Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(8): 105040, 2023 08.
Article in English | MEDLINE | ID: mdl-37442237

ABSTRACT

Cu/Zn-superoxide dismutase (CuZnSOD) is an enzyme that binds a copper and zinc ion and also forms an intramolecular disulfide bond. Together with the copper ion as the active site, the disulfide bond is completely conserved among these proteins; indeed, the disulfide bond plays critical roles in maintaining the catalytically competent conformation of CuZnSOD. Here, we found that a CuZnSOD protein in Paenibacillus lautus (PaSOD) has no Cys residue but exhibits a significant level of enzyme activity. The crystal structure of PaSOD revealed hydrophobic and hydrogen-bonding interactions in substitution for the disulfide bond of the other CuZnSOD proteins. Also notably, we determined that PaSOD forms a homodimer through an additional domain with a novel fold at the N terminus. While the advantages of lacking Cys residues and adopting a novel dimer configuration remain obscure, PaSOD does not require a disulfide-introducing/correcting system for maturation and could also avoid misfolding caused by aberrant thiol oxidations under an oxidative environment.


Subject(s)
Bacterial Proteins , Disulfides , Superoxide Dismutase-1 , Copper , Cysteine , Disulfides/chemistry , Superoxide Dismutase-1/chemistry , Zinc , Bacterial Proteins/chemistry , Paenibacillus , Protein Folding
2.
J Biol Chem ; 299(6): 104798, 2023 06.
Article in English | MEDLINE | ID: mdl-37156398

ABSTRACT

Canine degenerative myelopathy (DM), a fatal neurodegenerative disease in dogs, shares clinical and genetic features with amyotrophic lateral sclerosis, a human motor neuron disease. Mutations in the SOD1 gene encoding Cu/Zn superoxide dismutase (SOD1) cause canine DM and a subset of inherited human amyotrophic lateral sclerosis. The most frequent DM causative mutation is homozygous E40K mutation, which induces the aggregation of canine SOD1 but not of human SOD1. However, the mechanism through which canine E40K mutation induces species-specific aggregation of SOD1 remains unknown. By screening human/canine chimeric SOD1s, we identified that the humanized mutation of the 117th residue (M117L), encoded by exon 4, significantly reduced aggregation propensity of canine SOD1E40K. Conversely, introducing a mutation of leucine 117 to methionine, a residue homologous to canine, promoted E40K-dependent aggregation in human SOD1. M117L mutation improved protein stability and reduced cytotoxicity of canine SOD1E40K. Furthermore, crystal structural analysis of canine SOD1 proteins revealed that M117L increased the packing within the hydrophobic core of the ß-barrel structure, contributing to the increased protein stability. Our findings indicate that the structural vulnerability derived intrinsically from Met 117 in the hydrophobic core of the ß-barrel structure induces E40K-dependent species-specific aggregation in canine SOD1.


Subject(s)
Dog Diseases , Mutation , Neurodegenerative Diseases , Superoxide Dismutase-1 , Animals , Dogs , Humans , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/veterinary , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Dog Diseases/genetics , Dog Diseases/metabolism , Species Specificity
3.
Nat Commun ; 13(1): 1790, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35379805

ABSTRACT

Despite the recent clinical success of T cell checkpoint inhibition targeting the CTLA-4 and PD-1 pathways, many patients either fail to achieve objective responses or they develop resistance to therapy. In some cases, poor responses to checkpoint blockade have been linked to suboptimal CD28 costimulation and the inability to generate and maintain a productive adaptive anti-tumor immune response. To address this, here we utilize directed evolution to engineer a CD80 IgV domain with increased PD-L1 affinity and fuse this to an immunoglobulin Fc domain, creating a therapeutic (ALPN-202, davoceticept) capable of providing CD28 costimulation in a PD-L1-dependent fashion while also antagonizing PD-1 - PD-L1 and CTLA-4-CD80/CD86 interactions. We demonstrate that by combining CD28 costimulation and dual checkpoint inhibition, ALPN-202 enhances T cell activation and anti-tumor efficacy in cell-based assays and mouse tumor models more potently than checkpoint blockade alone and thus has the potential to generate potent, clinically meaningful anti-tumor immunity in humans.


