Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Cell ; 184(20): 5215-5229.e17, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34559986

ABSTRACT

Estrogen receptor α (ERα) is a hormone receptor and key driver for over 70% of breast cancers that has been studied for decades as a transcription factor. Unexpectedly, we discover that ERα is a potent non-canonical RNA-binding protein. We show that ERα RNA binding function is uncoupled from its activity to bind DNA and critical for breast cancer progression. Employing genome-wide cross-linking immunoprecipitation (CLIP) sequencing and a functional CRISPRi screen, we find that ERα-associated mRNAs sustain cancer cell fitness and elicit cellular responses to stress. Mechanistically, ERα controls different steps of RNA metabolism. In particular, we demonstrate that ERα RNA binding mediates alternative splicing of XBP1 and translation of the eIF4G2 and MCL1 mRNAs, which facilitates survival upon stress conditions and sustains tamoxifen resistance of cancer cells. ERα is therefore a multifaceted RNA-binding protein, and this activity transforms our knowledge of post-transcriptional regulation underlying cancer development and drug response.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Estrogen Receptor alpha/metabolism , RNA-Binding Proteins/metabolism , Animals , Base Sequence , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/chemistry , Eukaryotic Initiation Factor-4G/genetics , Eukaryotic Initiation Factor-4G/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Genomics , Humans , Mice, Inbred NOD , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Oncogenes , Protein Binding/drug effects , Protein Domains , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Physiological/drug effects , Stress, Physiological/genetics , Tamoxifen/pharmacology , X-Box Binding Protein 1/metabolism
2.
Cell ; 179(1): 132-146.e14, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31522887

ABSTRACT

Oligodendrocytes extend elaborate microtubule arbors that contact up to 50 axon segments per cell, then spiral around myelin sheaths, penetrating from outer to inner layers. However, how they establish this complex cytoarchitecture is unclear. Here, we show that oligodendrocytes contain Golgi outposts, an organelle that can function as an acentrosomal microtubule-organizing center (MTOC). We identify a specific marker for Golgi outposts-TPPP (tubulin polymerization promoting protein)-that we use to purify this organelle and characterize its proteome. In in vitro cell-free assays, recombinant TPPP nucleates microtubules. Primary oligodendrocytes from Tppp knockout (KO) mice have aberrant microtubule branching, mixed microtubule polarity, and shorter myelin sheaths when cultured on 3-dimensional (3D) microfibers. Tppp KO mice exhibit hypomyelination with shorter, thinner myelin sheaths and motor coordination deficits. Together, our data demonstrate that microtubule nucleation outside the cell body at Golgi outposts by TPPP is critical for elongation of the myelin sheath.


Subject(s)
Carrier Proteins/metabolism , Golgi Apparatus/metabolism , Microtubules/metabolism , Myelin Sheath/metabolism , Nerve Tissue Proteins/metabolism , Animals , Animals, Newborn , Axons/metabolism , Carrier Proteins/genetics , Cell-Free System/metabolism , Cells, Cultured , Escherichia coli/metabolism , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Organizing Center/metabolism , Nerve Tissue Proteins/genetics , Oligodendrocyte Precursor Cells/metabolism , Rats , Rats, Sprague-Dawley , Tubulin/metabolism
3.
Nat Immunol ; 16(6): 618-27, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25939025

ABSTRACT

A20 is an anti-inflammatory protein linked to multiple human diseases; however, the mechanisms by which A20 prevents inflammatory disease are incompletely defined. We found that A20-deficient T cells and fibroblasts were susceptible to caspase-independent and kinase RIPK3-dependent necroptosis. Global deficiency in RIPK3 significantly restored the survival of A20-deficient mice. A20-deficient cells exhibited exaggerated formation of RIPK1-RIPK3 complexes. RIPK3 underwent physiological ubiquitination at Lys5 (K5), and this ubiquitination event supported the formation of RIPK1-RIPK3 complexes. Both the ubiquitination of RIPK3 and formation of the RIPK1-RIPK3 complex required the catalytic cysteine of A20's deubiquitinating motif. Our studies link A20 and the ubiquitination of RIPK3 to necroptotic cell death and suggest additional mechanisms by which A20 might prevent inflammatory disease.


Subject(s)
Cysteine Endopeptidases/metabolism , Fibroblasts/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/physiology , Animals , Apoptosis/genetics , Catalytic Domain/genetics , Cysteine Endopeptidases/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Jurkat Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Necrosis/genetics , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitination/genetics , Ubiquitins/metabolism
4.
EMBO J ; 40(20): e107158, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34515347

ABSTRACT

Nucleolin is a multifunctional RNA Binding Protein (RBP) with diverse subcellular localizations, including the nucleolus in all eukaryotic cells, the plasma membrane in tumor cells, and the axon in neurons. Here we show that the glycine arginine rich (GAR) domain of nucleolin drives subcellular localization via protein-protein interactions with a kinesin light chain. In addition, GAR sequences mediate plasma membrane interactions of nucleolin. Both these modalities are in addition to the already reported involvement of the GAR domain in liquid-liquid phase separation in the nucleolus. Nucleolin transport to axons requires the GAR domain, and heterozygous GAR deletion mice reveal reduced axonal localization of nucleolin cargo mRNAs and enhanced sensory neuron growth. Thus, the GAR domain governs axonal transport of a growth controlling RNA-RBP complex in neurons, and is a versatile localization determinant for different subcellular compartments. Localization determination by GAR domains may explain why GAR mutants in diverse RBPs are associated with neurodegenerative disease.


Subject(s)
Cell Nucleolus/metabolism , Ganglia, Spinal/metabolism , Kinesins/metabolism , Neurons/metabolism , Phosphoproteins/chemistry , RNA-Binding Proteins/chemistry , Sciatic Nerve/metabolism , Amino Acid Sequence , Animals , Axonal Transport/genetics , Cell Line, Tumor , Cell Nucleolus/ultrastructure , Ganglia, Spinal/cytology , Gene Expression , HEK293 Cells , HeLa Cells , Humans , Kinesins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Neurons/cytology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Primary Cell Culture , Protein Domains , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sciatic Nerve/cytology , Nucleolin
5.
Mol Cell Proteomics ; 22(3): 100497, 2023 03.
Article in English | MEDLINE | ID: mdl-36642223

ABSTRACT

New protein synthesis is regulated both at the level of mRNA transcription and translation. RNA-Seq is effective at measuring levels of mRNA expression, but techniques to monitor mRNA translation are much more limited. Previously, we reported results from O-propargyl-puromycin (OPP) labeling of proteins undergoing active translation in a 2-h time frame, followed by biotinylation using click chemistry, affinity purification, and on-bead digestion to identify nascent proteins by mass spectrometry (OPP-ID). As with any on-bead digestion protocol, the problem of nonspecific binders complicated the rigorous categorization of nascent proteins by OPP-ID. Here, we incorporate a chemically cleavable linker, Dde biotin-azide, into the protocol (OPP-IDCL) to provide specific release of modified proteins from the streptavidin beads. Following capture, the Dde moiety is readily cleaved with 2% hydrazine, releasing nascent polypeptides bearing OPP plus a residual C3H8N4 tag. When results are compared side by side with the original OPP-ID method, change to a cleavable linker led to a dramatic reduction in the number of background proteins detected in controls and a concomitant increase in the number of proteins that could be characterized as newly synthesized. We evaluated the method's ability to detect nascent proteins at various submilligram protein input levels and showed that, when starting with only 100 µg of protein, ∼1500 nascent proteins could be identified with low background. Upon treatment of K562 cells with MLN128, a potent inhibitor of the mammalian target of rapamycin, prior to OPP treatment, we identified 1915 nascent proteins, the majority of which were downregulated upon inhibitor treatment. Repressed proteins with log2 FC <-1 revealed a complex network of functionally interacting proteins, with the largest cluster associated with translational initiation. Overall, incorporation of the Dde biotin-azide cleavable linker into our protocol has increased the depth and accuracy of profiling of nascent protein networks.


Subject(s)
Azides , Biotin , Proteins/chemistry , Peptides , RNA, Messenger
6.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35074918

ABSTRACT

MeCP2 is associated with Rett syndrome (RTT), MECP2 duplication syndrome, and a number of conditions with isolated features of these diseases, including autism, intellectual disability, and motor dysfunction. MeCP2 is known to broadly bind methylated DNA, but the precise molecular mechanism driving disease pathogenesis remains to be determined. Using proximity-dependent biotinylation (BioID), we identified a transcription factor 20 (TCF20) complex that interacts with MeCP2 at the chromatin interface. Importantly, RTT-causing mutations in MECP2 disrupt this interaction. TCF20 and MeCP2 are highly coexpressed in neurons and coregulate the expression of key neuronal genes. Reducing Tcf20 partially rescued the behavioral deficits caused by MECP2 overexpression, demonstrating a functional relationship between MeCP2 and TCF20 in MECP2 duplication syndrome pathogenesis. We identified a patient exhibiting RTT-like neurological features with a missense mutation in the PHF14 subunit of the TCF20 complex that abolishes the MeCP2-PHF14-TCF20 interaction. Our data demonstrate the critical role of the MeCP2-TCF20 complex for brain function.


Subject(s)
Methyl-CpG-Binding Protein 2/metabolism , Multiprotein Complexes/metabolism , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/metabolism , Transcription Factors/metabolism , Alleles , Animals , Biomarkers , Brain/metabolism , Disease Models, Animal , Disease Susceptibility , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Mutation , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Synapses/metabolism , Transcription Factors/genetics
7.
Immunity ; 42(1): 55-67, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25607459

ABSTRACT

Inappropriate inflammasome activation contributes to multiple human diseases, but the mechanisms by which inflammasomes are suppressed are poorly understood. The NF-κB inhibitor A20 is a ubiquitin-modifying enzyme that might be critical in preventing human inflammatory diseases. Here, we report that A20-deficient macrophages, unlike normal cells, exhibit spontaneous NLRP3 inflammasome activity to LPS alone. The kinase RIPK3, but not the adaptor MyD88, is required for this response. In normal cells, A20 constitutively associates with caspase-1 and pro-IL-1ß, and NLRP3 activation further promotes A20 recruitment to the inflammasome. Pro-IL-1ß also co-immunoprecipitates with RIPK1, RIPK3, caspase-1, and caspase-8 in a complex that is modified with K63-linked and unanchored polyubiquitin. In A20-deficient macrophages, this pro-IL-1ß-associated ubiquitination is markedly increased in a RIPK3-dependent manner. Mass spectrometric and mutational analyses reveal that K133 of pro-IL-1ß is a physiological ubiquitination site that supports processing. Our study reveals a mechanism by which A20 prevents inflammatory diseases.


Subject(s)
Carrier Proteins/metabolism , Cysteine Endopeptidases/metabolism , Inflammasomes/immunology , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/physiology , Animals , Cell Line , Cysteine Endopeptidases/genetics , DNA Mutational Analysis , Immune Tolerance , Interleukin-1beta/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred Strains , Mice, Knockout , Multiprotein Complexes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitination/genetics
8.
Mol Cell Proteomics ; 21(11): 100418, 2022 11.
Article in English | MEDLINE | ID: mdl-36180036

ABSTRACT

Importin ß1 (KPNB1) is a nucleocytoplasmic transport factor with critical roles in both cytoplasmic and nucleocytoplasmic transport, hence there is keen interest in the characterization of its subcellular interactomes. We found limited efficiency of BioID in the detection of importin complex cargos and therefore generated a highly specific and sensitive anti-KPNB1 monoclonal antibody to enable biotinylation by antibody recognition analysis of importin ß1 interactomes. The monoclonal antibody recognizes an epitope comprising residues 301-320 of human KPBN1 and strikingly is highly specific for cytoplasmic KPNB1 in diverse applications, with little reaction with KPNB1 in the nucleus. Biotinylation by antibody recognition with this novel antibody revealed numerous new interactors of importin ß1, expanding the KPNB1 interactome to cytoplasmic and signaling complexes that highlight potential new functions for the importins complex beyond nucleocytoplasmic transport. Data are available via ProteomeXchange with identifier PXD032728.


Subject(s)
Antibodies, Monoclonal , Karyopherins , Humans , Karyopherins/metabolism , Antibodies, Monoclonal/metabolism , beta Karyopherins/metabolism , Active Transport, Cell Nucleus , Cytoplasm/metabolism , Cell Nucleus/metabolism
9.
J Neurosci ; 42(10): 2065-2079, 2022 03 09.
Article in English | MEDLINE | ID: mdl-34987108

ABSTRACT

Ferroptosis is a caspase-independent, iron-dependent form of regulated necrosis extant in traumatic brain injury, Huntington disease, and hemorrhagic stroke. It can be activated by cystine deprivation leading to glutathione depletion, the insufficiency of the antioxidant glutathione peroxidase-4, and the hemolysis products hemoglobin and hemin. A cardinal feature of ferroptosis is extracellular signal-regulated kinase (ERK)1/2 activation culminating in its translocation to the nucleus. We have previously confirmed that the mitogen-activated protein (MAP) kinase kinase (MEK) inhibitor U0126 inhibits persistent ERK1/2 phosphorylation and ferroptosis. Here, we show that hemin exposure, a model of secondary injury in brain hemorrhage and ferroptosis, activated ERK1/2 in mouse neurons. Accordingly, MEK inhibitor U0126 protected against hemin-induced ferroptosis. Unexpectedly, U0126 prevented hemin-induced ferroptosis independent of its ability to inhibit ERK1/2 signaling. In contrast to classical ferroptosis in neurons or cancer cells, chemically diverse inhibitors of MEK did not block hemin-induced ferroptosis, nor did the forced expression of the ERK-selective MAP kinase phosphatase (MKP)3. We conclude that hemin or hemoglobin-induced ferroptosis, unlike glutathione depletion, is ERK1/2-independent. Together with recent studies, our findings suggest the existence of a novel subtype of neuronal ferroptosis relevant to bleeding in the brain that is 5-lipoxygenase-dependent, ERK-independent, and transcription-independent. Remarkably, our unbiased phosphoproteome analysis revealed dramatic differences in phosphorylation induced by two ferroptosis subtypes. As U0126 also reduced cell death and improved functional recovery after hemorrhagic stroke in male mice, our analysis also provides a template on which to build a search for U0126's effects in a variant of neuronal ferroptosis.SIGNIFICANCE STATEMENT Ferroptosis is an iron-dependent mechanism of regulated necrosis that has been linked to hemorrhagic stroke. Common features of ferroptotic death induced by diverse stimuli are the depletion of the antioxidant glutathione, production of lipoxygenase-dependent reactive lipids, sensitivity to iron chelation, and persistent activation of extracellular signal-regulated kinase (ERK) signaling. Unlike classical ferroptosis induced in neurons or cancer cells, here we show that ferroptosis induced by hemin is ERK-independent. Paradoxically, the canonical MAP kinase kinase (MEK) inhibitor U0126 blocks brain hemorrhage-induced death. Altogether, these data suggest that a variant of ferroptosis is unleashed in hemorrhagic stroke. We present the first, unbiased phosphoproteomic analysis of ferroptosis as a template on which to understand distinct paths to cell death that meet the definition of ferroptosis.


Subject(s)
Ferroptosis , Hemorrhagic Stroke , Animals , Antioxidants/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutathione/metabolism , Hemin/metabolism , Hemin/pharmacology , Hemoglobins/metabolism , Intracranial Hemorrhages/metabolism , Iron/metabolism , Male , Mice , Mitogen-Activated Protein Kinase Kinases/metabolism , Necrosis/metabolism , Neurons/metabolism , Phosphorylation
11.
Hum Mol Genet ; 28(9): 1498-1514, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30590647

ABSTRACT

Mutations in the microtubule-associated protein tau (MAPT) underlie multiple neurodegenerative disorders, yet the pathophysiological mechanisms are unclear. A novel variant in MAPT resulting in an alanine to threonine substitution at position 152 (A152T tau) has recently been described as a significant risk factor for both frontotemporal lobar degeneration and Alzheimer's disease. Here we use complementary computational, biochemical, molecular, genetic and imaging approaches in Caenorhabditis elegans and mouse models to interrogate the effects of the A152T variant on tau function. In silico analysis suggests that a threonine at position 152 of tau confers a new phosphorylation site. This finding is borne out by mass spectrometric survey of A152T tau phosphorylation in C. elegans and mouse. Optical pulse-chase experiments of Dendra2-tau demonstrate that A152T tau and phosphomimetic A152E tau exhibit increased diffusion kinetics and the ability to traverse across the axon initial segment more efficiently than wild-type (WT) tau. A C. elegans model of tauopathy reveals that A152T and A152E tau confer patterns of developmental toxicity distinct from WT tau, likely due to differential effects on retrograde axonal transport. These data support a role for phosphorylation of the variant threonine in A152T tau toxicity and suggest a mechanism involving impaired retrograde axonal transport contributing to human neurodegenerative disease.


Subject(s)
Alleles , Amino Acid Substitution , Genetic Variation , tau Proteins/genetics , tau Proteins/metabolism , Animals , Animals, Genetically Modified , Axonal Transport , Axons/metabolism , Caenorhabditis elegans , Disease Models, Animal , Disease Susceptibility , Humans , Mice , Mutation , Phosphorylation , Protein Binding , Synaptic Vesicles/metabolism , Tauopathies/etiology , Tauopathies/metabolism , Tauopathies/pathology
12.
J Cell Sci ; 132(7)2019 04 09.
Article in English | MEDLINE | ID: mdl-30837283

ABSTRACT

mTORC2 lies at the intersection of signaling pathways that control metabolism and ion transport through phosphorylation of the AGC-family kinases, the Akt and SGK1 proteins. How mTORC2 targets these functionally distinct downstream effectors in a context-specific manner is not known. Here, we show that the salt- and blood pressure-regulatory hormone, angiotensin II (AngII) stimulates selective mTORC2-dependent phosphorylation of SGK1 (S422) but not Akt (S473 and equivalent sites). Conventional PKC (cPKC), a critical mediator of the angiotensin type I receptor (AT1R, also known as AGTR1) signaling, regulates the subcellular localization of SIN1 (also known as MAPKAP1) and SGK1. Inhibition of cPKC catalytic activity disturbs SIN1 and SGK1 subcellular localization, re-localizing them from the nucleus and a perinuclear compartment to the plasma membrane in advance of hormonal stimulation. Surprisingly, pre-targeting of SIN1 and SGK1 to the plasma membrane prevents SGK1 S422 but not Akt S473 phosphorylation. Additionally, we identify three sites on SIN1 (S128, S315 and S356) that are phosphorylated in response to cPKC activation. Collectively, these data demonstrate that SGK1 activation occurs at a distinct subcellular compartment from that of Akt and suggests a mechanism for the selective activation of these functionally distinct mTORC2 targets through subcellular partitioning of mTORC2 activity.


Subject(s)
Immediate-Early Proteins/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , HEK293 Cells , Humans , Immediate-Early Proteins/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
13.
Mol Cell ; 52(5): 679-92, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24211266

ABSTRACT

SHOC2 is mutated in Noonan syndrome and plays a key role in the activation of the ERK-MAPK pathway, which is upregulated in the majority of human cancers. SHOC2 functions as a PP1-regulatory protein and as an effector of MRAS. Here we show that SHOC2 and MRAS form a complex with SCRIB, a polarity protein with tumor suppressor properties. SCRIB functions as a PP1-regulatory protein and antagonizes SHOC2-mediated RAF dephosphorylation through a mechanism involving competition for PP1 molecules within the same macromolecular complex. SHOC2 function is selectively required for the malignant properties of tumor cells with mutant RAS, and both MRAS and SHOC2 play a key role in polarized migration. We propose that MRAS, through its ability to recruit a complex with paradoxical components, coordinates ERK pathway spatiotemporal dynamics with polarity and that this complex plays a key role during tumorigenic growth.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/genetics , Membrane Proteins/genetics , Tumor Suppressor Proteins/genetics , ras Proteins/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line , Cell Movement/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Macromolecular Substances/metabolism , Membrane Proteins/metabolism , Phosphorylation , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Tumor Suppressor Proteins/metabolism , raf Kinases/genetics , raf Kinases/metabolism , ras Proteins/metabolism
14.
Proc Natl Acad Sci U S A ; 115(10): 2353-2358, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29467287

ABSTRACT

Regulation of gene expression at the level of protein synthesis is a crucial element in driving how the genetic landscape is expressed. However, we are still limited in technologies that can quantitatively capture the immediate proteomic changes that allow cells to respond to specific stimuli. Here, we present a method to capture and identify nascent proteomes in situ across different cell types without disturbing normal growth conditions, using O-propargyl-puromycin (OPP). Cell-permeable OPP rapidly labels nascent elongating polypeptides, which are subsequently conjugated to biotin-azide, using click chemistry, and captured with streptavidin beads, followed by digestion and analysis, using liquid chromatography-tandem mass spectrometry. Our technique of OPP-mediated identification (OPP-ID) allows detection of widespread proteomic changes within a short 2-hour pulse of OPP. We illustrate our technique by recapitulating alterations of proteomic networks induced by a potent mammalian target of rapamycin inhibitor, MLN128. In addition, by employing OPP-ID, we identify more than 2,100 proteins and uncover distinct protein networks underlying early erythroid progenitor and differentiation states not amenable to alternative approaches such as amino acid analog labeling. We present OPP-ID as a method to quantitatively identify nascent proteomes across an array of biological contexts while preserving the subtleties directing signaling in the native cellular environment.


Subject(s)
Cell Differentiation/physiology , Proteome/analysis , Proteomics/methods , Signal Transduction/physiology , Chromatography, Liquid , Drug Discovery , Humans , K562 Cells , Protein Biosynthesis , Proteome/chemistry , Proteome/metabolism , Puromycin/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tandem Mass Spectrometry
15.
J Am Chem Soc ; 142(45): 19085-19093, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33124817

ABSTRACT

Ferroptosis is an iron-dependent form of cell death resulting from loss or inhibition of cellular machinery that protects from the accumulation of lipid hydroperoxides. Ferroptosis likely serves a tumor suppressing function in normal cellular homeostasis, but certain cancers exploit and become highly dependent on specific nodes of the pathway, presumably to survive under conditions of increased oxidative stress and elevated labile ferrous iron levels. Here we introduce Ferroptosis Inducing Peroxide for Chemoproteomics-1 (FIPC-1), a reactivity-based probe that couples Fenton-type reaction with ferrous iron to subsequent protein labeling via concomitant carbon-centered radical generation. We show that FIPC-1 induces ferroptosis in susceptible cell types and labels cellular proteins in an iron-dependent fashion. Use of FIPC-1 in a quantitative chemoproteomics workflow reproducibly enriched protein targets in the thioredoxin, oxidoreductase, and protein disulfide isomerase (PDI) families, among others. In further interrogating the saturable targets of FIPC-1, we identified the PDI family member P4HB and the functionally uncharacterized protein NT5DC2, a member of the haloacid dehalogenase (HAD) superfamily, as previously unrecognized modulators of ferroptosis. Knockdown of these target genes sensitized cells to known ferroptosis inducers, while PACMA31, a previously reported inhibitor of P4HB, directly induced ferroptosis and was highly synergistic with erastin. Overall, this study introduces a new reactivity-based probe of the ferrous iron-dependent interactome and uncovers new targets for the therapeutic modulation of ferroptosis.


Subject(s)
Ferrous Compounds/chemistry , Molecular Probes/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Ferroptosis/drug effects , Ferrous Compounds/metabolism , Humans , Hydrogen Peroxide/chemistry , Iron/chemistry , Molecular Probes/chemical synthesis , Molecular Probes/pharmacology , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Peroxides/chemistry , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/metabolism , Thioredoxins/chemistry , Thioredoxins/metabolism
16.
Mol Cell Proteomics ; 17(11): 2091-2106, 2018 11.
Article in English | MEDLINE | ID: mdl-30038033

ABSTRACT

mRNA translation in axons enables neurons to introduce new proteins at sites distant from their cell body. mRNA-protein interactions drive this post-transcriptional regulation, yet knowledge of RNA binding proteins (RBP) in axons is limited. Here we used proteomics to identify RBPs interacting with the axonal localizing motifs of Nrn1, Hmgb1, Actb, and Gap43 mRNAs, revealing many novel RBPs in axons. Interestingly, no RBP is shared between all four RNA motifs, suggesting graded and overlapping specificities of RBP-mRNA pairings. A systematic assessment of axonal mRNAs interacting with hnRNP H1, hnRNP F, and hnRNP K, proteins that bound with high specificity to Nrn1 and Hmgb1, revealed that axonal mRNAs segregate into axon growth-associated RNA regulons based on hnRNP interactions. Axotomy increases axonal transport of hnRNPs H1, F, and K, depletion of these hnRNPs decreases axon growth and reduces axonal mRNA levels and axonal protein synthesis. Thus, subcellular hnRNP-interacting RNA regulons support neuronal growth and regeneration.


