Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Mol Cell ; 65(2): 361-370, 2017 Jan 19.
Article in English | MEDLINE | ID: mdl-28065596

ABSTRACT

Targeted mass spectrometry assays for protein quantitation monitor peptide surrogates, which are easily multiplexed to target many peptides in a single assay. However, these assays have generally not taken advantage of sample multiplexing, which allows up to ten analyses to occur in parallel. We present a two-dimensional multiplexing workflow that utilizes synthetic peptides for each protein to prompt the simultaneous quantification of >100 peptides from up to ten mixed sample conditions. We demonstrate that targeted analysis of unfractionated lysates (2 hr) accurately reproduces the quantification of fractionated lysates (72 hr analysis) while obviating the need for peptide detection prior to quantification. We targeted 131 peptides corresponding to 69 proteins across all 60 National Cancer Institute cell lines in biological triplicate, analyzing 180 samples in only 48 hr (the equivalent of 16 min/sample). These data further elucidated a correlation between the expression of key proteins and their cellular response to drug treatment.


Subject(s)
High-Throughput Screening Assays , Mass Spectrometry , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Proteome , Proteomics/methods , Antibiotics, Antineoplastic/pharmacology , Biomarkers/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Time Factors , Transcription Factors/metabolism , Workflow
2.
Mol Cell ; 57(5): 873-886, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25684204

ABSTRACT

BCL-2 is a negative regulator of apoptosis implicated in homeostatic and pathologic cell survival. The canonical anti-apoptotic mechanism involves entrapment of activated BAX by a groove on BCL-2, preventing BAX homo-oligomerization and mitochondrial membrane poration. The BCL-2 BH4 domain also confers anti-apoptotic functionality, but the mechanism is unknown. We find that a synthetic α-helical BH4 domain binds to BAX with nanomolar affinity and independently inhibits the conformational activation of BAX. Hydrogen-deuterium exchange mass spectrometry demonstrated that the N-terminal conformational changes in BAX induced by a triggering BIM BH3 helix were suppressed by the BCL-2 BH4 helix. Structural analyses localized the BH4 interaction site to a groove formed by residues of α1, α1-α2 loop, and α2-α3 and α5-α6 hairpins on the BAX surface. These data reveal a previously unappreciated binding site for targeted inhibition of BAX and suggest that the BCL-2 BH4 domain may participate in apoptosis blockade by a noncanonical interaction mechanism.


Subject(s)
Apoptosis , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/chemistry , bcl-2-Associated X Protein/chemistry , Amino Acid Sequence , Binding Sites/genetics , Deuterium Exchange Measurement/methods , HeLa Cells , Humans , Mass Spectrometry/methods , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Conformation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
3.
RNA ; 20(6): 867-81, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24713849

ABSTRACT

RNA is a central component of gene-silencing pathways that regulate diverse cellular processes. In the fission yeast Schizosaccharomyces pombe, an RNA-based mechanism represses meiotic gene expression during vegetative growth. This pathway depends on the zinc finger protein Red1, which is required to degrade meiotic mRNAs as well as to target histone H3 lysine 9 (H3K9) methylation, a repressive chromatin mark, to a subset of meiotic genes. However, the mechanism of Red1 function is unknown. Here we use affinity purification and mass spectrometry to identify a Red1-containing nuclear RNA silencing (NURS) complex. In addition to Red1, this complex includes the Mtl1, Red5, Ars2, Rmn1, and Iss10 proteins and associates with several other complexes that are involved in either signaling or mediating RNA silencing. By analyzing the effects of gene knockouts and inducible knockdown alleles, we show that NURS subunits regulate RNA degradation and H3K9 methylation at meiotic genes. We also identify roles for individual NURS subunits in interactions with Mmi1, an RNA-binding protein that marks meiotic RNAs for destruction, and the nuclear exosome RNA degradation complex. Finally, we show that the levels of H3K9 methylation at meiotic genes are not sufficient to restrict RNA polymerase II access or repress gene expression during vegetative growth. Our results demonstrate that Red1 partners with other proteins to silence meiotic gene expression at the post-transcriptional level. Conservation of a NURS-like complex in human cells suggests that this pathway plays an ancient and fundamental role in RNA silencing.


