , Macrophages/metabolism , Inflammation/metabolism , Gene Expression Regulation , Apoptosis/physiology
10.
J Cell Biochem ; 123(3): 691-696, 2022 03.
Article in English | MEDLINE | ID: mdl-35132673

ABSTRACT

Here, we present a peptide-based linear mixed models tool-PBLMM, a standalone desktop application for differential expression analysis of proteomics data. We also provide a Python package that allows streamlined data analysis workflows implementing the PBLMM algorithm. PBLMM is easy to use without scripting experience and calculates differential expression by peptide-based linear mixed regression models. We show that peptide-based models outperform classical methods of statistical inference of differentially expressed proteins. In addition, PBLMM exhibits superior statistical power in situations of low effect size and/or low sample size. Taken together our tool provides an easy-to-use, high-statistical-power method to infer differentially expressed proteins from proteomics data.


Subject(s)
Peptides , Proteomics , Algorithms , Linear Models , Peptides/analysis , Peptides/genetics , Proteins , Proteomics/methods , Software
11.
Blood ; 133(2): 168-179, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30498063

ABSTRACT

Autophagy maintains hematopoietic stem cell integrity and prevents malignant transformation. In addition to bulk degradation, selective autophagy serves as an intracellular quality control mechanism and requires autophagy receptors, such as p62 (SQSTM1), to specifically bridge the ubiquitinated cargos into autophagosomes. Here, we investigated the function of p62 in acute myeloid leukemia (AML) in vitro and in murine in vivo models of AML. Loss of p62 impaired expansion and colony-forming ability of leukemia cells and prolonged latency of leukemia development in mice. High p62 expression was associated with poor prognosis in human AML. Using quantitative mass spectrometry, we identified enrichment of mitochondrial proteins upon immunoprecipitation of p62. Loss of p62 significantly delayed removal of dysfunctional mitochondria, increased mitochondrial superoxide levels, and impaired mitochondrial respiration. Moreover, we demonstrated that the autophagy-dependent function of p62 is essential for cell growth and effective mitochondrial degradation by mitophagy. Our results highlight the prominent role of selective autophagy in leukemia progression, and specifically, the importance of mitophagy to maintain mitochondrial integrity.


Subject(s)
Autophagy , Leukemia, Experimental/pathology , Leukemia, Myeloid, Acute/pathology , Mitophagy , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/physiology , Animals , Follow-Up Studies , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Knockout , Prognosis , Survival Rate , Tumor Cells, Cultured
12.
Int J Mol Sci ; 22(6)2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33808970

ABSTRACT

Cell-free therapy using extracellular vesicles (EVs) from adipose-derived mesenchymal stromal/stem cells (ASCs) seems to be a safe and effective therapeutic option to support tissue and organ regeneration. The application of EVs requires particles with a maximum regenerative capability and hypoxic culture conditions as an in vitro preconditioning regimen has been shown to alter the molecular composition of released EVs. Nevertheless, the EV cargo after hypoxic preconditioning has not yet been comprehensively examined. The aim of the present study was the characterization of EVs from hypoxic preconditioned ASCs. We investigated the EV proteome and their effects on renal tubular epithelial cells in vitro. While no effect of hypoxia was observed on the number of released EVs and their protein content, the cargo of the proteins was altered. Proteomic analysis showed 41 increased or decreased proteins, 11 in a statistically significant manner. Furthermore, the uptake of EVs in epithelial cells and a positive effect on oxidative stress in vitro were observed. In conclusion, culture of ASCs under hypoxic conditions was demonstrated to be a promising in vitro preconditioning regimen, which alters the protein cargo and increases the anti-oxidative potential of EVs. These properties may provide new potential therapeutic options for regenerative medicine.


Subject(s)
Extracellular Vesicles/genetics , Proteome/genetics , Proteomics , Regenerative Medicine/methods , Cell- and Tissue-Based Therapy/trends , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Regeneration/genetics
13.
Biospektrum (Heidelb) ; 27(1): 40-45, 2021.
Article in German | MEDLINE | ID: mdl-33612988

ABSTRACT

Upon infection with SARS-CoV-2, a variety of changes happen inside the host cell. The virus hijacks host cell pathways for driving its own replication, while the host counteracts with response mechanisms. To gain a comprehensive understanding of COVID-19, caused by SARS-CoV-2 infection, and develop therapeutic strategies, it is crucial to observe these systematic changes in their entirety. In our recent studies, we followed the effects of SARS-CoV-2 infection on the human proteome, which led to the identification of several drugs that abolished viral proliferation in cells.

