Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
EMBO J ; 42(17): e113012, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37409490

ABSTRACT

Invasive bacteria enter the cytosol of host cells through initial uptake into bacteria-containing vacuoles (BCVs) and subsequent rupture of the BCV membrane, thereby exposing to the cytosol intraluminal, otherwise shielded danger signals such as glycans and sphingomyelin. The detection of glycans by galectin-8 triggers anti-bacterial autophagy, but how cells sense and respond to cytosolically exposed sphingomyelin remains unknown. Here, we identify TECPR1 (tectonin beta-propeller repeat containing 1) as a receptor for cytosolically exposed sphingomyelin, which recruits ATG5 into an E3 ligase complex that mediates lipid conjugation of LC3 independently of ATG16L1. TECPR1 binds sphingomyelin through its N-terminal DysF domain (N'DysF), a feature not shared by other mammalian DysF domains. Solving the crystal structure of N'DysF, we identified key residues required for the interaction, including a solvent-exposed tryptophan (W154) essential for binding to sphingomyelin-positive membranes and the conjugation of LC3 to lipids. Specificity of the ATG5/ATG12-E3 ligase responsible for the conjugation of LC3 is therefore conferred by interchangeable receptor subunits, that is, the canonical ATG16L1 and the sphingomyelin-specific TECPR1, in an arrangement reminiscent of certain multi-subunit ubiquitin E3 ligases.


Subject(s)
Microtubule-Associated Proteins , Sphingomyelins , Animals , Microtubule-Associated Proteins/metabolism , Autophagy-Related Proteins/metabolism , Carrier Proteins/metabolism , Autophagy , Ubiquitin-Protein Ligases/metabolism , Autophagy-Related Protein 5/metabolism , Mammals
2.
Science ; 379(6637): 1112-1117, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36758106

ABSTRACT

Certain inhibitor of apoptosis (IAP) family members are sentinel proteins that prevent untimely cell death by inhibiting caspases. Antagonists, including second mitochondria-derived activator of caspases (SMAC), regulate IAPs and drive cell death. Baculoviral IAP repeat-containing protein 6 (BIRC6), a giant IAP with dual E2 and E3 ubiquitin ligase activity, regulates programmed cell death through unknown mechanisms. We show that BIRC6 directly restricts executioner caspase-3 and -7 and ubiquitinates caspase-3, -7, and -9, working exclusively with noncanonical E1, UBA6. Notably, we show that SMAC suppresses both mechanisms. Cryo-electron microscopy structures of BIRC6 alone and in complex with SMAC reveal that BIRC6 is an antiparallel dimer juxtaposing the substrate-binding module against the catalytic domain. Furthermore, we discover that SMAC multisite binding to BIRC6 results in a subnanomolar affinity interaction, enabling SMAC to competitively displace caspases, thus antagonizing BIRC6 anticaspase function.


Subject(s)
Apoptosis , Caspase 3 , Caspase 7 , Caspase 9 , Inhibitor of Apoptosis Proteins , Ubiquitin-Protein Ligases , Humans , Caspase 3/metabolism , Caspase 7/metabolism , Caspase 9/metabolism , Cryoelectron Microscopy , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Catalytic Domain , Protein Multimerization
3.
Cell Rep ; 37(1): 109777, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34610306

ABSTRACT

Non-degradative ubiquitin chains and phosphorylation events govern signaling responses by innate immune receptors. The deubiquitinase CYLD in complex with SPATA2 is recruited to receptor signaling complexes by the ubiquitin ligase LUBAC and regulates Met1- and Lys63-linked polyubiquitin and receptor signaling outcomes. Here, we investigate the molecular determinants of CYLD activity. We reveal that two CAP-Gly domains in CYLD are ubiquitin-binding domains and demonstrate a requirement of CAP-Gly3 for CYLD activity and regulation of immune receptor signaling. Moreover, we identify a phosphorylation switch outside of the catalytic USP domain, which activates CYLD toward Lys63-linked polyubiquitin. The phosphorylated residue Ser568 is a novel tumor necrosis factor (TNF)-regulated phosphorylation site in CYLD and works in concert with Ser418 to enable CYLD-mediated deubiquitination and immune receptor signaling. We propose that phosphorylated CYLD, together with SPATA2 and LUBAC, functions as a ubiquitin-editing complex that balances Lys63- and Met1-linked polyubiquitin at receptor signaling complexes to promote LUBAC signaling.