Subject(s)
CD28 Antigens , Neoplasms , Animals , B7-1 Antigen/metabolism , CD28 Antigens/metabolism , Humans , Lymphocyte Activation , Mice , Neoplasms/drug therapy , Neoplasms/genetics , T-Lymphocytes
4.
Commun Biol ; 4(1): 1238, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34716405

ABSTRACT

C-phycocyanin (CPC), a blue pigment protein, is an indispensable component of giant phycobilisomes, which are light-harvesting antenna complexes in cyanobacteria that transfer energy efficiently to photosystems I and II. X-ray crystallographic and electron microscopy (EM) analyses have revealed the structure of CPC to be a closed toroidal hexamer by assembling two trimers. In this study, the structural characterization of non-conventional octameric CPC is reported for the first time. Analyses of the crystal and cryogenic EM structures of the native CPC from filamentous thermophilic cyanobacterium Thermoleptolyngbya sp. O-77 unexpectedly illustrated the coexistence of conventional hexamer and novel octamer. In addition, an unusual dimeric state, observed via analytical ultracentrifugation, was postulated to be a key intermediate structure in the assemble of the previously unobserved octamer. These observations provide new insights into the assembly processes of CPCs and the mechanism of energy transfer in the light-harvesting complexes.


Subject(s)
Bacterial Proteins/chemistry , Cyanobacteria/chemistry , Phycocyanin/chemistry
5.
J Med Chem ; 64(7): 3720-3746, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33769048

ABSTRACT

Autophagy is the common name for a number of lysosome-based degradation pathways of cytosolic cargos. The key components of autophagy are members of Atg8 family proteins involved in almost all steps of the process, from autophagosome formation to their selective fusion with lysosomes. In this study, we show that the homologous members of the human Atg8 family proteins, LC3A and LC3B, are druggable by a small molecule inhibitor novobiocin. Structure-activity relationship (SAR) studies of the 4-hydroxy coumarin core scaffold were performed, supported by a crystal structure of the LC3A dihydronovobiocin complex. The study reports the first nonpeptide inhibitors for these protein interaction targets and will lay the foundation for the development of more potent chemical probes for the Atg8 protein family which may also find applications for the development of autophagy-mediated degraders (AUTACs).


Subject(s)
4-Hydroxycoumarins/pharmacology , Autophagy/drug effects , Microtubule-Associated Proteins/metabolism , Protein Binding/drug effects , Sequestosome-1 Protein/metabolism , 4-Hydroxycoumarins/chemical synthesis , 4-Hydroxycoumarins/metabolism , HEK293 Cells , Humans , Ligands , Molecular Structure , Novobiocin/chemistry , Structure-Activity Relationship
6.
Cell Chem Biol ; 27(11): 1441-1451.e7, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32726587

ABSTRACT

Protein-protein interactions (PPIs) govern intracellular life, and identification of PPI inhibitors is challenging. Roadblocks in assay development stemming from weak binding affinities of natural PPIs impede progress in this field. We postulated that enhancing binding affinity of natural PPIs via protein engineering will aid assay development and hit discovery. This proof-of-principle study targets PPI between linear ubiquitin chains and NEMO UBAN domain, which activates NF-κB signaling. Using phage display, we generated ubiquitin variants that bind to the functional UBAN epitope with high affinity, act as competitive inhibitors, and structurally maintain the existing PPI interface. When utilized in assay development, variants enable generation of robust cell-based assays for chemical screening. Top compounds identified using this approach directly bind to UBAN and dampen NF-κB signaling. This study illustrates advantages of integrating protein engineering and chemical screening in hit identification, a development that we anticipate will have wide application in drug discovery.


Subject(s)
Biological Products/pharmacology , Drug Discovery , NF-kappa B/antagonists & inhibitors , Protein Engineering , Ubiquitin/antagonists & inhibitors , Biological Products/chemistry , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Molecular Structure , NF-kappa B/chemistry , NF-kappa B/metabolism , Protein Binding/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Ubiquitin/chemistry , Ubiquitin/metabolism
7.
Cell Rep ; 30(9): 3117-3126.e4, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130911

ABSTRACT

Timely completion of DNA replication is central to accurate cell division and to the maintenance of genomic stability. However, certain DNA-protein interactions can physically impede DNA replication fork progression. Cells remove or bypass these physical impediments by different mechanisms to preserve DNA macromolecule integrity and genome stability. In Saccharomyces cerevisiae, Wss1, the DNA-protein crosslink repair protease, allows cells to tolerate hydroxyurea-induced replication stress, but the underlying mechanism by which Wss1 promotes this function has remained unknown. Here, we report that Wss1 provides cells tolerance to replication stress by directly degrading core histone subunits that non-specifically and non-covalently bind to single-stranded DNA. Unlike Wss1-dependent proteolysis of covalent DNA-protein crosslinks, proteolysis of histones does not require Cdc48 nor SUMO-binding activities. Wss1 thus acts as a multi-functional protease capable of targeting a broad range of covalent and non-covalent DNA-binding proteins to preserve genome stability during adverse conditions.


Subject(s)
DNA Replication , Histones/metabolism , Proteolysis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/physiology , Stress, Physiological , DNA Replication/drug effects , Hydroxyurea/toxicity , Mutation/genetics , Proteolysis/drug effects , Saccharomyces cerevisiae/drug effects , Stress, Physiological/drug effects
8.
Autophagy ; 16(2): 256-270, 2020 02.
Article in English | MEDLINE | ID: mdl-30990354

ABSTRACT

Short linear motifs, known as LC3-interacting regions (LIRs), interact with mactoautophagy/autophagy modifiers (Atg8/LC3/GABARAP proteins) via a conserved universal mechanism. Typically, this includes the occupancy of 2 hydrophobic pockets on the surface of Atg8-family proteins by 2 specific aromatic and hydrophobic residues within the LIR motifs. Here, we describe an alternative mechanism of Atg8-family protein interaction with the non-canonical UBA5 LIR, an E1-like enzyme of the ufmylation pathway that preferentially interacts with GABARAP but not LC3 proteins. By solving the structures of both GABARAP and GABARAPL2 in complex with the UBA5 LIR, we show that in addition to the binding to the 2 canonical hydrophobic pockets (HP1 and HP2), a conserved tryptophan residue N-terminal of the LIR core sequence binds into a novel hydrophobic pocket on the surface of GABARAP proteins, which we term HP0. This mode of action is unique for UBA5 and accompanied by large rearrangements of key residues including the side chains of the gate-keeping K46 and the adjacent K/R47 in GABARAP proteins. Swapping mutations in LC3B and GABARAPL2 revealed that K/R47 is the key residue in the specific binding of GABARAP proteins to UBA5, with synergetic contributions of the composition and dynamics of the loop L3. Finally, we elucidate the physiological relevance of the interaction and show that GABARAP proteins regulate the localization and function of UBA5 on the endoplasmic reticulum membrane in a lipidation-independent manner.Abbreviations: ATG: AuTophaGy-related; EGFP: enhanced green fluorescent protein; GABARAP: GABA-type A receptor-associated protein; ITC: isothermal titration calorimetry; KO: knockout; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; NMR: nuclear magnetic resonance; RMSD: root-mean-square deviation of atomic positions; TKO: triple knockout; UBA5: ubiquitin like modifier activating enzyme 5.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 8 Family/metabolism , Intracellular Membranes/metabolism , Microtubule-Associated Proteins/metabolism , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/metabolism , Amino Acid Motifs , Amino Acid Sequence , Apoptosis Regulatory Proteins/chemistry , Autophagy-Related Protein 8 Family/chemistry , Autophagy-Related Protein 8 Family/genetics , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Lysine/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Models, Molecular , Mutation/genetics , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Secondary , Structure-Activity Relationship
9.
Nat Commun ; 9(1): 1017, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523821

ABSTRACT

Ultraviolet (UV) light radiation induces the formation of bulky photoproducts in the DNA that globally affect transcription and splicing. However, the signaling pathways and mechanisms that link UV-light-induced DNA damage to changes in RNA metabolism remain poorly understood. Here we employ quantitative phosphoproteomics and protein kinase inhibition to provide a systems view on protein phosphorylation patterns induced by UV light and uncover the dependencies of phosphorylation events on the canonical DNA damage signaling by ATM/ATR and the p38 MAP kinase pathway. We identify RNA-binding proteins as primary substrates and 14-3-3 as direct readers of p38-MK2-dependent phosphorylation induced by UV light. Mechanistically, we show that MK2 phosphorylates the RNA-binding subunit of the NELF complex NELFE on Serine 115. NELFE phosphorylation promotes the recruitment of 14-3-3 and rapid dissociation of the NELF complex from chromatin, which is accompanied by RNA polymerase II elongation.


Subject(s)
DNA Damage/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA/metabolism , Ultraviolet Rays/adverse effects , p38 Mitogen-Activated Protein Kinases/metabolism , 14-3-3 Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Chromatin/metabolism , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Phosphorylation , RNA Polymerase II/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism
10.
Biochemistry ; 56(47): 6281-6291, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29094929

ABSTRACT

Serratia marcescens secretes a lipase, LipA, through a type I secretion system (T1SS). The T1SS for LipA, the Lip system, is composed of an inner membrane ABC transporter with its nucleotide-binding domains (NBD), LipB, a membrane fusion protein, LipC, and an outer membrane channel protein, LipD. Passenger protein secreted by this system has been functionally and structurally characterized well, but relatively little information about the transporter complex is available. Here, we report the crystallographic studies of LipC without the membrane anchor region, LipC-, and the NBD of LipB (LipB-NBD). LipC- crystallographic analysis has led to the determination of the structure of the long α-helical and lipoyl domains, but not the area where it interacts with LipB, suggesting that the region is flexible without LipB. The long α-helical domain has three α-helices, which interacts with LipD in the periplasm. LipB-NBD has the common overall architecture and ATP hydrolysis activity of ABC transporter NBDs. Using the predicted models of full-length LipB and LipD, the overall structural insight into the Lip system is discussed.


Subject(s)
Bacterial Proteins/chemistry , Lipase/chemistry , Lipase/metabolism , Membrane Fusion Proteins/chemistry , Membrane Fusion/physiology , Nucleotides/metabolism , Serratia marcescens/enzymology , Bacterial Proteins/metabolism , Crystallography, X-Ray , Membrane Fusion Proteins/metabolism , Nucleotides/chemistry , Protein Conformation
11.
Cell Death Dis ; 8(6): e2900, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28661476

ABSTRACT

Rho GTPases control fundamental cellular processes and Cdc42 is a well-studied member of the family that controls filopodia formation and cell migration. Although the regulation of Cdc42 activity by nucleotide binding is well documented, the mechanisms driving its proteostasis are not clear. Here, we demonstrate that the highly conserved, RING domain containing E3 ubiquitin ligase XIAP controls the protein stability of Cdc42. XIAP binds to Cdc42 and directly conjugates poly ubiquitin chains to the Lysine 166 of Cdc42 targeting it for proteasomal degradation. Depletion of XIAP led to an increased protein stability and activity of Cdc42 in normal and tumor cells. Consistently, loss of XIAP enhances filopodia formation in a Cdc42-dependent manner and this phenomenon phenocopies EGF stimulation. Further, XIAP depletion promotes lung colonization of tumor cells in mice in a Cdc42-dependent manner. These observations shed molecular insights into ubiquitin-dependent regulation of Cdc42 and that of actin cytoskeleton.


Subject(s)
Apoptosis/genetics , Neoplasms/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics , cdc42 GTP-Binding Protein/genetics , Actin Cytoskeleton/genetics , Animals , Cell Movement/genetics , HeLa Cells , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Protein Stability , Pseudopodia/genetics , Signal Transduction/genetics , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism , Xenograft Model Antitumor Assays , cdc42 GTP-Binding Protein/metabolism
12.
Nature ; 538(7625): 402-405, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27732584

ABSTRACT

The post-translational modification of proteins with polyubiquitin regulates virtually all aspects of cell biology. Eight distinct chain linkage types co-exist in polyubiquitin and are independently regulated in cells. This 'ubiquitin code' determines the fate of the modified protein. Deubiquitinating enzymes of the ovarian tumour (OTU) family regulate cellular signalling by targeting distinct linkage types within polyubiquitin, and understanding their mechanisms of linkage specificity gives fundamental insights into the ubiquitin system. Here we reveal how the deubiquitinase Cezanne (also known as OTUD7B) specifically targets Lys11-linked polyubiquitin. Crystal structures of Cezanne alone and in complex with monoubiquitin and Lys11-linked diubiquitin, in combination with hydrogen-deuterium exchange mass spectrometry, enable us to reconstruct the enzymatic cycle in great detail. An intricate mechanism of ubiquitin-assisted conformational changes activates the enzyme, and while all chain types interact with the enzymatic S1 site, only Lys11-linked chains can bind productively across the active site and stimulate catalytic turnover. Our work highlights the plasticity of deubiquitinases and indicates that new conformational states can occur when a true substrate, such as diubiquitin, is bound at the active site.


Subject(s)
Deubiquitinating Enzymes/metabolism , Endopeptidases/metabolism , Lysine/metabolism , Polyubiquitin/metabolism , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Deubiquitinating Enzymes/chemistry , Deubiquitinating Enzymes/genetics , Deuterium Exchange Measurement , Endopeptidases/chemistry , Endopeptidases/genetics , Enzyme Activation , Humans , Mass Spectrometry , Models, Molecular , Protein Binding , Protein Conformation , Substrate Specificity , Ubiquitination , Ubiquitins/metabolism
13.
J Biol Chem ; 291(17): 9025-41, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26929408

ABSTRACT

The covalent conjugation of ubiquitin-fold modifier 1 (UFM1) to proteins generates a signal that regulates transcription, response to cell stress, and differentiation. Ufmylation is initiated by ubiquitin-like modifier activating enzyme 5 (UBA5), which activates and transfers UFM1 to ubiquitin-fold modifier-conjugating enzyme 1 (UFC1). The details of the interaction between UFM1 and UBA5 required for UFM1 activation and its downstream transfer are however unclear. In this study, we described and characterized a combined linear LC3-interacting region/UFM1-interacting motif (LIR/UFIM) within the C terminus of UBA5. This single motif ensures that UBA5 binds both UFM1 and light chain 3/γ-aminobutyric acid receptor-associated proteins (LC3/GABARAP), two ubiquitin (Ub)-like proteins. We demonstrated that LIR/UFIM is required for the full biological activity of UBA5 and for the effective transfer of UFM1 onto UFC1 and a downstream protein substrate both in vitro and in cells. Taken together, our study provides important structural and functional insights into the interaction between UBA5 and Ub-like modifiers, improving the understanding of the biology of the ufmylation pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Microtubule-Associated Proteins/metabolism , Protein Processing, Post-Translational/physiology , Proteins/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Motifs , Apoptosis Regulatory Proteins , HEK293 Cells , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Protein Structure, Secondary , Proteins/chemistry , Proteins/genetics , Structure-Activity Relationship , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics
14.
J Cell Sci ; 129(5): 875-80, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26906419

ABSTRACT

Ubiquitin plays an essential role in modulating protein functions, and deregulation of the ubiquitin system leads to the development of multiple human diseases. Owing to its molecular features, ubiquitin can form various homo- and heterotypic polymers on substrate proteins, thereby provoking distinct cellular responses. The concept of multifaceted ubiquitin chains encoding different functions has been substantiated in recent years. It has been established that all possible ubiquitin linkage types are utilized for chain assembly and propagation of specific signals in vivo. In addition, branched ubiquitin chains and phosphorylated ubiquitin molecules have been put under the spotlight recently. The development of novel technologies has provided detailed insights into the structure and function of previously poorly understood ubiquitin signals. In this Cell Science at a Glance article and accompanying poster, we provide an update on the complexity of ubiquitin chains and their physiological relevance.


Subject(s)
Ubiquitin/chemistry , Ubiquitination , Amino Acid Sequence , Animals , Humans , Immunity, Innate , Mitophagy , Proteolysis , Signal Transduction , Ubiquitin/physiology
15.
Mol Cell ; 60(1): 89-104, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26431026

ABSTRACT

Hereditary spastic paraplegias (HSPs) are a diverse group of neurodegenerative diseases that are characterized by axonopathy of the corticospinal motor neurons. A mutation in the gene encoding for Tectonin ß-propeller containing protein 2 (TECPR2) causes HSP that is complicated by neurological symptoms. While TECPR2 is a human ATG8 binding protein and positive regulator of autophagy, the exact function of TECPR2 is unknown. Here, we show that TECPR2 associates with several trafficking components, among them the COPII coat protein SEC24D. TECPR2 is required for stabilization of SEC24D protein levels, maintenance of functional ER exit sites (ERES), and efficient ER export in a manner dependent on binding to lipidated LC3C. TECPR2-deficient HSP patient cells display alterations in SEC24D abundance and ER export efficiency. Additionally, TECPR2 and LC3C are required for autophagosome formation, possibly through maintaining functional ERES. Collectively, these results reveal that TECPR2 functions as molecular scaffold linking early secretion pathway and autophagy.