14.
Br J Cancer ; 122(9): 1391-1398, 2020 04.
Article in English | MEDLINE | ID: mdl-32203214

ABSTRACT

BACKGROUND: The amino acid serine is an important substrate for biosynthesis and redox homeostasis. We investigated whether glioblastoma (GBM) cells are dependent on serine for survival under conditions of the tumour microenvironment. METHODS: Serine availability in GBM cells was modulated pharmacologically, genetically and by adjusting serine and glycine concentrations in the culture medium. Cells were investigated for regulation of serine metabolism, proliferation, sensitivity to hypoxia-induced cell death and redox homeostasis. RESULTS: Hypoxia-induced expression of phosphoglycerate dehydrogenase (PHGDH) and the mitochondrial serine hydroxymethyltransferase (SHMT2) was observed in three of five tested glioma cell lines. Nuclear factor erythroid 2-related factor (Nrf) 2 activation also induced PHGDH and SHMT2 expression in GBM cells. Low levels of endogenous PHGDH as well as PHGDH gene suppression resulted in serine dependency for cell growth. Pharmacological inhibition of PHGDH with CBR-5884 reduced proliferation and sensitised cells profoundly to hypoxia-induced cell death. This effect was accompanied by an increase in reactive oxygen species and a decrease in the NADPH/NADP+ ratio. Similarly, hypoxia-induced cell death was enhanced by PHGDH gene suppression and reduced by PHGDH overexpression. CONCLUSIONS: Serine facilitates adaptation of GBM cells to conditions of the tumour microenvironment and its metabolism could be a plausible therapeutic target.


Subject(s)
Glioblastoma/metabolism , Glycine Hydroxymethyltransferase/genetics , NF-E2-Related Factor 2/genetics , Phosphoglycerate Dehydrogenase/genetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Homeostasis/drug effects , Humans , Oxidation-Reduction/drug effects , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Serine/metabolism , Tumor Microenvironment/drug effects
15.
Anal Chem ; 92(12): 8041-8045, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32442378

ABSTRACT

Pulsed Stable Isotope Labeling in Cell culture (SILAC) approaches allow measurement of protein dynamics, including protein translation and degradation. However, its use for quantifying acute changes has been limited due to low labeled peptide stoichiometry. Here, we describe the use of instrument logic to select peaks of interest via targeted mass differences (TMD) for overcoming this limitation. Comparing peptides artificially mixed at low heavy-to-light stoichiometry measured using standard data dependent acquisition with or without TMD revealed 2-3-fold increases in identification without significant loss in quantification precision for both MS2 and MS3 methods. Our benchmarked method approach increased throughput by reducing the necessary machine time. We anticipate that all pulsed SILAC measurements, combined with tandem mass tagging (TMT) or not, would greatly benefit from instrument logic based approaches.


Subject(s)
Isotope Labeling , Neoplasm Proteins/analysis , Chromatography, Liquid , HeLa Cells , Humans , Mass Spectrometry , Neoplasm Proteins/metabolism , Tumor Cells, Cultured
17.
Cell Death Discov ; 10(1): 8, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38182566

ABSTRACT

Glioblastoma is an incurable brain tumor with a median survival below two years. Trials investigating targeted therapy with inhibitors of the kinase mTOR have produced ambiguous results. Especially combination of mTOR inhibition with standard temozolomide radiochemotherapy has resulted in reduced survival in a phase II clinical trial. To date, this phenomenon is only poorly understood. To recreate the therapeutic setting in vitro, we exposed glioblastoma cell lines to co-treatment with rapamycin and temozolomide and assessed cell viability, DNA damage and reactive oxygen species. Additionally, we employed a novel translatomic based mass spectrometry approach ("mePROD") to analyze acute changes in translated proteins. mTOR inhibition with rapamycin protected glioblastoma cells from temozolomide toxicity. Following co-treatment of temozolomide with rapamycin, an increased translation of reactive oxygen species (ROS)-detoxifying proteins was detected by mass spectrometry. This was accompanied by improved ROS-homeostasis and reduced DNA damage. Additionally, rapamycin induced the expression of the DNA repair enzyme O-6-methylguanine-DNA methyltransferase (MGMT) in glioblastoma cells with an unmethylated MGMT gene promotor. Inhibition of mTOR antagonized the cytotoxic effects of temozolomide in vitro. The induction of antioxidant defences and MGMT are two underlying candidate mechanisms. Further functional experiments in vitro and in vivo are warranted to characterize this effect that appears relevant for combinatorial therapeutic strategies.

18.
Cell Death Differ ; 30(7): 1710-1725, 2023 07.
Article in English | MEDLINE | ID: mdl-37202505

ABSTRACT

SREBP2 is a master regulator of the mevalonate pathway (MVP), a biosynthetic process that drives the synthesis of dolichol, heme A, ubiquinone and cholesterol and also provides substrates for protein prenylation. Here, we identify SREBP2 as a novel substrate for USP28, a deubiquitinating enzyme that is frequently upregulated in squamous cancers. Our results show that silencing of USP28 reduces expression of MVP enzymes and lowers metabolic flux into this pathway. We also show that USP28 binds to mature SREBP2, leading to its deubiquitination and stabilisation. USP28 depletion rendered cancer cells highly sensitive to MVP inhibition by statins, which was rescued by the addition of geranyl-geranyl pyrophosphate. Analysis of human tissue microarrays revealed elevated expression of USP28, SREBP2 and MVP enzymes in lung squamous cell carcinoma (LSCC) compared to lung adenocarcinoma (LADC). Moreover, CRISPR/Cas-mediated deletion of SREBP2 selectively attenuated tumour growth in a KRas/p53/LKB1 mutant mouse model of lung cancer. Finally, we demonstrate that statins synergise with a dual USP28/25 inhibitor to reduce viability of SCC cells. Our findings suggest that combinatorial targeting of MVP and USP28 could be a therapeutic strategy for the treatment of squamous cell carcinomas.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Lung Neoplasms , Mice , Animals , Humans , Mevalonic Acid/metabolism , Lung Neoplasms/genetics , Carcinoma, Squamous Cell/genetics , Ubiquitin Thiolesterase/metabolism
19.
Methods Mol Biol ; 2428: 75-87, 2022.
Article in English | MEDLINE | ID: mdl-35171474

ABSTRACT

Multiplexed enhanced protein dynamic mass spectrometry (mePROD MS) enables robust quantification of translation in cell culture. Tandem mass tags (TMT) are combined with pulsed stable isotope labeling in cell culture (pSILAC) to monitor newly synthesized proteins on a proteome wide scale. While approaches combining pSILAC and TMT typically require long labeling times to reach sufficient intensity of the newly synthesized peptides in the mass spectrometer, mePROD uses a carrier signal that boosts the survey scan intensity and strongly increases identification rates. Hence, this protocol provides an easy and cost-efficient method to profile proteome-wide translatome changes at a temporal resolution of minutes.


Subject(s)
Proteome , Proteomics , Isotope Labeling/methods , Mass Spectrometry , Peptides/analysis , Proteome/metabolism , Proteomics/methods
20.
Methods Mol Biol ; 2477: 179-193, 2022.
Article in English | MEDLINE | ID: mdl-35524118

ABSTRACT

Selective Ribosome Profiling (SeRP) is an emerging methodology, developed to capture cotranslational interactions in vivo. To date, SeRP is the only method that can directly capture, in near-codon resolution, ribosomes in action. Thus, SeRP allows us to study the mechanisms of protein synthesis and the network of protein-protein interactions that are formed already during synthesis. Here we report, in detail, the protocol for purification of ribosome- and Nascent-Chain associated factors, followed by isolation of ribosome-protected mRNA footprints, cDNA library generation and subsequent data analysis.


Subject(s)
Protein Biosynthesis , Ribosomes , Codon/metabolism , Gene Library , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL