Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Cell ; 172(3): 590-604.e13, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29373831

ABSTRACT

Stress granules (SGs) are transient ribonucleoprotein (RNP) aggregates that form during cellular stress and are increasingly implicated in human neurodegeneration. To study the proteome and compositional diversity of SGs in different cell types and in the context of neurodegeneration-linked mutations, we used ascorbate peroxidase (APEX) proximity labeling, mass spectrometry, and immunofluorescence to identify ∼150 previously unknown human SG components. A highly integrated, pre-existing SG protein interaction network in unstressed cells facilitates rapid coalescence into larger SGs. Approximately 20% of SG diversity is stress or cell-type dependent, with neuronal SGs displaying a particularly complex repertoire of proteins enriched in chaperones and autophagy factors. Strengthening the link between SGs and neurodegeneration, we demonstrate aberrant dynamics, composition, and subcellular distribution of SGs in cells from amyotrophic lateral sclerosis (ALS) patients. Using three Drosophila ALS/FTD models, we identify SG-associated modifiers of neurotoxicity in vivo. Altogether, our results highlight SG proteins as central to understanding and ultimately targeting neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cytoplasmic Granules/metabolism , Protein Interaction Maps , Ribonucleoproteins/metabolism , Stress, Physiological , Animals , Drosophila melanogaster , HEK293 Cells , HeLa Cells , Humans , Neurons/metabolism , Protein Transport
2.
Immunity ; 55(3): 512-526.e9, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263569

ABSTRACT

Dual blockade of the PD-1 and TIGIT coinhibitory receptors on T cells shows promising early results in cancer patients. Here, we studied the mechanisms whereby PD-1 and/or TIGIT blockade modulate anti-tumor CD8+ T cells. Although PD-1 and TIGIT are thought to regulate different costimulatory receptors (CD28 and CD226), effectiveness of PD-1 or TIGIT inhibition in preclinical tumor models was reduced in the absence of CD226. CD226 expression associated with clinical benefit in patients with non-small cell lung carcinoma (NSCLC) treated with anti-PD-L1 antibody atezolizumab. CD226 and CD28 were co-expressed on NSCLC infiltrating CD8+ T cells poised for expansion. Mechanistically, PD-1 inhibited phosphorylation of both CD226 and CD28 via its ITIM-containing intracellular domain (ICD); TIGIT's ICD was dispensable, with TIGIT restricting CD226 co-stimulation by blocking interaction with their common ligand PVR (CD155). Thus, full restoration of CD226 signaling, and optimal anti-tumor CD8+ T cell responses, requires blockade of TIGIT and PD-1, providing a mechanistic rationale for combinatorial targeting in the clinic.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Antigens, Differentiation, T-Lymphocyte/metabolism , CD28 Antigens/metabolism , Humans , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Receptors, Immunologic/metabolism
3.
Mol Cell ; 81(17): 3468-3480.e7, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34314700

ABSTRACT

HECT ubiquitin ligases play essential roles in metazoan development and physiology. The HECT ligase HUWE1 is central to the cellular stress response by mediating degradation of key death or survival factors, including Mcl1, p53, DDIT4, and Myc. Although mutations in HUWE1 and related HECT ligases are widely implicated in human disease, our molecular understanding remains limited. Here we present a comprehensive investigation of full-length HUWE1, deepening our understanding of this class of enzymes. The N-terminal ∼3,900 amino acids of HUWE1 are indispensable for proper ligase function, and our cryo-EM structures of HUWE1 offer a complete molecular picture of this large HECT ubiquitin ligase. HUWE1 forms an alpha solenoid-shaped assembly with a central pore decorated with protein interaction modules. Structures of HUWE1 variants linked to neurodevelopmental disorders as well as of HUWE1 bound to a model substrate link the functions of this essential enzyme to its three-dimensional organization.


Subject(s)
Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Cryoelectron Microscopy/methods , HEK293 Cells , Humans , Stress, Physiological/physiology , Structure-Activity Relationship , Tumor Suppressor Proteins/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
4.
EMBO J ; 42(4): e112275, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36350249

ABSTRACT

Nearly one-third of nascent proteins are initially targeted to the endoplasmic reticulum (ER), where they are correctly folded and assembled before being delivered to their final cellular destinations. To prevent the accumulation of misfolded membrane proteins, ER-associated degradation (ERAD) removes these client proteins from the ER membrane to the cytosol in a process known as retrotranslocation. Our previous work demonstrated that rhomboid pseudoprotease Dfm1 is involved in the retrotranslocation of ubiquitinated membrane integral ERAD substrates. Herein, we found that Dfm1 associates with the SPOTS complex, which is composed of serine palmitoyltransferase (SPT) enzymes and accessory components that are critical for catalyzing the first rate-limiting step of the sphingolipid biosynthesis pathway. Furthermore, Dfm1 employs an ERAD-independent role for facilitating the ER export and endosome- and Golgi-associated degradation (EGAD) of Orm2, which is a major antagonist of SPT activity. Given that the accumulation of human Orm2 homologs, ORMDLs, is associated with various pathologies, our study serves as a molecular foothold for understanding how dysregulation of sphingolipid metabolism leads to various diseases.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Sphingolipids , Humans , Sphingolipids/metabolism , Ubiquitin/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Homeostasis
5.
Plant Cell ; 35(1): 469-487, 2023 01 02.
Article in English | MEDLINE | ID: mdl-36227066

ABSTRACT

Polarization of cells prior to asymmetric cell division is crucial for correct cell divisions, cell fate, and tissue patterning. In maize (Zea mays) stomatal development, the polarization of subsidiary mother cells (SMCs) prior to asymmetric division is controlled by the BRICK (BRK)-PANGLOSS (PAN)-RHO FAMILY GTPASE (ROP) pathway. Two catalytically inactive receptor-like kinases, PAN2 and PAN1, are required for correct division plane positioning. Proteins in the BRK-PAN-ROP pathway are polarized in SMCs, with the polarization of each protein dependent on the previous one. As most of the known proteins in this pathway do not physically interact, possible interactors that might participate in the pathway are yet to be described. We identified WEAK CHLOROPLAST MOVEMENT UNDER BLUE LIGHT 1 (WEB1)/PLASTID MOVEMENT IMPAIRED 2 (PMI2)-RELATED (WPR) proteins as players during SMC polarization in maize. WPRs physically interact with PAN receptors and polarly accumulate in SMCs. The polarized localization of WPR proteins depends on PAN2 but not PAN1. CRISPR-Cas9-induced mutations result in division plane defects in SMCs, and ectopic expression of WPR-RFP results in stomatal defects and alterations to the actin cytoskeleton. We show that certain WPR proteins directly interact with F-actin through their N-terminus. Our data implicate WPR proteins as potentially regulating actin filaments, providing insight into their molecular function. These results demonstrate that WPR proteins are important for cell polarization.


Subject(s)
Plant Proteins , Plant Stomata , Zea mays , Actin Cytoskeleton/metabolism , Cell Division , Plant Proteins/genetics , Plant Proteins/metabolism , Zea mays/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Plant Stomata/growth & development , Plant Stomata/metabolism , Cell Polarity/genetics , Cell Polarity/physiology
6.
Plant Cell ; 35(7): 2678-2693, 2023 06 26.
Article in English | MEDLINE | ID: mdl-37017144

ABSTRACT

Formative asymmetric divisions produce cells with different fates and are critical for development. We show the maize (Zea mays) myosin XI protein, OPAQUE1 (O1), is necessary for asymmetric divisions during maize stomatal development. We analyzed stomatal precursor cells before and during asymmetric division to determine why o1 mutants have abnormal division planes. Cell polarization and nuclear positioning occur normally in the o1 mutant, and the future site of division is correctly specified. The defect in o1 becomes apparent during late cytokinesis, when the phragmoplast forms the nascent cell plate. Initial phragmoplast guidance in o1 is normal; however, as phragmoplast expansion continues o1 phragmoplasts become misguided. To understand how O1 contributes to phragmoplast guidance, we identified O1-interacting proteins. Maize kinesins related to the Arabidopsis thaliana division site markers PHRAGMOPLAST ORIENTING KINESINs (POKs), which are also required for correct phragmoplast guidance, physically interact with O1. We propose that different myosins are important at multiple steps of phragmoplast expansion, and the O1 actin motor and POK-like microtubule motors work together to ensure correct late-stage phragmoplast guidance.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Zea mays/genetics , Zea mays/metabolism , Kinesins/metabolism , Asymmetric Cell Division , Cytokinesis/genetics , Microtubules/metabolism , Arabidopsis/metabolism , Myosins/genetics , Myosins/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Vesicular Transport Proteins/metabolism
7.
Cell ; 141(2): 220-2, 2010 Apr 16.
Article in English | MEDLINE | ID: mdl-20403317

ABSTRACT

Ubiquitin-dependent processes are critical for propagating antiviral defense signals during viral infection. In this issue, Zeng et al. (2010) describe how viral RNA and unanchored ubiquitin chains conspire to promote activation of RIG-I and the host cell's antiviral transcriptional program.

8.
Cell ; 143(6): 951-65, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21145461

ABSTRACT

Dynamic reorganization of signaling systems frequently accompanies pathway perturbations, yet quantitative studies of network remodeling by pathway stimuli are lacking. Here, we report the development of a quantitative proteomics platform centered on multiplex absolute quantification (AQUA) technology to elucidate the architecture of the cullin-RING ubiquitin ligase (CRL) network and to evaluate current models of dynamic CRL remodeling. Current models suggest that CRL complexes are controlled by cycles of CRL deneddylation and CAND1 binding. Contrary to expectations, acute CRL inhibition with MLN4924, an inhibitor of the NEDD8-activating enzyme, does not result in a global reorganization of the CRL network. Examination of CRL complex stoichiometry reveals that, independent of cullin neddylation, a large fraction of cullins are assembled with adaptor modules, whereas only a small fraction are associated with CAND1. These studies suggest an alternative model of CRL dynamicity where the abundance of adaptor modules, rather than cycles of neddylation and CAND1 binding, drives CRL network organization.


Subject(s)
Cullin Proteins/metabolism , Cell Line , Cullin Proteins/chemistry , Cyclopentanes/pharmacology , NEDD8 Protein , Protein Processing, Post-Translational , Proteomics , Pyrimidines/pharmacology , Transcription Factors/metabolism , Ubiquitins/antagonists & inhibitors , Ubiquitins/metabolism
9.
Mol Cell ; 65(4): 751-760.e4, 2017 Feb 16.
Article in English | MEDLINE | ID: mdl-28132843

ABSTRACT

Ribosomes that experience terminal stalls during translation are resolved by ribosome-associated quality control (QC) pathways that oversee mRNA and nascent chain destruction and recycle ribosomal subunits. The proximal factors that sense stalled ribosomes and initiate mammalian ribosome-associated QC events remain undefined. We demonstrate that the ZNF598 ubiquitin ligase and the 40S ribosomal protein RACK1 help to resolve poly(A)-induced stalled ribosomes. They accomplish this by regulating distinct and overlapping regulatory 40S ribosomal ubiquitylation events. ZNF598 primarily mediates regulatory ubiquitylation of RPS10 and RPS20, whereas RACK1 regulates RPS2, RPS3, and RPS20 ubiquitylation. Gain or loss of ZNF598 function or mutations that block RPS10 or RPS20 ubiquitylation result in defective resolution of stalled ribosomes and subsequent readthrough of poly(A)-containing stall sequences. Together, our results indicate that ZNF598, RACK1, and 40S regulatory ubiquitylation plays a pivotal role in mammalian ribosome-associated QC pathways.


Subject(s)
Carrier Proteins/metabolism , GTP-Binding Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , Receptors, Cell Surface/metabolism , Ribosomal Proteins/metabolism , Ribosomes/enzymology , Ubiquitin/metabolism , Ubiquitination , Carrier Proteins/genetics , GTP-Binding Proteins/genetics , HCT116 Cells , HEK293 Cells , Humans , Neoplasm Proteins/genetics , RNA Interference , RNA, Messenger/genetics , Receptors for Activated C Kinase , Receptors, Cell Surface/genetics , Ribosomal Proteins/genetics , Ribosomes/genetics , Transfection
10.
Cell ; 138(2): 389-403, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19615732

ABSTRACT

Deubiquitinating enzymes (Dubs) function to remove covalently attached ubiquitin from proteins, thereby controlling substrate activity and/or abundance. For most Dubs, their functions, targets, and regulation are poorly understood. To systematically investigate Dub function, we initiated a global proteomic analysis of Dubs and their associated protein complexes. This was accomplished through the development of a software platform called CompPASS, which uses unbiased metrics to assign confidence measurements to interactions from parallel nonreciprocal proteomic data sets. We identified 774 candidate interacting proteins associated with 75 Dubs. Using Gene Ontology, interactome topology classification, subcellular localization, and functional studies, we link Dubs to diverse processes, including protein turnover, transcription, RNA processing, DNA damage, and endoplasmic reticulum-associated degradation. This work provides the first glimpse into the Dub interaction landscape, places previously unstudied Dubs within putative biological pathways, and identifies previously unknown interactions and protein complexes involved in this increasingly important arm of the ubiquitin-proteasome pathway.


Subject(s)
Endopeptidases/genetics , Endopeptidases/metabolism , Proteomics , Software , Databases, Protein , Endopeptidases/chemistry , Endoplasmic Reticulum/metabolism , Gene Expression , Humans , Tandem Mass Spectrometry , Ubiquitin/metabolism
11.
J Cell Sci ; 134(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33912921

ABSTRACT

Viral infection both activates stress signaling pathways and redistributes ribosomes away from host mRNAs to translate viral mRNAs. The intricacies of this ribosome shuffle from host to viral mRNAs are poorly understood. Here, we uncover a role for the ribosome-associated quality control (RQC) factor ZNF598 during vaccinia virus mRNA translation. ZNF598 acts on collided ribosomes to ubiquitylate 40S subunit proteins uS10 (RPS20) and eS10 (RPS10), initiating RQC-dependent nascent chain degradation and ribosome recycling. We show that vaccinia infection enhances uS10 ubiquitylation, indicating an increased burden on RQC pathways during viral propagation. Consistent with an increased RQC demand, we demonstrate that vaccinia virus replication is impaired in cells that either lack ZNF598 or express a ubiquitylation-deficient version of uS10. Using SILAC-based proteomics and concurrent RNA-seq analysis, we determine that translation, but not transcription of vaccinia virus mRNAs is compromised in cells with deficient RQC activity. Additionally, vaccinia virus infection reduces cellular RQC activity, suggesting that co-option of ZNF598 by vaccinia virus plays a critical role in translational reprogramming that is needed for optimal viral propagation.


Subject(s)
Vaccinia virus , Vaccinia , Carrier Proteins/metabolism , HEK293 Cells , Humans , Protein Biosynthesis , Quality Control , Ribosomes/metabolism , Vaccinia/genetics , Vaccinia virus/genetics
12.
Mol Cell ; 59(1): 35-49, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26051182

ABSTRACT

Insults to ER homeostasis activate the unfolded protein response (UPR), which elevates protein folding and degradation capacity and attenuates protein synthesis. While a role for ubiquitin in regulating the degradation of misfolded ER-resident proteins is well described, ubiquitin-dependent regulation of translational reprogramming during the UPR remains uncharacterized. Using global quantitative ubiquitin proteomics, we identify evolutionarily conserved, site-specific regulatory ubiquitylation of 40S ribosomal proteins. We demonstrate that these events occur on assembled cytoplasmic ribosomes and are stimulated by both UPR activation and translation inhibition. We further show that ER stress-stimulated regulatory 40S ribosomal ubiquitylation occurs on a timescale similar to eIF2α phosphorylation, is dependent upon PERK signaling, and is required for optimal cell survival during chronic UPR activation. In total, these results reveal regulatory 40S ribosomal ubiquitylation as an important facet of eukaryotic translational control.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Eukaryotic Initiation Factor-2/metabolism , Ribosome Subunits, Small, Eukaryotic/metabolism , Unfolded Protein Response/genetics , eIF-2 Kinase/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Survival , Drosophila/genetics , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Humans , Molecular Sequence Data , Phosphorylation , Protein Biosynthesis/genetics , Saccharomyces cerevisiae/genetics , Ubiquitination
13.
Proc Natl Acad Sci U S A ; 117(14): 7950-7960, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32193347

ABSTRACT

Intracellular pathogen infection leads to proteotoxic stress in host organisms. Previously we described a physiological program in the nematode Caenorhabditis elegans called the intracellular pathogen response (IPR), which promotes resistance to proteotoxic stress and appears to be distinct from canonical proteostasis pathways. The IPR is controlled by PALS-22 and PALS-25, proteins of unknown biochemical function, which regulate expression of genes induced by natural intracellular pathogens. We previously showed that PALS-22 and PALS-25 regulate the mRNA expression of the predicted ubiquitin ligase component cullin cul-6, which promotes thermotolerance in pals-22 mutants. However, it was unclear whether CUL-6 acted alone, or together with other cullin-ring ubiquitin ligase components, which comprise a greatly expanded gene family in C. elegans Here we use coimmunoprecipitation studies paired with genetic analysis to define the cullin-RING ligase components that act together with CUL-6 to promote thermotolerance. First, we identify a previously uncharacterized RING domain protein in the TRIM family we named RCS-1, which acts as a core component with CUL-6 to promote thermotolerance. Next, we show that the Skp-related proteins SKR-3, SKR-4, and SKR-5 act redundantly to promote thermotolerance with CUL-6. Finally, we screened F-box proteins that coimmunoprecipitate with CUL-6 and find that FBXA-158 and FBXA-75 promote thermotolerance. In summary, we have defined the three core components and two F-box adaptors of a cullin-RING ligase complex that promotes thermotolerance as part of the IPR in C. elegans, which adds to our understanding of how organisms cope with proteotoxic stress.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/immunology , Cullin Proteins/metabolism , F-Box Proteins/metabolism , Microsporidia/immunology , Thermotolerance/immunology , Animals , Animals, Genetically Modified , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/immunology , Cullin Proteins/genetics , Cullin Proteins/immunology , F-Box Proteins/immunology , Host-Pathogen Interactions/immunology , Models, Animal , Proteostasis/immunology
14.
Nature ; 537(7620): 328-38, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27629639

ABSTRACT

The cellular proteome is a complex microcosm of structural and regulatory networks that requires continuous surveillance and modification to meet the dynamic needs of the cell. It is therefore crucial that the protein flux of the cell remains in balance to ensure proper cell function. Genetic alterations that range from chromosome imbalance to oncogene activation can affect the speed, fidelity and capacity of protein biogenesis and degradation systems, which often results in proteome imbalance. An improved understanding of the causes and consequences of proteome imbalance is helping to reveal how these systems can be targeted to treat diseases such as cancer.


Subject(s)
Proteome/metabolism , Animals , Autophagy , Chromosome Aberrations , Gene Dosage , Homeostasis/drug effects , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Protein Biosynthesis/drug effects , Protein Transport , Proteome/genetics , Ribosomes/drug effects , Ribosomes/metabolism
15.
Mol Cell ; 52(1): 9-24, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24035498

ABSTRACT

Modular cullin-RING E3 ubiquitin ligases (CRLs) use substrate binding adaptor proteins to specify target ubiquitylation. Many of the ~200 human CRL adaptor proteins remain poorly studied due to a shortage of efficient methods to identify biologically relevant substrates. Here, we report the development of parallel adaptor capture (PAC) proteomics and its use to systematically identify candidate targets for the leucine-rich repeat family of F-box proteins (FBXLs) that function with SKP1-CUL1-F-box protein (SCF) E3s. In validation experiments, we identify the unstudied F-box protein FBXL17 as a regulator of the NFR2 oxidative stress pathway. We demonstrate that FBXL17 controls the transcription of the NRF2 target HMOX1 via turnover of the transcriptional repressor BACH1 in the absence or presence of extrinsic oxidative stress. This work identifies a role for SCF(FBXL17) in controlling the threshold for NRF2-dependent gene activation and provides a framework for elucidating the functions of CRL adaptor proteins.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , F-Box Proteins/metabolism , Fanconi Anemia Complementation Group Proteins/metabolism , Proteomics , SKP Cullin F-Box Protein Ligases/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , F-Box Proteins/genetics , Fanconi Anemia Complementation Group Proteins/genetics , Gene Expression Regulation , HCT116 Cells , HEK293 Cells , HeLa Cells , Heme Oxygenase-1/metabolism , Humans , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Proteomics/methods , RNA Interference , Reproducibility of Results , SKP Cullin F-Box Protein Ligases/genetics , Transcription, Genetic , Transfection
16.
Plant Physiol ; 177(3): 1170-1186, 2018 07.
Article in English | MEDLINE | ID: mdl-29794020