Subject(s)
Autophagy , Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Transport , Spastic Paraplegia, Hereditary/genetics , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins , Carrier Proteins/genetics , HeLa Cells , Humans , Mutation , Nerve Tissue Proteins/genetics , Spastic Paraplegia, Hereditary/metabolism , Vesicular Transport Proteins/metabolism
16.
Nature ; 522(7556): 354-8, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26040720

ABSTRACT

The endoplasmic reticulum (ER) is the largest intracellular endomembrane system, enabling protein and lipid synthesis, ion homeostasis, quality control of newly synthesized proteins and organelle communication. Constant ER turnover and modulation is needed to meet different cellular requirements and autophagy has an important role in this process. However, its underlying regulatory mechanisms remain unexplained. Here we show that members of the FAM134 reticulon protein family are ER-resident receptors that bind to autophagy modifiers LC3 and GABARAP, and facilitate ER degradation by autophagy ('ER-phagy'). Downregulation of FAM134B protein in human cells causes an expansion of the ER, while FAM134B overexpression results in ER fragmentation and lysosomal degradation. Mutant FAM134B proteins that cause sensory neuropathy in humans are unable to act as ER-phagy receptors. Consistently, disruption of Fam134b in mice causes expansion of the ER, inhibits ER turnover, sensitizes cells to stress-induced apoptotic cell death and leads to degeneration of sensory neurons. Therefore, selective ER-phagy via FAM134 proteins is indispensable for mammalian cell homeostasis and controls ER morphology and turnover in mice and humans.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins , Biomarkers/metabolism , Cell Line , Endoplasmic Reticulum/chemistry , Female , Gene Deletion , Humans , Intracellular Signaling Peptides and Proteins , Lysosomes/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Phagosomes/metabolism , Protein Binding , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/pathology
17.
Mol Cell ; 57(6): 995-1010, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25684205

ABSTRACT

The small Rho GTPase RAC1 is an essential regulator of cellular signaling that controls actin rearrangements and cell motility. Here, we identify a novel CUL3 RING ubiquitin ligase complex, containing the substrate adaptors KBTBD6 and KBTBD7, that mediates ubiquitylation and proteasomal degradation of TIAM1, a RAC1-specific GEF. Increasing the abundance of TIAM1 by depletion of KBTBD6 and/or KBTBD7 leads to elevated RAC1 activity, changes in actin morphology, loss of focal adhesions, reduced proliferation, and enhanced invasion. KBTBD6 and KBTBD7 employ ATG8 family-interacting motifs to bind preferentially to GABARAP proteins. Surprisingly, ubiquitylation and degradation of TIAM1 by CUL3(KBTBD6/KBTBD7) depends on its binding to GABARAP proteins. Our study reveals that recruitment of CUL3(KBTBD6/KBTBD7) to GABARAP-containing vesicles regulates the abundance of membrane-associated TIAM1 and subsequently spatially restricted RAC1 signaling. Besides their role in autophagy and trafficking, we uncovered a previously unknown function of GABARAP proteins as membrane-localized signaling scaffolds.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cullin Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Microtubule-Associated Proteins/metabolism , Trans-Activators/metabolism , rac1 GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Apoptosis Regulatory Proteins , Autophagy-Related Protein 8 Family , Cullin Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Humans , Intracellular Signaling Peptides and Proteins , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Protein Multimerization , Signal Transduction , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Trans-Activators/genetics , Ubiquitination , rac1 GTP-Binding Protein/genetics
18.
Cell Host Microbe ; 17(1): 58-71, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25500191