Subject(s)
Meiosis/genetics , RNA Interference/physiology , RNA, Nuclear/genetics , Carrier Proteins/genetics , Chromatin/genetics , Exosomes/genetics , Gene Expression Regulation, Fungal/genetics , Histones/genetics , Methylation , RNA Polymerase II/genetics , RNA Stability/genetics , RNA, Messenger/genetics , Schizosaccharomyces pombe Proteins/genetics , mRNA Cleavage and Polyadenylation Factors/genetics
4.
Proc Natl Acad Sci U S A ; 110(11): E986-95, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23404709

ABSTRACT

Proapoptotic B-cell lymphoma 2 (BCL-2) antagonist/killer (BAK) and BCL-2-associated X (BAX) form toxic mitochondrial pores in response to cellular stress. Whereas BAX resides predominantly in the cytosol, BAK is constitutively localized to the outer mitochondrial membrane. Select BCL-2 homology domain 3 (BH3) helices activate BAX directly by engaging an α1/α6 trigger site. The inability to express full-length BAK has hampered full dissection of its activation mechanism. Here, we report the production of full-length, monomeric BAK by mutagenesis-based solubilization of its C-terminal α-helical surface. Recombinant BAK autotranslocates to mitochondria but only releases cytochrome c upon BH3 triggering. A direct activation mechanism was explicitly demonstrated using a liposomal system that recapitulates BAK-mediated release upon addition of BH3 ligands. Photoreactive BH3 helices mapped both triggering and autointeractions to the canonical BH3-binding pocket of BAK, whereas the same ligands crosslinked to the α1/α6 site of BAX. Thus, activation of both BAK and BAX is initiated by direct BH3-interaction but at distinct trigger sites. These structural and biochemical insights provide opportunities for developing proapoptotic agents that activate the death pathway through direct but differential engagement of BAK and BAX.


Subject(s)
Mitochondria, Liver/chemistry , Mitochondria, Liver/metabolism , bcl-2 Homologous Antagonist-Killer Protein/chemistry , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Animals , Apoptosis/physiology , Humans , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/chemistry , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
5.
Anal Chem ; 87(19): 9855-63, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26308379

ABSTRACT

Isobaric labeling strategies for mass spectrometry-based proteomics enable multiplexed simultaneous quantification of samples and therefore substantially increase the sample throughput in proteomics. However, despite these benefits, current limits to multiplexing capacity are prohibitive for large sample sizes and impose limitations on experimental design. Here, we introduce a novel mechanism for increasing the multiplexing density of isobaric reagents. We present Combinatorial Isobaric Mass Tags (CMTs), an isobaric labeling architecture with the unique ability to generate multiple series of reporter ions simultaneously. We demonstrate that utilization of multiple reporter ion series improves multiplexing capacity of CMT with respect to a commercially available isobaric labeling reagent with preserved quantitative accuracy and depth of coverage in complex mixtures. We provide a blueprint for the realization of 16-plex reagents with 1 Da spacing between reporter ions and up to 28-plex at 6 mDa spacing using only 5 heavy isotopes per reagent. We anticipate that this improvement in multiplexing capacity will further advance the application of quantitative proteomics, particularly in high-throughput screening assays.


