Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 223(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38578285

ABSTRACT

IRE1α is an endoplasmic reticulum (ER) sensor that recognizes misfolded proteins to induce the unfolded protein response (UPR). We studied cholera toxin (CTx), which invades the ER and activates IRE1α in host cells, to understand how unfolded proteins are recognized. Proximity labeling colocalized the enzymatic and metastable A1 segment of CTx (CTxA1) with IRE1α in live cells, where we also found that CTx-induced IRE1α activation enhanced toxicity. In vitro, CTxA1 bound the IRE1α lumenal domain (IRE1αLD), but global unfolding was not required. Rather, the IRE1αLD recognized a seven-residue motif within an edge ß-strand of CTxA1 that must locally unfold for binding. Binding mapped to a pocket on IRE1αLD normally occupied by a segment of the IRE1α C-terminal flexible loop implicated in IRE1α oligomerization. Mutation of the CTxA1 recognition motif blocked CTx-induced IRE1α activation in live cells, thus linking the binding event with IRE1α signal transduction and induction of the UPR.


Subject(s)
Cholera Toxin , Endoribonucleases , Protein Serine-Threonine Kinases , Unfolded Protein Response , Cholera Toxin/genetics , Cholera Toxin/metabolism , Endoplasmic Reticulum Stress , Endoribonucleases/genetics , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Humans , Animals , Mice , Cell Line
2.
EMBO Rep ; 24(12): e56997, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-37975164

ABSTRACT

Planar cell polarity (PCP) signaling polarizes epithelial cells within the plane of an epithelium. Core PCP signaling components adopt asymmetric subcellular localizations within cells to both polarize and coordinate polarity between cells. Achieving subcellular asymmetry requires additional effectors, including some mediating post-translational modifications of core components. Identification of such proteins is challenging due to pleiotropy. We used mass spectrometry-based proximity labeling proteomics to identify such regulators in the Drosophila wing. We identified the catalytic subunit of protein phosphatase1, Pp1-87B, and show that it regulates core protein polarization. Pp1-87B interacts with the core protein Van Gogh and at least one serine/threonine kinase, Dco/CKIε, that is known to regulate PCP. Pp1-87B modulates Van Gogh subcellular localization and directs its dephosphorylation in vivo. PNUTS, a Pp1 regulatory subunit, also modulates PCP. While the direct substrate(s) of Pp1-87B in control of PCP is not known, our data support the model that cycling between phosphorylated and unphosphorylated forms of one or more core PCP components may regulate acquisition of asymmetry. Finally, our screen serves as a resource for identifying additional regulators of PCP signaling.


Subject(s)
Drosophila Proteins , Membrane Proteins , Animals , Cell Polarity/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
3.
bioRxiv ; 2023 Sep 13.
Article in English | MEDLINE | ID: mdl-37745534

ABSTRACT

PCP signaling polarizes epithelial cells within the plane of an epithelium. Core PCP signaling components adopt asymmetric subcellular localizations within cells to both polarize and coordinate polarity between cells. Achieving subcellular asymmetry requires additional effectors, including some mediating post-translational modifications of core components. Identification of such proteins is challenging due to pleiotropy. We used mass spectrometry-based proximity labeling proteomics to identify such regulators in the Drosophila wing. We identified the catalytic subunit of Protein Phosphatase1, Pp1-87B, and show that it regulates core protein polarization. Pp1-87B interacts with the core protein Van Gogh and at least one Serine/Threonine kinase, Dco/CKIε, that is known to regulate PCP. Pp1-87B modulates Van Gogh subcellular localization and directs its dephosphorylation in vivo. PNUTS, a Pp1 regulatory subunit, also modulates PCP. While the direct substrate(s) of Pp1-87B in control of PCP is not known, our data support the model that cycling between phosphorylated and unphosphorylated forms of one or more core PCP components may regulate acquisition of asymmetry. Finally, our screen serves as a resource for identifying additional regulators of PCP signaling.