Subject(s)
Axons/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Nucleotide Motifs/genetics , RNA, Messenger/genetics , Regulon/genetics , 5' Untranslated Regions/genetics , Animals , Axonal Transport/genetics , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Male , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Binding , Protein Biosynthesis , RNA Transport/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
17.
Genes Dev ; 26(13): 1421-6, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22751498

ABSTRACT

The Ras/mitogen-activated protein kinase (MAPK) pathway plays a critical role in transducing mitogenic signals from receptor tyrosine kinases. Loss-of-function mutations in one feedback regulator of Ras/MAPK signaling, SPRED1 (Sprouty-related protein with an EVH1 domain), cause Legius syndrome, an autosomal dominant human disorder that resembles Neurofibromatosis-1 (NF1). Spred1 functions as a negative regulator of the Ras/MAPK pathway; however, the underlying molecular mechanism is poorly understood. Here we show that neurofibromin, the NF1 gene product, is a Spred1-interacting protein that is necessary for Spred1's inhibitory function. We show that Spred1 binding induces the plasma membrane localization of NF1, which subsequently down-regulates Ras-GTP levels. This novel mechanism for the regulation of neurofibromin provides a molecular bridge for understanding the overlapping pathophysiology of NF1 and Legius syndrome.


Subject(s)
Cafe-au-Lait Spots/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neurofibromatosis 1/metabolism , Neurofibromin 1/metabolism , Repressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cafe-au-Lait Spots/genetics , Cells, Cultured , Humans , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System , Membrane Proteins/genetics , Mice , Neurofibromatosis 1/genetics , Neurofibromin 1/genetics , Protein Binding , Repressor Proteins/genetics
18.
Mol Cell Proteomics ; 16(1): 39-56, 2017 01.
Article in English | MEDLINE | ID: mdl-27834733

ABSTRACT

Glioblastoma multiformes (GBMs) are high-grade astrocytomas and the most common brain malignancies. Primary GBMs are often associated with disturbed RAS signaling, and expression of oncogenic HRAS results in a malignant phenotype in glioma cell lines. Secondary GBMs arise from lower-grade astrocytomas, have slower progression than primary tumors, and contain IDH1 mutations in over 70% of cases. Despite significant amount of accumulating genomic and transcriptomic data, the fundamental mechanistic differences of gliomagenesis in these two types of high-grade astrocytoma remain poorly understood. Only a few studies have attempted to investigate the proteome, phosphorylation signaling, and epigenetic regulation in astrocytoma. In the present study, we applied quantitative phosphoproteomics to identify the main signaling differences between oncogenic HRAS and mutant IDH1-driven glioma cells as models of primary and secondary GBM, respectively. Our analysis confirms the driving roles of the MAPK and PI3K/mTOR signaling pathways in HRAS driven cells and additionally uncovers dysregulation of other signaling pathways. Although a subset of the signaling changes mediated by HRAS could be reversed by a MEK inhibitor, dual inhibition of MEK and PI3K resulted in more complete reversal of the phosphorylation patterns produced by HRAS expression. In contrast, cells expressing mutant IDH1 did not show significant activation of MAPK or PI3K/mTOR pathways. Instead, global downregulation of protein expression was observed. Targeted proteomic analysis of histone modifications identified significant histone methylation, acetylation, and butyrylation changes in the mutant IDH1 expressing cells, consistent with a global transcriptional repressive state. Our findings offer novel mechanistic insight linking mutant IDH1 associated inhibition of histone demethylases with specific histone modification changes to produce global transcriptional repression in secondary glioblastoma. Our proteomic datasets are available for download and provide a comprehensive catalogue of alterations in protein abundance, phosphorylation, and histone modifications in oncogenic HRAS and IDH1 driven astrocytoma cells beyond the transcriptomic level.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Isocitrate Dehydrogenase/genetics , Phosphoproteins/analysis , Proteomics/methods , Proto-Oncogene Proteins p21(ras)/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/metabolism , Histone Code , Histones/metabolism , Humans , Mitogen-Activated Protein Kinases/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Protein Interaction Maps , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
19.
Am J Respir Cell Mol Biol ; 55(5): 623-632, 2016 11.
Article in English | MEDLINE | ID: mdl-27281349

ABSTRACT

Lung inflammation in premature infants contributes to the development of bronchopulmonary dysplasia (BPD), a chronic lung disease with long-term sequelae. Pilot studies administering budesonide suspended in surfactant have found reduced BPD without the apparent adverse effects that occur with systemic dexamethasone therapy. Our objective was to determine budesonide potency, stability, and antiinflammatory effects in human fetal lung. We cultured explants of second-trimester fetal lung with budesonide or dexamethasone and used microscopy, immunoassays, RNA sequencing, liquid chromatography/tandem mass spectrometry, and pulsating bubble surfactometry. Budesonide suppressed secreted chemokines IL-8 and CCL2 (MCP-1) within 4 hours, reaching a 90% decrease at 12 hours, which was fully reversed 72 hours after removal of the steroid. Half-maximal effects occurred at 0.04-0.05 nM, representing a fivefold greater potency than for dexamethasone. Budesonide significantly induced 3.6% and repressed 2.8% of 14,500 sequenced mRNAs by 1.6- to 95-fold, including 119 genes that contribute to the glucocorticoid inflammatory transcriptome; some are known targets of nuclear factor-κB. By global proteomics, 22 secreted inflammatory proteins were hormonally regulated. Two glucocorticoid-regulated genes of interest because of their association with lung disease are CHI3L1 and IL1RL1. Budesonide retained activity in the presence of surfactant and did not alter its surface properties. There was some formation of palmitate-budesonide in lung tissue but no detectable metabolism to inactive 16α-hydroxy prednisolone. We concluded that budesonide is a potent and stable antiinflammatory glucocorticoid in human fetal lung in vitro, supporting a beneficial antiinflammatory response to lung-targeted budesonide:surfactant treatment of infants for the prevention of BPD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Budesonide/pharmacology , Fetus/drug effects , Lung/embryology , Anti-Inflammatory Agents/metabolism , Budesonide/metabolism , Chemokines/metabolism , Dexamethasone/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Humans , Lung/drug effects , Pneumonia/genetics , Pneumonia/pathology , Surface Tension/drug effects , Time Factors
20.
Mol Cell Proteomics ; 12(12): 3653-65, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24019147

ABSTRACT

Brassinosteroids (BRs) are essential phytohormones for plant growth and development. BRs are perceived by the cell surface receptor kinase BRI1, and downstream signal transduction through multiple components leads to activation of the transcription factors BZR1 and BZR2/BES1. BZR1 activity is highly controlled by BR through reversible phosphorylation, protein degradation, and nucleocytoplasmic shuttling. To further understand the molecular function of BZR1, we performed tandem affinity purification of the BZR1 complex and identified BZR1-associated proteins using mass spectrometry. These BZR1-associated proteins included several known BR signaling components, such as BIN2, BSK1, 14-3-3λ, and PP2A, as well as a large number of proteins with previously unknown functions in BR signal transduction, including the kinases MKK5 and MAPK4, histone deacetylase 19, cysteine proteinase inhibitor 6, a DEAD-box RNA helicase, cysteine endopeptidases RD21A and RD21B, calmodulin-binding transcription activator 5, ubiquitin protease 12, cyclophilin 59, and phospholipid-binding protein synaptotagmin A. Their interactions with BZR1 were confirmed by in vivo and in vitro assays. Furthermore, MKK5 was found to phosphorylate BZR1 in vitro. This study demonstrates an effective method for purifying proteins associated with low-abundance transcription factors, and identifies new BZR1-interacting proteins with potentially important roles in BR response.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/drug effects , Brassinosteroids/pharmacology , Gene Expression Regulation, Plant , Nuclear Proteins/metabolism , Plant Growth Regulators/pharmacology , Signal Transduction , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Chromatography, Affinity , DNA-Binding Proteins , Mass Spectrometry , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Molecular Sequence Annotation , Nuclear Proteins/genetics , Phosphorylation , Protein Binding , Protein Interaction Mapping , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Transport , Proteolysis
SELECTION OF CITATIONS
SEARCH DETAIL