Subject(s)
Mass Spectrometry/methods , Peptides/analysis , Proteomics/methods , High-Throughput Screening Assays/methods , Indicators and Reagents/chemistry , Ions/chemistry
6.
Cell Metab ; 23(5): 867-80, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27133130

ABSTRACT

Mammary epithelial cells transition between periods of proliferation and quiescence during development, menstrual cycles, and pregnancy, and as a result of oncogenic transformation. Utilizing an organotypic 3D tissue culture model coupled with quantitative metabolomics and proteomics, we identified significant differences in glutamate utilization between proliferating and quiescent cells. Relative to quiescent cells, proliferating cells catabolized more glutamate via transaminases to couple non-essential amino acid (NEAA) synthesis to α-ketoglutarate generation and tricarboxylic acid (TCA) cycle anaplerosis. As cells transitioned to quiescence, glutamine consumption and transaminase expression were reduced, while glutamate dehydrogenase (GLUD) was induced, leading to decreased NEAA synthesis. Highly proliferative human tumors display high transaminase and low GLUD expression, suggesting that proliferating cancer cells couple glutamine consumption to NEAA synthesis to promote biosynthesis. These findings describe a competitive and partially redundant relationship between transaminases and GLUD, and they reveal how coupling of glutamate-derived carbon and nitrogen metabolism can be regulated to support cell proliferation.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Glutamic Acid/metabolism , Mammary Glands, Human/cytology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Proliferation , Cells, Cultured , Female , Glutamate Dehydrogenase/metabolism , Humans , Metabolomics , Models, Biological , Nitrogen Isotopes , Phosphatidylinositol 3-Kinases/metabolism , Proteomics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transaminases/metabolism
7.
Methods Enzymol ; 544: 25-48, 2014.
Article in English | MEDLINE | ID: mdl-24974285

ABSTRACT

Protein interactions dictate a myriad of cellular activities that maintain health or cause disease. Dissecting these binding partnerships, and especially their sites of interaction, fuels the discovery of signaling pathways, disease mechanisms, and next-generation therapeutics. We previously applied all-hydrocarbon peptide stapling to chemically restore α-helical shape to bioactive motifs that become unfolded when taken out of context from native signaling proteins. For example, we developed stabilized alpha-helices of BCL-2 domains (SAHBs) to dissect and target protein interactions of the BCL-2 family, a critical network that regulates the apoptotic pathway. SAHBs are α-helical surrogates that bind both stable and transient physiologic interactors and have effectively uncovered novel sites of BCL-2 family protein interaction. To leverage stapled peptides for proteomic discovery, we describe our conversion of SAHBs into photoreactive agents that irreversibly capture their protein targets and facilitate rapid identification of the peptide helix binding sites. We envision that the development of photoreactive stapled peptides will accelerate the discovery of novel and unanticipated protein interactions and how they impact health and disease.


Subject(s)
Mass Spectrometry/methods , Peptides/metabolism , Photosensitizing Agents/metabolism , Protein Interaction Mapping/methods , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Animals , Apoptosis , Binding Sites , Humans , Models, Molecular , Molecular Sequence Data , Peptides/analysis , Photosensitizing Agents/analysis , Protein Structure, Secondary , Protein Structure, Tertiary , Proteomics , Proto-Oncogene Proteins c-bcl-2/analysis
8.
Nat Struct Mol Biol ; 21(1): 36-42, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24317490

ABSTRACT

Glucokinase (GK) is a glucose-phosphorylating enzyme that regulates insulin release and hepatic metabolism, and its loss of function is implicated in diabetes pathogenesis. GK activators (GKAs) are attractive therapeutics in diabetes; however, clinical data indicate that their benefits can be offset by hypoglycemia, owing to marked allosteric enhancement of the enzyme's glucose affinity. We show that a phosphomimetic of the BCL-2 homology 3 (BH3) α-helix derived from human BAD, a GK-binding partner, increases the enzyme catalytic rate without dramatically changing glucose affinity, thus providing a new mechanism for pharmacologic activation of GK. Remarkably, BAD BH3 phosphomimetic mediates these effects by engaging a new region near the enzyme's active site. This interaction increases insulin secretion in human islets and restores the function of naturally occurring human GK mutants at the active site. Thus, BAD phosphomimetics may serve as a new class of GKAs.