Subject(s)
Deubiquitinating Enzyme CYLD/metabolism , Cell Line, Tumor , Crystallography, X-Ray , Deubiquitinating Enzyme CYLD/antagonists & inhibitors , Deubiquitinating Enzyme CYLD/genetics , Endopeptidases/chemistry , Endopeptidases/genetics , Endopeptidases/metabolism , Humans , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Phosphorylation , Polyubiquitin/metabolism , Protein Binding , Protein Domains , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin/metabolism
4.
J Cell Biol ; 219(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32356865

ABSTRACT

The Aurora B chromosomal passenger complex (CPC) is a conserved regulator of mitosis. Its functions require localization first to the chromosome arms and then centromeres in mitosis and subsequently the central spindle in anaphase. Here, we analyze the requirements for core CPC subunits, survivin and INCENP, and the mitotic kinesin-like protein 2 (MKLP2) in targeting to these distinct localizations. Centromere recruitment of the CPC requires interaction of survivin with histone H3 phosphorylated at threonine 3, and we provide a complete structure of this assembly. Furthermore, we show that the INCENP RRKKRR-motif is required for both centromeric localization of the CPC in metaphase and MKLP2-dependent transport in anaphase. MKLP2 and DNA bind competitively to this motif, and INCENP T59 phosphorylation acts as a switch preventing MKLP2 binding in metaphase. In anaphase, CPC binding promotes the microtubule-dependent ATPase activity of MKLP2. These results explain how centromere targeting of the CPC in mitosis is coupled to its movement to the central spindle in anaphase.


Subject(s)
Anaphase , Aurora Kinase B/metabolism , Chromatin/metabolism , Histones/metabolism , Kinesins/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , Aurora Kinase B/chemistry , Aurora Kinase B/genetics , Binding, Competitive , Centromere/metabolism , Centromere/ultrastructure , Chromatin/ultrastructure , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , HeLa Cells , Histones/chemistry , Histones/genetics , Humans , Kinesins/chemistry , Kinesins/genetics , Metaphase , Microtubules/metabolism , Microtubules/ultrastructure , Models, Molecular , Phosphorylation , Protein Binding , Protein Structure, Secondary , Protein Transport , Sequence Alignment , Sequence Homology, Amino Acid , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Survivin/chemistry , Survivin/genetics , Survivin/metabolism
5.
Nat Commun ; 10(1): 4320, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31541095

ABSTRACT

OTULIN (OTU Deubiquitinase With Linear Linkage Specificity) specifically hydrolyzes methionine1 (Met1)-linked ubiquitin chains conjugated by LUBAC (linear ubiquitin chain assembly complex). Here we report on the mass spectrometric identification of the OTULIN interactor SNX27 (sorting nexin 27), an adaptor of the endosomal retromer complex responsible for protein recycling to the cell surface. The C-terminal PDZ-binding motif (PDZbm) in OTULIN associates with the cargo-binding site in the PDZ domain of SNX27. By solving the structure of the OTU domain in complex with the PDZ domain, we demonstrate that a second interface contributes to the selective, high affinity interaction of OTULIN and SNX27. SNX27 does not affect OTULIN catalytic activity, OTULIN-LUBAC binding or Met1-linked ubiquitin chain homeostasis. However, via association, OTULIN antagonizes SNX27-dependent cargo loading, binding of SNX27 to the VPS26A-retromer subunit and endosome-to-plasma membrane trafficking. Thus, we define an additional, non-catalytic function of OTULIN in the regulation of SNX27-retromer assembly and recycling to the cell surface.