ABSTRACT

Eukaryotic circadian clocks utilize the ubiquitin proteasome system to precisely degrade clock proteins. In plants, the F-box-type E3 ubiquitin ligases ZEITLUPE (ZTL), FLAVIN-BINDING, KELCH REPEAT, F-BOX1 (FKF1), and LOV KELCH PROTEIN2 (LKP2) regulate clock period and couple the clock to photoperiodic flowering in response to end-of-day light conditions. To better understand their functions, we expressed decoy ZTL, FKF1, and LKP2 proteins that associate with target proteins but are unable to ubiquitylate their targets in Arabidopsis (Arabidopsis thaliana). These dominant-negative forms of the proteins inhibit the ubiquitylation of target proteins and allow for the study of ubiquitylation-independent and -dependent functions of ZTL, FKF1, and LKP2. We demonstrate the effects of expressing ZTL, FKF1, and LKP2 decoys on the circadian clock and flowering time. Furthermore, the decoy E3 ligases trap substrate interactions, and using immunoprecipitation-mass spectrometry, we identify interacting partners. We focus studies on the clock transcription factor CCA1 HIKING EXPEDITION (CHE) and show that ZTL interacts directly with CHE and can mediate CHE ubiquitylation. We also demonstrate that CHE protein is degraded in the dark and that degradation is reduced in a ztl mutant plant, showing that CHE is a bona fide ZTL target protein. This work increases our understanding of the genetic and biochemical roles for ZTL, FKF1, and LKP2 and also demonstrates an effective methodology for studying complicated genetic redundancy among E3 ubiquitin ligases.


Subject(s)
Arabidopsis Proteins/metabolism , Circadian Clocks/physiology , Repressor Proteins/metabolism , Arabidopsis/physiology , Arabidopsis Proteins/genetics , Flowers/physiology , Gene Expression Regulation, Plant , Plants, Genetically Modified , Protein Domains , Protein Interaction Maps , Repressor Proteins/genetics , Ubiquitination
17.
Mol Cell ; 44(2): 325-40, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21906983

ABSTRACT

Despite the diverse biological pathways known to be regulated by ubiquitylation, global identification of substrates that are targeted for ubiquitylation has remained a challenge. To globally characterize the human ubiquitin-modified proteome (ubiquitinome), we utilized a monoclonal antibody that recognizes diglycine (diGly)-containing isopeptides following trypsin digestion. We identify ~19,000 diGly-modified lysine residues within ~5000 proteins. Using quantitative proteomics we monitored temporal changes in diGly site abundance in response to both proteasomal and translational inhibition, indicating both a dependence on ongoing translation to observe alterations in site abundance and distinct dynamics of individual modified lysines in response to proteasome inhibition. Further, we demonstrate that quantitative diGly proteomics can be utilized to identify substrates for cullin-RING ubiquitin ligases. Interrogation of the ubiquitinome allows for not only a quantitative assessment of alterations in protein homeostasis fidelity, but also identification of substrates for individual ubiquitin pathway enzymes.


Subject(s)
Proteome/metabolism , Ubiquitin/metabolism , Cells, Cultured , Cullin Proteins/metabolism , Glycylglycine/genetics , HCT116 Cells , Humans , Lysine/genetics , Proteomics , Ubiquitination
18.
J Biol Chem ; 292(8): 3112-3128, 2017 02 24.
Article in English | MEDLINE | ID: mdl-28077573

ABSTRACT

A surprising feature of endoplasmic reticulum (ER)-associated degradation (ERAD) is the movement, or retrotranslocation, of ubiquitinated substrates from the ER lumen or membrane to the cytosol where they are degraded by the 26S proteasome. Multispanning ER membrane proteins, called ERAD-M substrates, are retrotranslocated to the cytosol as full-length intermediates during ERAD, and we have investigated how they maintain substrate solubility. Using an in vivo assay, we show that retrotranslocated ERAD-M substrates are moved to the cytoplasm as part of the normal ERAD pathway, where they are part of a solely proteinaceous complex. Using proteomics and direct biochemical confirmation, we found that Cdc48 serves as a critical "retrochaperone" for these ERAD-M substrates. Cdc48 binding to retrotranslocated, ubiquitinated ERAD-M substrates is required for their solubility; removal of the polyubiquitin chains or competition for binding by addition of free polyubiquitin liberated Cdc48 from retrotranslocated proteins and rendered them insoluble. All components of the canonical Cdc48 complex Cdc48-Npl4-Ufd1 were present in solubilized ERAD-M substrates. This function of the complex was observed for both HRD and DOA pathway substrates. Thus, in addition to the long known ATP-dependent extraction of ERAD substrates during retrotranslocation, the Cdc48 complex is generally and critically needed for the solubility of retrotranslocated ERAD-M intermediates.