ABSTRACT

The host endolysosomal compartment is often manipulated by intracellular bacterial pathogens. Salmonella (Salmonella enterica serovar Typhimurium) secrete numerous effector proteins, including SifA, through a specialized type III secretion system to hijack the host endosomal system and generate the Salmonella-containing vacuole (SCV). To form this replicative niche, Salmonella targets the Rab7 GTPase to recruit host membranes through largely unknown mechanisms. We show that Pleckstrin homology domain-containing protein family member 1 (PLEKHM1), a lysosomal adaptor, is targeted by Salmonella through direct interaction with SifA. By binding the PLEKHM1 PH2 domain, Salmonella utilize a complex containing PLEKHM1, Rab7, and the HOPS tethering complex to mobilize phagolysosomal membranes to the SCV. Depletion of PLEKHM1 causes a profound defect in SCV morphology with multiple bacteria accumulating in enlarged structures and significantly dampens Salmonella proliferation in multiple cell types and mice. Thus, PLEKHM1 provides a critical interface between pathogenic infection and the host endolysosomal system.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bacterial Proteins/metabolism , Glycoproteins/metabolism , Host-Pathogen Interactions , Membrane Glycoproteins/metabolism , Salmonella typhimurium/growth & development , Vacuoles/microbiology , Animals , Autophagy-Related Proteins , Carrier Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Protein Binding , Protein Interaction Mapping , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
19.
Mol Cell ; 54(3): 349-61, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24726327

ABSTRACT

Linear ubiquitin chains are implicated in the regulation of the NF-κB pathway, immunity, and inflammation. They are synthesized by the LUBAC complex containing the catalytic subunit HOIL-1-interacting protein (HOIP) and are disassembled by the linear ubiquitin-specific deubiquitinase OTULIN. Little is known about the regulation of these opposing activities. Here we demonstrate that HOIP and OTULIN interact and act as a bimolecular editing pair for linear ubiquitin signals in vivo. The HOIP PUB domain binds to the PUB interacting motif (PIM) of OTULIN and the chaperone VCP/p97. Structural studies revealed the basis of high-affinity interaction with the OTULIN PIM. The conserved Tyr56 of OTULIN makes critical contacts with the HOIP PUB domain, and its phosphorylation negatively regulates this interaction. Functionally, HOIP binding to OTULIN is required for the recruitment of OTULIN to the TNF receptor complex and to counteract HOIP-dependent activation of the NF-κB pathway.


Subject(s)
Endopeptidases/chemistry , NF-kappa B/metabolism , Ubiquitin-Protein Ligases/chemistry , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Crystallography, X-Ray , Endopeptidases/metabolism , HeLa Cells , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Protein Structure, Secondary , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Thermodynamics , Ubiquitin-Protein Ligases/metabolism , Valosin Containing Protein
20.
Cell ; 154(1): 169-84, 2013 Jul 03.
Article in English | MEDLINE | ID: mdl-23827681

ABSTRACT

Sixteen ovarian tumor (OTU) family deubiquitinases (DUBs) exist in humans, and most members regulate cell-signaling cascades. Several OTU DUBs were reported to be ubiquitin (Ub) chain linkage specific, but comprehensive analyses are missing, and the underlying mechanisms of linkage specificity are unclear. Using Ub chains of all eight linkage types, we reveal that most human OTU enzymes are linkage specific, preferring one, two, or a defined subset of linkage types, including unstudied atypical Ub chains. Biochemical analysis and five crystal structures of OTU DUBs with or without Ub substrates reveal four mechanisms of linkage specificity. Additional Ub-binding domains, the ubiquitinated sequence in the substrate, and defined S1' and S2 Ub-binding sites on the OTU domain enable OTU DUBs to distinguish linkage types. We introduce Ub chain restriction analysis, in which OTU DUBs are used as restriction enzymes to reveal linkage type and the relative abundance of Ub chains on substrates.


Subject(s)
Endopeptidases/chemistry , Endopeptidases/metabolism , Ovarian Neoplasms/enzymology , Ubiquitination , Catalysis , Catalytic Domain , Crystallography, X-Ray , Endopeptidases/genetics , Female , Humans , Models, Molecular , Ovarian Neoplasms/metabolism , Protein Structure, Tertiary , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/metabolism , Ubiquitins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...