Subject(s)
Endopeptidases/metabolism , Endosomes/metabolism , Sorting Nexins/metabolism , Binding Sites , Crystallography, X-Ray , Endopeptidases/chemistry , Gene Knockout Techniques , Glucose Transporter Type 1/metabolism , HEK293 Cells , Humans , Models, Molecular , PDZ Domains , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Transport , Sorting Nexins/chemistry , Sorting Nexins/genetics , Ubiquitination , Vesicular Transport Proteins/metabolism
6.
EMBO Mol Med ; 11(3)2019 03.
Article in English | MEDLINE | ID: mdl-30804083

ABSTRACT

The deubiquitinase OTULIN removes methionine-1 (M1)-linked polyubiquitin signals conjugated by the linear ubiquitin chain assembly complex (LUBAC) and is critical for preventing TNF-driven inflammation in OTULIN-related autoinflammatory syndrome (ORAS). Five ORAS patients have been reported, but how dysregulated M1-linked polyubiquitin signalling causes their symptoms is unclear. Here, we report a new case of ORAS in which an OTULIN-Gly281Arg mutation leads to reduced activity and stability in vitro and in cells. In contrast to OTULIN-deficient monocytes, in which TNF signalling and NF-κB activation are increased, loss of OTULIN in patient-derived fibroblasts leads to a reduction in LUBAC levels and an impaired response to TNF Interestingly, both patient-derived fibroblasts and OTULIN-deficient monocytes are sensitised to certain types of TNF-induced death, and apoptotic cells are evident in ORAS patient skin lesions. Remarkably, haematopoietic stem cell transplantation leads to complete resolution of inflammatory symptoms, including fevers, panniculitis and diarrhoea. Therefore, haematopoietic cells are necessary for clinical manifestation of ORAS Together, our data suggest that ORAS pathogenesis involves hyper-inflammatory immune cells and TNF-induced death of both leukocytes and non-haematopoietic cells.


Subject(s)
Endopeptidases/metabolism , Inflammation/metabolism , Cell Death/genetics , Cell Death/physiology , Endopeptidases/chemistry , Endopeptidases/deficiency , Female , Fibroblasts/metabolism , Humans , Inflammation/genetics , Male , Mutation/genetics , NF-kappa B/metabolism , Protein Processing, Post-Translational , Proteomics , Signal Transduction/genetics , Signal Transduction/physiology , Ubiquitin/metabolism , Ubiquitination/genetics , Ubiquitination/physiology
7.
Methods Mol Biol ; 1844: 73-84, 2018.
Article in English | MEDLINE | ID: mdl-30242704

ABSTRACT

The availability of different polyubiquitin chains of specific linkage types has changed the appreciation of the specificity in the ubiquitin (Ub) system. Numerous E2 Ub-conjugating enzymes and E3 Ub ligases, Ub-binding domains (UBDs), and deubiquitinases (DUBs) are now known to assemble, bind, or hydrolyze individual linkage types, respectively. Biochemical and structural studies of these processes require milligram quantities of pure polyUb. Here we describe protocols that allow the enzymatic synthesis and purification of six of the eight homotypic polyUb chains through the use of chain-specific Ub ligases and DUBs.


Subject(s)
Protein Interaction Domains and Motifs , Ubiquitin/metabolism , Humans , Mutation , Polyubiquitin/chemistry , Polyubiquitin/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin/isolation & purification , Ubiquitin-Conjugating Enzymes/isolation & purification , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
8.
Cell Rep ; 23(2): 470-484, 2018 Apr 10.
Article in English | MEDLINE | ID: mdl-29642005

ABSTRACT

Tumor necrosis factor (TNF) is an inflammatory cytokine that can signal cell survival or cell death. The mechanisms that switch between these distinct outcomes remain poorly defined. Here, we show that the E3 ubiquitin ligase Mind Bomb-2 (MIB2) regulates TNF-induced cell death by inactivating RIPK1 via inhibitory ubiquitylation. Although depletion of MIB2 has little effect on NF-κB activation, it sensitizes cells to RIPK1- and caspase-8-dependent cell death. We find that MIB2 represses the cytotoxic potential of RIPK1 by ubiquitylating lysine residues in the C-terminal portion of RIPK1. Our data suggest that ubiquitin conjugation of RIPK1 interferes with RIPK1 oligomerization and RIPK1-FADD association. Disruption of MIB2-mediated ubiquitylation, either by mutation of MIB2's E3 activity or RIPK1's ubiquitin-acceptor lysines, sensitizes cells to RIPK1-mediated cell death. Together, our findings demonstrate that Mind Bomb E3 ubiquitin ligases can function as additional checkpoint of cytokine-induced cell death, selectively protecting cells from the cytotoxic effects of TNF.


Subject(s)
Apoptosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases/metabolism , Apoptosis/drug effects , Caspase 8/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Protein Multimerization/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
9.
Mol Cell ; 69(4): 566-580.e5, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29452637

ABSTRACT

Tumor necrosis factor (TNF) can drive inflammation, cell survival, and death. While ubiquitylation-, phosphorylation-, and nuclear factor κB (NF-κB)-dependent checkpoints suppress the cytotoxic potential of TNF, it remains unclear whether ubiquitylation can directly repress TNF-induced death. Here, we show that ubiquitylation regulates RIPK1's cytotoxic potential not only via activation of downstream kinases and NF-kB transcriptional responses, but also by directly repressing RIPK1 kinase activity via ubiquitin-dependent inactivation. We find that the ubiquitin-associated (UBA) domain of cellular inhibitor of apoptosis (cIAP)1 is required for optimal ubiquitin-lysine occupancy and K48 ubiquitylation of RIPK1. Independently of IKK and MK2, cIAP1-mediated and UBA-assisted ubiquitylation suppresses RIPK1 kinase auto-activation and, in addition, marks it for proteasomal degradation. In the absence of a functional UBA domain of cIAP1, more active RIPK1 kinase accumulates in response to TNF, causing RIPK1 kinase-mediated cell death and systemic inflammatory response syndrome. These results reveal a direct role for cIAP-mediated ubiquitylation in controlling RIPK1 kinase activity and preventing TNF-mediated cytotoxicity.


Subject(s)
Baculoviral IAP Repeat-Containing 3 Protein/physiology , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/physiology , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ubiquitin/metabolism , Animals , Apoptosis , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitination
10.
Org Lett ; 19(24): 6490-6493, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29172548

ABSTRACT

An optimized large scale and highly reproducible route to orthogonally protected γ-thiolysine is reported. Its utility in the synthesis of bifunctional ubiquitin monomers is demonstrated. These ubiquitin synthons are employed in polymerization reactions giving access to synthetic poly-ubiquitin chains of defined linkage.

11.
Cell Chem Biol ; 24(10): 1299-1313.e7, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-28919039

ABSTRACT

The methionine 1 (M1)-specific deubiquitinase (DUB) OTULIN acts as a negative regulator of nuclear factor κB signaling and immune homeostasis. By replacing Gly76 in distal ubiquitin (Ub) by dehydroalanine we designed the diubiquitin (diUb) activity-based probe UbG76Dha-Ub (OTULIN activity-based probe [ABP]) that couples to the catalytic site of OTULIN and thereby captures OTULIN in its active conformation. The OTULIN ABP displays high selectivity for OTULIN and does not label other M1-cleaving DUBs, including CYLD. The only detectable cross-reactivities were the labeling of USP5 (Isopeptidase T) and an ATP-dependent assembly of polyOTULIN ABP chains via Ub-activating E1 enzymes. Both cross-reactivities were abolished by the removal of the C-terminal Gly in the ABP's proximal Ub, yielding the specific OTULIN probe UbG76Dha-UbΔG76 (OTULIN ABPΔG76). Pull-downs demonstrate that substrate-bound OTULIN associates with the linear ubiquitin chain assembly complex (LUBAC). Thus, we present a highly selective ABP for OTULIN that will facilitate studying the cellular function of this essential DUB.


Subject(s)
Endopeptidases/metabolism , Molecular Probes/chemistry , Molecular Probes/metabolism , Ubiquitin/metabolism , Ubiquitination , Catalytic Domain , Endopeptidases/chemistry , HEK293 Cells , Humans , Substrate Specificity
12.
Biochem Soc Trans ; 44(6): 1581-1602, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27913667

ABSTRACT

The post-translational modification of proteins provides a rapid and versatile system for regulating all signalling pathways. Protein ubiquitination is one such type of post-translational modification involved in controlling numerous cellular processes. The unique ability of ubiquitin to form polyubiquitin chains creates a highly complex code responsible for different subsequent signalling outcomes. Specialised enzymes ('writers') generate the ubiquitin code, whereas other enzymes ('erasers') disassemble it. Importantly, the ubiquitin code is deciphered by different ubiquitin-binding proteins ('readers') functioning to elicit particular cellular responses. Ten years ago, the methionine1 (Met1)-linked (linear) polyubiquitin code was first identified and the intervening years have witnessed a seismic shift in our understanding of Met1-linked polyubiquitin in cellular processes, particularly inflammatory signalling. This review will discuss the molecular mechanisms of specificity determination within Met1-linked polyubiquitin signalling.


Subject(s)
Methionine/metabolism , Polyubiquitin/metabolism , Signal Transduction , Ubiquitinated Proteins/metabolism , Animals , Binding Sites/genetics , Endopeptidases/chemistry , Endopeptidases/genetics , Endopeptidases/metabolism , Humans , Methionine/genetics , Models, Molecular , Polyubiquitin/chemistry , Polyubiquitin/genetics , Protein Domains , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitinated Proteins/chemistry , Ubiquitinated Proteins/genetics
13.
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
14.
Mol Cell ; 63(6): 990-1005, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27591049

ABSTRACT

The linear ubiquitin chain assembly complex (LUBAC) regulates immune signaling, and its function is regulated by the deubiquitinases OTULIN and CYLD, which associate with the catalytic subunit HOIP. However, the mechanism through which CYLD interacts with HOIP is unclear. We here show that CYLD interacts with HOIP via spermatogenesis-associated protein 2 (SPATA2). SPATA2 interacts with CYLD through its non-canonical PUB domain, which binds the catalytic CYLD USP domain in a CYLD B-box-dependent manner. Significantly, SPATA2 binding activates CYLD-mediated hydrolysis of ubiquitin chains. SPATA2 also harbors a conserved PUB-interacting motif that selectively docks into the HOIP PUB domain. In cells, SPATA2 is recruited to the TNF receptor 1 signaling complex and is required for CYLD recruitment. Loss of SPATA2 increases ubiquitination of LUBAC substrates and results in enhanced NOD2 signaling. Our data reveal SPATA2 as a high-affinity binding partner of CYLD and HOIP, and a regulatory component of LUBAC-mediated NF-κB signaling.


Subject(s)
NF-kappa B/chemistry , Proteins/chemistry , Tumor Suppressor Proteins/chemistry , Ubiquitin-Protein Ligases/chemistry , Ubiquitin/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Deubiquitinating Enzyme CYLD , Endopeptidases/chemistry , Endopeptidases/genetics , Endopeptidases/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gene Expression Regulation , Humans , Immunity, Innate , Kinetics , Molecular Docking Simulation , NF-kappa B/genetics , NF-kappa B/immunology , Nod2 Signaling Adaptor Protein/chemistry , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Proteins/genetics , Proteins/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Substrate Specificity , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , Ubiquitin/genetics , Ubiquitin/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology
15.
Cell ; 166(5): 1215-1230.e20, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27523608

ABSTRACT

Methionine-1 (M1)-linked ubiquitin chains regulate the activity of NF-κB, immune homeostasis, and responses to infection. The importance of negative regulators of M1-linked chains in vivo remains poorly understood. Here, we show that the M1-specific deubiquitinase OTULIN is essential for preventing TNF-associated systemic inflammation in humans and mice. A homozygous hypomorphic mutation in human OTULIN causes a potentially fatal autoinflammatory condition termed OTULIN-related autoinflammatory syndrome (ORAS). Four independent OTULIN mouse models reveal that OTULIN deficiency in immune cells results in cell-type-specific effects, ranging from over-production of inflammatory cytokines and autoimmunity due to accumulation of M1-linked polyubiquitin and spontaneous NF-κB activation in myeloid cells to downregulation of M1-polyubiquitin signaling by degradation of LUBAC in B and T cells. Remarkably, treatment with anti-TNF neutralizing antibodies ameliorates inflammation in ORAS patients and rescues mouse phenotypes. Hence, OTULIN is critical for restraining life-threatening spontaneous inflammation and maintaining immune homeostasis.


Subject(s)
Autoimmune Diseases/genetics , Autoimmunity/genetics , Deubiquitinating Enzymes/metabolism , Endopeptidases/metabolism , Inflammation/genetics , Animals , Antibodies, Neutralizing/therapeutic use , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , B-Lymphocytes/immunology , Cytokines/metabolism , Deubiquitinating Enzymes/genetics , Disease Models, Animal , Endopeptidases/genetics , Germ-Line Mutation , Humans , Inflammation/immunology , Inflammation/therapy , Infliximab/therapeutic use , Methionine/metabolism , Mice , Mice, Mutant Strains , Myeloid Cells/immunology , Polyubiquitin/metabolism , Sequence Deletion , Syndrome , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
16.
Cell Rep ; 14(12): 2846-58, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26997266

ABSTRACT

Innate immune signaling relies on the deposition of non-degradative polyubiquitin at receptor-signaling complexes, but how these ubiquitin modifications are regulated by deubiquitinases remains incompletely understood. Met1-linked ubiquitin (Met1-Ub) is assembled by the linear ubiquitin assembly complex (LUBAC), and this is counteracted by the Met1-Ub-specific deubiquitinase OTULIN, which binds to the catalytic LUBAC subunit HOIP. In this study, we report that HOIP also interacts with the deubiquitinase CYLD but that CYLD does not regulate ubiquitination of LUBAC components. Instead, CYLD limits extension of Lys63-Ub and Met1-Ub conjugated to RIPK2 to restrict signaling and cytokine production. Accordingly, Met1-Ub and Lys63-Ub were individually required for productive NOD2 signaling. Our study thus suggests that LUBAC, through its associated deubiquitinases, coordinates the deposition of not only Met1-Ub but also Lys63-Ub to ensure an appropriate response to innate immune receptor activation.


Subject(s)
Deubiquitinating Enzymes/metabolism , Immunity, Innate , Lysine/metabolism , Methionine/metabolism , Signal Transduction , Ubiquitin/metabolism , Catalytic Domain , Cell Line, Tumor , Cytokines/metabolism , Deubiquitinating Enzymes/antagonists & inhibitors , Deubiquitinating Enzymes/genetics , Endopeptidases/chemistry , Endopeptidases/genetics , Endopeptidases/metabolism , HEK293 Cells , Humans , Lysine/chemistry , Methionine/chemistry , Mutagenesis, Site-Directed , NF-kappa B/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
17.
Chembiochem ; 17(9): 816-20, 2016 05 03.
Article in English | MEDLINE | ID: mdl-26996281

ABSTRACT

Deubiquitinating enzymes (DUBs) are proteases that fulfill crucial roles in the ubiquitin (Ub) system, by deconjugation of Ub from its targets and disassembly of polyUb chains. The specificity of a DUB towards one of the polyUb chain linkages largely determines the ultimate signaling function. We present a novel set of diubiquitin FRET probes, comprising all seven isopeptide linkages, for the absolute quantification of chain cleavage specificity of DUBs by means of Michaelis-Menten kinetics. Each probe is equipped with a FRET pair consisting of Rhodamine110 and tetramethylrhodamine to allow the fully synthetic preparation of the probes by SPPS and NCL. Our synthetic strategy includes the introduction of N,N'-Boc-protected 5-carboxyrhodamine as a convenient building block in peptide chemistry. We demonstrate the value of our probes by quantifying the linkage specificities of a panel of nine DUBs in a high-throughput manner.


Subject(s)
Deubiquitinating Enzymes/metabolism , Ubiquitin/metabolism , Chromatography, High Pressure Liquid , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Kinetics , Ubiquitination
18.
FEBS J ; 283(1): 39-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26503766

ABSTRACT

Modification of proteins with Met1-linked 'linear' ubiquitin chains has emerged as a key regulatory signal to control inflammatory signalling via the master regulator, the transcription factor nuclear factor κB (NF-κB). While the assembly machinery, the linear ubiquitin chain assembly complex (LUBAC), and receptors for this ubiquitin chain type have been known for years, it was less clear which deubiquitinating enzymes (DUBs) hydrolyse Met1 linkages specifically. In 2013, two labs reported the previously unannotated protein FAM105B/OTULIN to be this missing Met1 linkage-specific DUB. Structural studies have revealed how OTULIN achieves its remarkable specificity, employing a mechanism of ubiquitin-assisted catalysis in which a glutamate residue on the substrate complements the active site of the enzyme. The specificity of OTULIN enables it to regulate global levels of Met1-linked polyubiquitin in cells. This ability led to investigations of NF-κB activation from new angles, and also revealed involvement of Met1-polyubiquitin in Wnt signalling. Interestingly, OTULIN directly interacts with LUBAC, and this interaction is dynamic and can be regulated by OTULIN phosphorylation. This provides a new paradigm for how individual linkage types can be regulated by dedicated enzyme complexes mediating assembly and removal. Here we review what has been learned about OTULIN's mechanism, regulation and function, discuss the open questions in the field, and discuss how DUBs regulate the NF-κB response.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Endopeptidases/metabolism , Polyubiquitin/metabolism , Signal Transduction , Animals , Humans
19.
Sci Rep ; 5: 16826, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26585104

ABSTRACT

This paper proposes a mechanism of controlling the diameter and separation of metallic nanorods from physical vapor deposition through self-organized seeds and experimentally demonstrates the feasibility using Ag as the prototype metal, In as the seed, and Si the substrate. Being non-wetting on Si substrates, deposited In atoms self-organize into islands. Subsequently deposited Ag atoms attach to In islands, rather than to Si substrates, due to preferential bonding and geometrical shadowing. The experimental results show that self-organized In seeds of 5 nm nominal thickness give rise to the best separation and the smallest diameter of Ag nanorods.

20.
Sci Rep ; 5: 9260, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25851026

ABSTRACT

This paper reports combined hydrophobicity and mechanical durability through the nanoscale engineering of surfaces in the form of nanorod-polymer composites. Specifically, the hydrophobicity derives from nanoscale features of mechanically hard ZnO nanorods and the mechanical durability derives from the composite structure of a hard ZnO nanorod core and soft polymer shell. Experimental characterization correlates the morphology of the nanoengineered surfaces with the combined hydrophobicity and mechanical durability, and reveals the responsible mechanisms. Such surfaces may find use in applications, such as boat hulls, that benefit from hydrophobicity and require mechanical durability.

SELECTION OF CITATIONS
SEARCH DETAIL
...