Subject(s)
Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/metabolism , Endoplasmic Reticulum-Associated Degradation , Saccharomyces cerevisiae/metabolism , Endoplasmic Reticulum/metabolism , Hydroxymethylglutaryl CoA Reductases/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/metabolism , Solubility , Ubiquitin/metabolism , Ubiquitination , Valosin Containing Protein
19.
RNA ; 22(3): 373-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26763119

ABSTRACT

The zinc finger protein tristetraprolin (TTP) promotes translation repression and degradation of mRNAs containing AU-rich elements (AREs). Although much attention has been directed toward understanding the decay process and machinery involved, the translation repression role of TTP has remained poorly understood. Here we identify the cap-binding translation repression 4EHP-GYF2 complex as a cofactor of TTP. Immunoprecipitation and in vitro pull-down assays demonstrate that TTP associates with the 4EHP-GYF2 complex via direct interaction with GYF2, and mutational analyses show that this interaction occurs via conserved tetraproline motifs of TTP. Mutant TTP with diminished 4EHP-GYF2 binding is impaired in its ability to repress a luciferase reporter ARE-mRNA. 4EHP knockout mouse embryonic fibroblasts (MEFs) display increased induction and slower turnover of TTP-target mRNAs as compared to wild-type MEFs. Our work highlights the function of the conserved tetraproline motifs of TTP and identifies 4EHP-GYF2 as a cofactor in translational repression and mRNA decay by TTP.


Subject(s)
AU Rich Elements , Eukaryotic Initiation Factor-4E/metabolism , Proline/metabolism , RNA Caps/metabolism , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Tristetraprolin/metabolism , Animals , Cell Line , Eukaryotic Initiation Factor-4E/genetics , Hydrolysis , Mice , Mice, Knockout , Protein Binding , Tristetraprolin/chemistry
20.
Mol Cell Proteomics ; 15(8): 2576-93, 2016 08.
Article in English | MEDLINE | ID: mdl-27185884

ABSTRACT

Protein homeostasis dysfunction has been implicated in the development and progression of aging related human pathologies. There is a need for the establishment of quantitative methods to evaluate global protein homoeostasis function. As the ubiquitin (ub) proteasome system plays a key role in regulating protein homeostasis, we applied quantitative proteomic methods to evaluate the sensitivity of site-specific ubiquitylation events as markers for protein homeostasis dysfunction. Here, we demonstrate that the ub-modified proteome can exceed the sensitivity of engineered fluorescent reporters as a marker for proteasome dysfunction and can provide unique signatures for distinct proteome challenges which is not possible with engineered reporters. We demonstrate that combining ub-proteomics with subcellular fractionation can effectively separate degradative and regulatory ubiquitylation events on distinct protein populations. Using a recently developed potent inhibitor of the critical protein homeostasis factor p97/VCP, we demonstrate that distinct insults to protein homeostasis function can elicit robust and largely unique alterations to the ub-modified proteome. Taken together, we demonstrate that proteomic approaches to monitor the ub-modified proteome can be used to evaluate global protein homeostasis and can be used to monitor distinct functional outcomes for spatially separated protein populations.


Subject(s)
Proteasome Inhibitors/pharmacology , Proteome/metabolism , Proteomics/methods , Ubiquitins/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Binding Sites , Cell Cycle Proteins/antagonists & inhibitors , Chromatography, Liquid , HCT116 Cells , Homeostasis/drug effects , Humans , Protein Interaction Maps , Proteome/chemistry , Proteome/drug effects , Tandem Mass Spectrometry , Valosin Containing Protein
SELECTION OF CITATIONS
SEARCH DETAIL