4.
Neuron ; 110(14): 2299-2314.e8, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35613619

ABSTRACT

Transcription factors specify the fate and connectivity of developing neurons. We investigate how a lineage-specific transcription factor, Acj6, controls the precise dendrite targeting of Drosophila olfactory projection neurons (PNs) by regulating the expression of cell-surface proteins. Quantitative cell-surface proteomic profiling of wild-type and acj6 mutant PNs in intact developing brains, and a proteome-informed genetic screen identified PN surface proteins that execute Acj6-regulated wiring decisions. These include canonical cell adhesion molecules and proteins previously not associated with wiring, such as Piezo, whose mechanosensitive ion channel activity is dispensable for its function in PN dendrite targeting. Comprehensive genetic analyses revealed that Acj6 employs unique sets of cell-surface proteins in different PN types for dendrite targeting. Combined expression of Acj6 wiring executors rescued acj6 mutant phenotypes with higher efficacy and breadth than expression of individual executors. Thus, Acj6 controls wiring specificity of different neuron types by specifying distinct combinatorial expression of cell-surface executors.


Subject(s)
Drosophila Proteins , Olfactory Receptor Neurons , Animals , Dendrites/physiology , Drosophila/metabolism , Drosophila Proteins/metabolism , Ion Channels/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Olfactory Pathways/physiology , Olfactory Receptor Neurons/metabolism , POU Domain Factors/metabolism , Proteomics , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Nat Commun ; 12(1): 2382, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33888706

ABSTRACT

Conventional approaches to identify secreted factors that regulate homeostasis are limited in their abilities to identify the tissues/cells of origin and destination. We established a platform to identify secreted protein trafficking between organs using an engineered biotin ligase (BirA*G3) that biotinylates, promiscuously, proteins in a subcellular compartment of one tissue. Subsequently, biotinylated proteins are affinity-enriched and identified from distal organs using quantitative mass spectrometry. Applying this approach in Drosophila, we identify 51 muscle-secreted proteins from heads and 269 fat body-secreted proteins from legs/muscles, including CG2145 (human ortholog ENDOU) that binds directly to muscles and promotes activity. In addition, in mice, we identify 291 serum proteins secreted from conditional BirA*G3 embryo stem cell-derived teratomas, including low-abundance proteins with hormonal properties. Our findings indicate that the communication network of secreted proteins is vast. This approach has broad potential across different model systems to identify cell-specific secretomes and mediators of interorgan communication in health or disease.


Subject(s)
Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/metabolism , Proteomics/methods , Repressor Proteins/metabolism , Staining and Labeling/methods , Animals , Animals, Genetically Modified , Biotin/metabolism , Biotinylation , Carbon-Nitrogen Ligases/genetics , Cell Line , Disease Models, Animal , Drosophila , Embryonic Stem Cells , Escherichia coli Proteins/genetics , Female , Humans , Male , Mice , Protein Engineering , Protein Transport , Repressor Proteins/genetics , Tandem Mass Spectrometry/methods , Teratoma/diagnosis , Teratoma/pathology
6.
Nat Commun ; 12(1): 734, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33531470

ABSTRACT

Driver genes with a mutually exclusive mutation pattern across tumor genomes are thought to have overlapping roles in tumorigenesis. In contrast, we show here that mutually exclusive prostate cancer driver alterations involving the ERG transcription factor and the ubiquitin ligase adaptor SPOP are synthetic sick. At the molecular level, the incompatible cancer pathways are driven by opposing functions in SPOP. ERG upregulates wild type SPOP to dampen androgen receptor (AR) signaling and sustain ERG activity through degradation of the bromodomain histone reader ZMYND11. Conversely, SPOP-mutant tumors stabilize ZMYND11 to repress ERG-function and enable oncogenic androgen receptor signaling. This dichotomy regulates the response to therapeutic interventions in the AR pathway. While mutant SPOP renders tumor cells susceptible to androgen deprivation therapies, ERG promotes sensitivity to high-dose androgen therapy and pharmacological inhibition of wild type SPOP. More generally, these results define a distinct class of antagonistic cancer drivers and a blueprint toward their therapeutic exploitation.


Subject(s)
Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Prostatic Neoplasms/metabolism , Repressor Proteins/metabolism , Transcriptional Regulator ERG/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Animals , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Male , Mice , Mice, Nude , Mutation/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Prostatic Neoplasms/genetics , Protein Binding , Proteomics , Receptors, Androgen/metabolism , Repressor Proteins/genetics , Signal Transduction/physiology , Transcriptional Regulator ERG/genetics , Ubiquitin-Protein Ligase Complexes/genetics
7.
Blood ; 137(16): 2209-2220, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33512474

ABSTRACT

Casitas B-lineage lymphoma (CBL) encodes an E3 ubiquitin ligase and signaling adaptor that regulates receptor and nonreceptor tyrosine kinases. Recurrent CBL mutations occur in myeloid neoplasms, including 10% to 20% of chronic myelomonocytic leukemia (CMML) cases, and selectively disrupt the protein's E3 ubiquitin ligase activity. CBL mutations have been associated with poor prognosis, but the oncogenic mechanisms and therapeutic implications of CBL mutations remain incompletely understood. We combined functional assays and global mass spectrometry to define the phosphoproteome, CBL interactome, and mechanism of signaling activation in a panel of cell lines expressing an allelic series of CBL mutations. Our analyses revealed that increased LYN activation and interaction with mutant CBL are key drivers of enhanced CBL phosphorylation, phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1) recruitment, and downstream phosphatidylinositol 3-kinase (PI3K)/AKT signaling in CBL-mutant cells. Signaling adaptor domains of CBL, including the tyrosine kinase-binding domain, proline-rich region, and C-terminal phosphotyrosine sites, were all required for the oncogenic function of CBL mutants. Genetic ablation or dasatinib-mediated inhibition of LYN reduced CBL phosphorylation, CBL-PIK3R1 interaction, and PI3K/AKT signaling. Furthermore, we demonstrated in vitro and in vivo antiproliferative efficacy of dasatinib in CBL-mutant cell lines and primary CMML. Overall, these mechanistic insights into the molecular function of CBL mutations provide rationale to explore the therapeutic potential of LYN inhibition in CBL-mutant myeloid malignancies.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-cbl/genetics , src-Family Kinases/metabolism , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , Mutation , Protein Interaction Maps , Proto-Oncogene Proteins c-cbl/metabolism , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 117(22): 12143-12154, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32424107

ABSTRACT

Proximity labeling catalyzed by promiscuous enzymes, such as TurboID, have enabled the proteomic analysis of subcellular regions difficult or impossible to access by conventional fractionation-based approaches. Yet some cellular regions, such as organelle contact sites, remain out of reach for current PL methods. To address this limitation, we split the enzyme TurboID into two inactive fragments that recombine when driven together by a protein-protein interaction or membrane-membrane apposition. At endoplasmic reticulum-mitochondria contact sites, reconstituted TurboID catalyzed spatially restricted biotinylation, enabling the enrichment and identification of >100 endogenous proteins, including many not previously linked to endoplasmic reticulum-mitochondria contacts. We validated eight candidates by biochemical fractionation and overexpression imaging. Overall, split-TurboID is a versatile tool for conditional and spatially specific proximity labeling in cells.


Subject(s)
Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Proteome/analysis , Biotinylation , HEK293 Cells , Humans , Proteome/metabolism , Staining and Labeling
9.
Cell ; 180(2): 373-386.e15, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31955847

ABSTRACT

Molecular interactions at the cellular interface mediate organized assembly of single cells into tissues and, thus, govern the development and physiology of multicellular organisms. Here, we developed a cell-type-specific, spatiotemporally resolved approach to profile cell-surface proteomes in intact tissues. Quantitative profiling of cell-surface proteomes of Drosophila olfactory projection neurons (PNs) in pupae and adults revealed global downregulation of wiring molecules and upregulation of synaptic molecules in the transition from developing to mature PNs. A proteome-instructed in vivo screen identified 20 cell-surface molecules regulating neural circuit assembly, many of which belong to evolutionarily conserved protein families not previously linked to neural development. Genetic analysis further revealed that the lipoprotein receptor LRP1 cell-autonomously controls PN dendrite targeting, contributing to the formation of a precise olfactory map. These findings highlight the power of temporally resolved in situ cell-surface proteomic profiling in discovering regulators of brain wiring.


Subject(s)
Olfactory Pathways/metabolism , Olfactory Receptor Neurons/metabolism , Proteomics/methods , Animals , Axons/metabolism , Brain/metabolism , Dendrites/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/metabolism , Neurogenesis/physiology , Olfactory Nerve/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/physiology , Receptors, Lipoprotein/metabolism , Smell/physiology
10.
Nat Commun ; 11(1): 359, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953384

ABSTRACT

Protein ubiquitylation is involved in a plethora of cellular processes. While antibodies directed at ubiquitin remnants (K-ɛ-GG) have improved the ability to monitor ubiquitylation using mass spectrometry, methods for highly multiplexed measurement of ubiquitylation in tissues and primary cells using sub-milligram amounts of sample remains a challenge. Here, we present a highly sensitive, rapid and multiplexed protocol termed UbiFast for quantifying ~10,000 ubiquitylation sites from as little as 500 µg peptide per sample from cells or tissue in a TMT10plex in ca. 5 h. High-field Asymmetric Waveform Ion Mobility Spectrometry (FAIMS) is used to improve quantitative accuracy for posttranslational modification analysis. We use the approach to rediscover substrates of the E3 ligase targeting drug lenalidomide and to identify proteins modulated by ubiquitylation in models of basal and luminal human breast cancer. The sensitivity and speed of the UbiFast method makes it suitable for large-scale studies in primary tissue samples.


Subject(s)
Proteins/metabolism , Proteome/analysis , Translational Research, Biomedical/methods , Ubiquitin/metabolism , Ubiquitination/physiology , Animals , Breast Neoplasms , Casein Kinase Ialpha , Female , HeLa Cells , Humans , Ikaros Transcription Factor , Mass Spectrometry/methods , Mice , Multiple Myeloma , Protein Processing, Post-Translational , Proteomics/methods , Sensitivity and Specificity , Staining and Labeling , Ubiquitin-Protein Ligases/metabolism
11.
Nat Biotechnol ; 38(1): 108, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31748691

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Mol Cancer Res ; 18(4): 574-584, 2020 04.
Article in English | MEDLINE | ID: mdl-31862696

ABSTRACT

Genomic analysis of lung adenocarcinomas has revealed that the MGA gene, which encodes a heterodimeric partner of the MYC-interacting protein MAX, is significantly mutated or deleted in lung adenocarcinomas. Most of the mutations are loss of function for MGA, suggesting that MGA may act as a tumor suppressor. Here, we characterize both the molecular and cellular role of MGA in lung adenocarcinomas and illustrate its functional relevance in the MYC pathway. Although MGA and MYC interact with the same binding partner, MAX, and recognize the same E-box DNA motif, we show that the molecular function of MGA appears to be antagonistic to that of MYC. Using mass spectrometry-based affinity proteomics, we demonstrate that MGA interacts with a noncanonical PCGF6-PRC1 complex containing MAX and E2F6 that is involved in gene repression, while MYC is not part of this MGA complex, in agreement with previous studies describing the interactomes of E2F6 and PCGF6. Chromatin immunoprecipitation-sequencing and RNA sequencing assays show that MGA binds to and represses genes that are bound and activated by MYC. In addition, we show that, as opposed to the MYC oncoprotein, MGA acts as a negative regulator for cancer cell proliferation. Our study defines a novel MYC/MAX/MGA pathway, in which MYC and MGA play opposite roles in protein interaction, transcriptional regulation, and cellular proliferation. IMPLICATIONS: This study expands the range of key cancer-associated genes whose dysregulation is functionally equivalent to MYC activation and places MYC within a linear pathway analogous to cell-cycle or receptor tyrosine kinase/RAS/RAF pathways in lung adenocarcinomas.


Subject(s)
Adenocarcinoma of Lung/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Lung Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation/physiology , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutation , Proto-Oncogene Proteins c-myc/genetics
14.
Blood ; 132(14): 1535-1544, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30064974

ABSTRACT

Thalidomide and its derivatives, lenalidomide and pomalidomide, are clinically effective treatments for multiple myeloma and myelodysplastic syndrome with del(5q). These molecules lack activity in murine models, limiting investigation of their therapeutic activity or toxicity in vivo. Here, we report the development of a mouse model that is sensitive to thalidomide derivatives because of a single amino acid change in the direct target of thalidomide derivatives, cereblon (Crbn). In human cells, thalidomide and its analogs bind CRBN and recruit protein targets to the CRL4CRBN E3 ubiquitin ligase, resulting in their ubiquitination and subsequent degradation by the proteasome. We show that mice with a single I391V amino acid change in Crbn exhibit thalidomide-induced degradation of drug targets previously identified in human cells, including Ikaros (Ikzf1), Aiolos (Ikzf3), Zfp91, and casein kinase 1a1 (Ck1α), both in vitro and in vivo. We use the Crbn I391V model to demonstrate that the in vivo therapeutic activity of lenalidomide in del(5q) myelodysplastic syndrome can be explained by heterozygous expression of Ck1α in del(5q) cells. We found that lenalidomide acts on hematopoietic stem cells with heterozygous expression of Ck1α and inactivation of Trp53 causes lenalidomide resistance. We further demonstrate that Crbn I391V is sufficient to confer thalidomide-induced fetal loss in mice, capturing a major toxicity of this class of drugs. Further study of the Crbn I391V model will provide valuable insights into the in vivo efficacy and toxicity of this class of drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Lenalidomide/pharmacology , Myelodysplastic Syndromes/drug therapy , Nerve Tissue Proteins/genetics , Point Mutation , Thalidomide/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents/chemistry , Casein Kinase I/metabolism , Disease Models, Animal , Female , Hematopoiesis/drug effects , Lenalidomide/chemistry , Male , Mice , Mice, Inbred C57BL , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Nerve Tissue Proteins/metabolism , Thalidomide/analogs & derivatives
15.
Nat Biotechnol ; 36(9): 880-887, 2018 10.
Article in English | MEDLINE | ID: mdl-30125270

ABSTRACT

Protein interaction networks and protein compartmentalization underlie all signaling and regulatory processes in cells. Enzyme-catalyzed proximity labeling (PL) has emerged as a new approach to study the spatial and interaction characteristics of proteins in living cells. However, current PL methods require over 18 h of labeling time or utilize chemicals with limited cell permeability or high toxicity. We used yeast display-based directed evolution to engineer two promiscuous mutants of biotin ligase, TurboID and miniTurbo, which catalyze PL with much greater efficiency than BioID or BioID2, and enable 10-min PL in cells with non-toxic and easily deliverable biotin. Furthermore, TurboID extends biotin-based PL to flies and worms.


Subject(s)
Protein Interaction Mapping , Catalysis , Enzymes/metabolism , Mutation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
16.
Genes Dev ; 32(13-14): 929-943, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29950492

ABSTRACT

While a mutation in C9ORF72 is the most common genetic contributor to amyotrophic lateral sclerosis (ALS), much remains to be learned concerning the function of the protein normally encoded at this locus. To elaborate further on functions for C9ORF72, we used quantitative mass spectrometry-based proteomics to identify interacting proteins in motor neurons and found that its long isoform complexes with and stabilizes SMCR8, which further enables interaction with WDR41. To study the organismal and cellular functions for this tripartite complex, we generated Smcr8 loss-of-function mutant mice and found that they developed phenotypes also observed in C9orf72 loss-of-function animals, including autoimmunity. Along with a loss of tolerance for many nervous system autoantigens, we found increased lysosomal exocytosis in Smcr8 mutant macrophages. In addition to elevated surface Lamp1 (lysosome-associated membrane protein 1) expression, we also observed enhanced secretion of lysosomal components-phenotypes that we subsequently observed in C9orf72 loss-of-function macrophages. Overall, our findings demonstrate that C9ORF72 and SMCR8 have interdependent functions in suppressing autoimmunity as well as negatively regulating lysosomal exocytosis-processes of potential importance to ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Autoimmunity/genetics , Carrier Proteins/metabolism , Exocytosis/genetics , Lysosomes/metabolism , Animals , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Carrier Proteins/genetics , Gene Expression Regulation/genetics , Humans , Lymph Nodes/pathology , Lysosomal-Associated Membrane Protein 1/genetics , Macrophages/pathology , Mice , Mice, Knockout , Mutation , Protein Isoforms , Protein Stability , Splenomegaly/genetics
17.
Nat Methods ; 14(12): 1167-1170, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039416

ABSTRACT

Although purification of biotinylated molecules is highly efficient, identifying specific sites of biotinylation remains challenging. We show that anti-biotin antibodies enable unprecedented enrichment of biotinylated peptides from complex peptide mixtures. Live-cell proximity labeling using APEX peroxidase followed by anti-biotin enrichment and mass spectrometry yielded over 1,600 biotinylation sites on hundreds of proteins, an increase of more than 30-fold in the number of biotinylation sites identified compared to streptavidin-based enrichment of proteins.


Subject(s)
Antibodies/metabolism , Biotin/metabolism , Peptides/chemistry , Proteins/chemistry , Biotechnology/methods , Biotinylation , Chromatography, Liquid , HEK293 Cells , Humans , Jurkat Cells , Proteins/isolation & purification , Staining and Labeling , Streptavidin/metabolism , Tandem Mass Spectrometry
18.
Nat Med ; 23(9): 1046-1054, 2017 09.
Article in English | MEDLINE | ID: mdl-28805821

ABSTRACT

It is generally assumed that recurrent mutations within a given cancer driver gene elicit similar drug responses. Cancer genome studies have identified recurrent but divergent missense mutations affecting the substrate-recognition domain of the ubiquitin ligase adaptor SPOP in endometrial and prostate cancers. The therapeutic implications of these mutations remain incompletely understood. Here we analyzed changes in the ubiquitin landscape induced by endometrial cancer-associated SPOP mutations and identified BRD2, BRD3 and BRD4 proteins (BETs) as SPOP-CUL3 substrates that are preferentially degraded by endometrial cancer-associated SPOP mutants. The resulting reduction of BET protein levels sensitized cancer cells to BET inhibitors. Conversely, prostate cancer-specific SPOP mutations resulted in impaired degradation of BETs, promoting their resistance to pharmacologic inhibition. These results uncover an oncogenomics paradox, whereby mutations mapping to the same domain evoke opposing drug susceptibilities. Specifically, we provide a molecular rationale for the use of BET inhibitors to treat patients with endometrial but not prostate cancer who harbor SPOP mutations.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Carcinoma, Endometrioid/genetics , Carcinosarcoma/genetics , Endometrial Neoplasms/genetics , Neoplasms, Cystic, Mucinous, and Serous/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Prostatic Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Transcription Factors/metabolism , Acetanilides/pharmacology , Adenocarcinoma, Clear Cell/metabolism , Animals , Apoptosis/drug effects , Azepines/pharmacology , Carcinoma, Endometrioid/metabolism , Carcinosarcoma/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, Liquid , Cullin Proteins/metabolism , Drug Resistance, Neoplasm , Endometrial Neoplasms/metabolism , Epigenesis, Genetic , Female , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Male , Mass Spectrometry , Mice, Nude , Molecular Targeted Therapy , Mutation , Neoplasm Transplantation , Neoplasms, Cystic, Mucinous, and Serous/metabolism , Nuclear Proteins/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Ubiquitination
19.
Elife ; 62017 04 25.
Article in English | MEDLINE | ID: mdl-28441135

ABSTRACT

The cytosol-facing membranes of cellular organelles contain proteins that enable signal transduction, regulation of morphology and trafficking, protein import and export, and other specialized processes. Discovery of these proteins by traditional biochemical fractionation can be plagued with contaminants and loss of key components. Using peroxidase-mediated proximity biotinylation, we captured and identified endogenous proteins on the outer mitochondrial membrane (OMM) and endoplasmic reticulum membrane (ERM) of living human fibroblasts. The proteomes of 137 and 634 proteins, respectively, are highly specific and highlight 94 potentially novel mitochondrial or ER proteins. Dataset intersection identified protein candidates potentially localized to mitochondria-ER contact sites. We found that one candidate, the tail-anchored, PDZ-domain-containing OMM protein SYNJ2BP, dramatically increases mitochondrial contacts with rough ER when overexpressed. Immunoprecipitation-mass spectrometry identified ribosome-binding protein 1 (RRBP1) as SYNJ2BP's ERM binding partner. Our results highlight the power of proximity biotinylation to yield insights into the molecular composition and function of intracellular membranes.


Subject(s)
Endoplasmic Reticulum/chemistry , Intracellular Membranes/chemistry , Membrane Proteins/analysis , Mitochondria/chemistry , Proteomics/methods , Animals , Biotinylation/methods , COS Cells , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , Peroxidase/metabolism
20.
Cell Chem Biol ; 24(3): 404-414, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28238724

ABSTRACT

A recurring challenge in cell biology is to define the molecular components of macromolecular complexes of interest. The predominant method, immunoprecipitation, recovers only strong interaction partners that survive cell lysis and repeated detergent washes. We explored peroxidase-catalyzed proximity biotinylation, APEX, as an alternative, focusing on the mitochondrial nucleoid, the dynamic macromolecular complex that houses the mitochondrial genome. Via 1-min live-cell biotinylation followed by quantitative, ratiometric mass spectrometry, we enriched 37 nucleoid proteins, seven of which had never previously been associated with the nucleoid. The specificity of our dataset was very high, and we validated three hits by follow-up studies. For one novel nucleoid-associated protein, FASTKD1, we discovered a role in downregulation of mitochondrial complex I via specific repression of ND3 mRNA. Our study demonstrates that APEX is a powerful tool for mapping macromolecular complexes in living cells, and can identify proteins and pathways that have been missed by traditional approaches.


Subject(s)
DNA, Mitochondrial/metabolism , Mitochondria/metabolism , Peroxidase/metabolism , Biocatalysis , Biotinylation , Electron Transport Complex I/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Microscopy, Electron , Microscopy, Fluorescence , Mitochondria/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...