Subject(s)
Glucokinase/metabolism , Phosphoproteins/physiology , Allosteric Regulation , Animals , Enzyme Activation , Humans
9.
Chem Biol ; 20(7): 888-902, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23890007

ABSTRACT

PUMA is a proapoptotic BCL-2 family member that drives the apoptotic response to a diversity of cellular insults. Deciphering the spectrum of PUMA interactions that confer its context-dependent proapoptotic properties remains a high priority goal. Here, we report the synthesis of PUMA SAHBs, structurally stabilized PUMA BH3 helices that, in addition to broadly targeting antiapoptotic proteins, directly bind to proapoptotic BAX. NMR, photocrosslinking, and biochemical analyses revealed that PUMA SAHBs engage an α1/α6 trigger site on BAX to initiate its functional activation. We further demonstrated that a cell-permeable PUMA SAHB analog induces apoptosis in neuroblastoma cells and, like expressed PUMA protein, engages BCL-2, MCL-1, and BAX. Thus, we find that PUMA BH3 is a dual antiapoptotic inhibitor and proapoptotic direct activator, and its mimetics may serve as effective pharmacologic triggers of apoptosis in resistant human cancers.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Animals , Caspase 3/metabolism , Caspase 7/metabolism , Enzyme Activation , HEK293 Cells , Humans , Mice , Models, Molecular , Molecular Sequence Data , Porosity , Protein Structure, Tertiary , Proteomics , Substrate Specificity , bcl-2-Associated X Protein/metabolism
10.
Mol Cell Biol ; 33(8): 1487-502, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23382074

ABSTRACT

Although many functions and targets have been attributed to the histone and protein deacetylase SIRT1, a comprehensive analysis of SIRT1 binding proteins yielding a high-confidence interaction map has not been established. Using a comparative statistical analysis of binding partners, we have assembled a high-confidence SIRT1 interactome. Employing this method, we identified the deubiquitinating enzyme ubiquitin-specific protease 22 (USP22), a component of the deubiquitinating module (DUBm) of the SAGA transcriptional coactivating complex, as a SIRT1-interacting partner. We found that this interaction is highly specific, requires the ZnF-UBP domain of USP22, and is disrupted by the inactivating H363Y mutation within SIRT1. Moreover, we show that USP22 is acetylated on multiple lysine residues and that alteration of a single lysine (K129) within the ZnF-UBP domain is sufficient to alter interaction of the DUBm with the core SAGA complex. Furthermore, USP22-mediated recruitment of SIRT1 activity promotes the deacetylation of individual SAGA complex components. Our results indicate an important role of SIRT1-mediated deacetylation in regulating the formation of DUBm subcomplexes within the larger SAGA complex.


Subject(s)
Protein Interaction Maps , Sirtuin 1/metabolism , Thiolester Hydrolases/metabolism , Acetylation , Cell Line , HEK293 Cells , Humans , Mutation , Protein Binding , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering , Sirtuin 1/genetics , Thiolester Hydrolases/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Ubiquitin Thiolesterase , Ubiquitination
11.
Chem Biol ; 17(12): 1325-33, 2010 Dec 22.
Article in English | MEDLINE | ID: mdl-21168768

ABSTRACT

Defining protein interactions forms the basis for discovery of biological pathways, disease mechanisms, and opportunities for therapeutic intervention. To harness the robust binding affinity and selectivity of structured peptides for interactome discovery, we engineered photoreactive stapled BH3 peptide helices that covalently capture their physiologic BCL-2 family targets. The crosslinking α helices covalently trap both static and dynamic protein interactors, and enable rapid identification of interaction sites, providing a critical link between interactome discovery and targeted drug design.


Subject(s)
Peptides/chemistry , Proto-Oncogene Proteins c-bcl-2/chemistry , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Humans , Molecular Sequence Data , Peptides/chemical synthesis , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/metabolism , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL