Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Microbiol ; 121(3): 453-469, 2024 03.
Article in English | MEDLINE | ID: mdl-37612878

ABSTRACT

Leishmania is the causative agent of the tropical neglected disease leishmaniasis and infects macrophages as its definitive host cell. In order to sustain and propagate infections, Leishmania parasites have to complete cycles of exit and re-infection. Yet, the mechanism driving the parasite spread to other cells remains unclear. Recent studies reported pro-inflammatory monocytes as replicative niche of Leishmania major and showed prolonged expression of IL-1ß at the site of infection, indicating an activation of the NLRP3 inflammasome and pointing toward pyroptosis as a possible mechanism of parasite spread. To address the species-specific inflammasome activation of human cells, we characterized the BLaER1 monocytes as a model for L. major infection. We found that BLaER1 monocytes support infection and activation by Leishmania parasites to the same extent as primary human macrophages. Harnessing the possibilities of this infection model, we first showed that BLaER1 GSDMD-/- cells, which carry a deletion of the pore-forming protein gasdermin D, are more resistant to pyroptotic cell death and, concomitantly, display a strongly delayed release of intracellular parasite. Using that knockout in a co-incubation assay in comparison with wild-type BLaER1 cells, we demonstrate that impairment of the pyroptosis pathway leads to lower rates of parasite spread to new host cells, thus, implicating pyroptotic cell death as a possible exit mechanism of L. major in pro-inflammatory microenvironments.


Subject(s)
Inflammasomes , Leishmania , Humans , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Pyroptosis/physiology , Phosphate-Binding Proteins/metabolism , Macrophages , Leishmania/metabolism , Interleukin-1beta/metabolism
2.
EMBO Rep ; 23(12): e55839, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36268590

ABSTRACT

ZBP1 is an interferon-induced cytosolic nucleic acid sensor that facilitates antiviral responses via RIPK3. Although ZBP1-mediated programmed cell death is widely described, whether and how it promotes inflammatory signaling is unclear. Here, we report a ZBP1-induced inflammatory signaling pathway mediated by K63- and M1-linked ubiquitin chains, which depends on RIPK1 and RIPK3 as scaffolds independently of cell death. In human HT29 cells, ZBP1 associated with RIPK1 and RIPK3 as well as ubiquitin ligases cIAP1 and LUBAC. ZBP1-induced K63- and M1-linked ubiquitination of RIPK1 and ZBP1 to promote TAK1- and IKK-mediated inflammatory signaling and cytokine production. Inhibition of caspase activity suppressed ZBP1-induced cell death but enhanced cytokine production in a RIPK1- and RIPK3 kinase activity-dependent manner. Lastly, we provide evidence that ZBP1 signaling contributes to SARS-CoV-2-induced cytokine production. Taken together, we describe a ZBP1-RIPK3-RIPK1-mediated inflammatory signaling pathway relayed by the scaffolding role of RIPKs and regulated by caspases, which may induce inflammation when ZBP1 is activated below the threshold needed to trigger a cell death response.


Subject(s)
Cell Death , RNA-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases , Humans , Cytokines , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Ubiquitin , RNA-Binding Proteins/genetics , HT29 Cells , Inflammation
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34561306

ABSTRACT

The COVID-19 pandemic highlights the importance of efficient and safe vaccine development. Vaccine adjuvants are essential to boost and tailor the immune response to the corresponding pathogen. To allow for an educated selection, we assessed the effect of different adjuvants on human monocyte-derived dendritic cells (DCs) and their ability to polarize innate and adaptive immune responses. In contrast to commonly used adjuvants, such as aluminum hydroxide, Toll-like receptor (TLR) agonists induced robust phenotypic and functional DC maturation. In a DC-lymphocyte coculture system, we investigated the ensuing immune reactions. While monophosphoryl lipid A synthetic, a TLR4 ligand, induced checkpoint inhibitors indicative for immune exhaustion, the TLR7/8 agonist Resiquimod (R848) induced prominent type-1 interferon and interleukin 6 responses and robust CTL, B-cell, and NK-cell proliferation, which is particularly suited for antiviral immune responses. The recently licensed COVID-19 vaccines, BNT162b and mRNA-1273, are both based on single-stranded RNA. Indeed, we could confirm that the cytokine profile induced by lipid-complexed RNA was almost identical to the pattern induced by R848. Although this awaits further investigation, our results suggest that their efficacy involves the highly efficient antiviral response pattern stimulated by the RNAs' TLR7/8 activation.


Subject(s)
Adjuvants, Immunologic/pharmacology , COVID-19/immunology , Dendritic Cells/immunology , Immunity, Cellular/drug effects , SARS-CoV-2/immunology , T-Lymphocytes/immunology , Adolescent , Adult , Aged , Female , Humans , Imidazoles/pharmacology , Lipid A/analogs & derivatives , Lipid A/pharmacology , Male , Middle Aged , Toll-Like Receptors/immunology
4.
EMBO J ; 40(6): e106094, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33576509

ABSTRACT

The assembly of a specific polymeric ubiquitin chain on a target protein is a key event in the regulation of numerous cellular processes. Yet, the mechanisms that govern the selective synthesis of particular polyubiquitin signals remain enigmatic. The homologous ubiquitin-conjugating (E2) enzymes Ubc1 (budding yeast) and Ube2K (mammals) exclusively generate polyubiquitin linked through lysine 48 (K48). Uniquely among E2 enzymes, Ubc1 and Ube2K harbor a ubiquitin-binding UBA domain with unknown function. We found that this UBA domain preferentially interacts with ubiquitin chains linked through lysine 63 (K63). Based on structural modeling, in vitro ubiquitination experiments, and NMR studies, we propose that the UBA domain aligns Ubc1 with K63-linked polyubiquitin and facilitates the selective assembly of K48/K63-branched ubiquitin conjugates. Genetic and proteomics experiments link the activity of the UBA domain, and hence the formation of this unusual ubiquitin chain topology, to the maintenance of cellular proteostasis.


Subject(s)
Polyubiquitin/biosynthesis , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination/physiology , Computer Simulation , Models, Structural , Protein Domains , Proteomics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction/physiology , Ubiquitin-Conjugating Enzymes/genetics
5.
Science ; 370(6513): 203-208, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32817270

ABSTRACT

The spike protein (S) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is required for cell entry and is the primary focus for vaccine development. In this study, we combined cryo-electron tomography, subtomogram averaging, and molecular dynamics simulations to structurally analyze S in situ. Compared with the recombinant S, the viral S was more heavily glycosylated and occurred mostly in the closed prefusion conformation. We show that the stalk domain of S contains three hinges, giving the head unexpected orientational freedom. We propose that the hinges allow S to scan the host cell surface, shielded from antibodies by an extensive glycan coat. The structure of native S contributes to our understanding of SARS-CoV-2 infection and potentially to the development of safe vaccines.


Subject(s)
Betacoronavirus/chemistry , Molecular Dynamics Simulation , Spike Glycoprotein, Coronavirus/chemistry , Cryoelectron Microscopy , Electron Microscope Tomography , Glycosylation , Humans , Protein Domains , Protein Multimerization , SARS-CoV-2
6.
Elife ; 92020 07 02.
Article in English | MEDLINE | ID: mdl-32614325

ABSTRACT

Ubiquitin ligases (E3s) embedded in the endoplasmic reticulum (ER) membrane regulate essential cellular activities including protein quality control, calcium flux, and sterol homeostasis. At least 25 different, transmembrane domain (TMD)-containing E3s are predicted to be ER-localised, but for most their organisation and cellular roles remain poorly defined. Using a comparative proteomic workflow, we mapped over 450 protein-protein interactions for 21 stably expressed, full-length E3s. Bioinformatic analysis linked ER-E3s and their interactors to multiple homeostatic, regulatory, and metabolic pathways. Among these were four membrane-embedded interactors of RNF26, a polytopic E3 whose abundance is auto-regulated by ubiquitin-proteasome dependent degradation. RNF26 co-assembles with TMEM43, ENDOD1, TMEM33 and TMED1 to form a complex capable of modulating innate immune signalling through the cGAS-STING pathway. This RNF26 complex represents a new modulatory axis of STING and innate immune signalling at the ER membrane. Collectively, these data reveal the broad scope of regulation and differential functionalities mediated by ER-E3s for both membrane-tethered and cytoplasmic processes.


Subject(s)
Endoplasmic Reticulum/metabolism , Immunity, Innate , Protein Interaction Mapping , Protein Interaction Maps , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Proteomics
7.
Front Immunol ; 9: 1772, 2018.
Article in English | MEDLINE | ID: mdl-30108591

ABSTRACT

Tumor necrosis factor α (TNFα) drives the pathophysiology of human autoimmune diseases and consequently, neutralizing antibodies (Abs) or Ab-derived molecules directed against TNFα are essential therapeutics. As treatment with several TNFα blockers has been reported to entail a higher risk of infectious diseases such as leishmaniasis, we established an in vitro model based on Leishmania-infected human macrophages, co-cultured with autologous T-cells, for the analysis and comparison of anti-TNFα therapeutics. We demonstrate that neutralization of soluble TNFα (sTNFα) by the anti-TNFα Abs Humira®, Remicade®, and its biosimilar Remsima® negatively affects infection as treatment with these agents significantly reduces Leishmania-induced T-cell proliferation and increases the number of infected macrophages. By contrast, we show that blockade of sTNFα by Cimzia® does not affect T-cell proliferation and infection rates. Moreover, compared to Remicade®, treatment with Cimzia® does not impair the expression of cytolytic effector proteins in proliferating T-cells. Our data demonstrate that Cimzia® supports parasite control through its conjugated polyethylene glycol (PEG) moiety as PEGylation of Remicade® improves the clearance of intracellular Leishmania. This effect can be linked to complement activation, with levels of complement component C5a being increased upon treatment with Cimzia® or a PEGylated form of Remicade®. Taken together, we provide an in vitro model of human leishmaniasis that allows direct comparison of different anti-TNFα agents. Our results enhance the understanding of the efficacy and adverse effects of TNFα blockers and they contribute to evaluate anti-TNFα therapy for patients living in countries with a high prevalence of leishmaniasis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Leishmania/drug effects , Macrophages/drug effects , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adalimumab/immunology , Adalimumab/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Cells, Cultured , Certolizumab Pegol/immunology , Certolizumab Pegol/pharmacology , Coculture Techniques , Humans , Infliximab/immunology , Infliximab/pharmacology , Leishmania/immunology , Leishmania/physiology , Leishmaniasis/drug therapy , Leishmaniasis/immunology , Leishmaniasis/parasitology , Macrophages/immunology , Macrophages/parasitology , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
8.
Int J Med Microbiol ; 308(1): 228-236, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29169848

ABSTRACT

Phagocytosis is essential for uptake and elimination of pathogenic microorganisms. Autophagy is a highly conserved mechanism for incorporation of cellular constituents to replenish nutrients by degradation. Recently, parts of the autophagy machinery - above all microtubule-associated protein 1 light chain 3 (LC3) - were found to be specifically recruited to phagosomal membranes resulting in phagosome-lysosome fusion and efficient degradation of internalized cargo in a process termed LC3-associated phagocytosis (LAP). Many pathogenic bacterial, fungal and parasitic microorganisms reside within LAP-targeted single-membrane phagosomes or vacuoles after infection of host cells. In this minireview we describe the state of knowledge on the interaction of pathogens with LAP or LAP-like pathways and report on various pathogens that have evolved strategies to circumvent degradation in LAP compartments.


Subject(s)
Bacteria/pathogenicity , Fungi/pathogenicity , Microtubule-Associated Proteins/metabolism , Parasites/pathogenicity , Phagocytosis , Animals , Bacteria/immunology , Bacteria/metabolism , Fungi/immunology , Fungi/metabolism , Humans , Immune Evasion , Microtubule-Associated Proteins/immunology , Parasites/immunology , Parasites/metabolism , Phagosomes/metabolism , Phagosomes/microbiology , Phagosomes/parasitology , Vacuoles/metabolism , Vacuoles/microbiology , Vacuoles/parasitology
9.
Gut ; 66(6): 1060-1073, 2017 06.
Article in English | MEDLINE | ID: mdl-26953272

ABSTRACT

OBJECTIVE: Patients with Niemann-Pick disease type C1 (NPC1), a lysosomal lipid storage disorder that causes neurodegeneration and liver damage, can present with IBD, but neither the significance nor the functional mechanism of this association is clear. We studied bacterial handling and antibacterial autophagy in patients with NPC1. DESIGN: We characterised intestinal inflammation in 14 patients with NPC1 who developed IBD. We investigated bacterial handling and cytokine production of NPC1 monocytes or macrophages in vitro and compared NPC1-associated functional defects to those caused by IBD-associated nucleotide-binding oligomerization domain-containing protein 2 (NOD2) variants or mutations in X-linked inhibitor of apoptosis (XIAP). RESULTS: Patients with the lysosomal lipid storage disorder NPC1 have increased susceptibility to early-onset fistulising colitis with granuloma formation, reminiscent of Crohn's disease (CD). Mutations in NPC1 cause impaired autophagy due to defective autophagosome function that abolishes NOD2-mediated bacterial handling in vitro similar to variants in NOD2 or XIAP deficiency. In contrast to genetic NOD2 and XIAP variants, NPC1 mutations do not impair NOD2-receptor-interacting kinase 2 (RIPK2)-XIAP-dependent cytokine production. Pharmacological activation of autophagy can rescue bacterial clearance in macrophages in vitro by increasing the autophagic flux and bypassing defects in NPC1. CONCLUSIONS: NPC1 confers increased risk of early-onset severe CD. Our data support the concept that genetic defects at different checkpoints of selective autophagy cause a shared outcome of CD-like immunopathology linking monogenic and polygenic forms of IBD. Muramyl dipeptide-driven cytokine responses and antibacterial autophagy induction are parallel and independent signalling cascades downstream of the NOD2-RIPK2-XIAP complex.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Autophagy/genetics , Crohn Disease/genetics , Granuloma/genetics , Macrophages/drug effects , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/physiopathology , Nod2 Signaling Adaptor Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Adolescent , Adult , Anti-Bacterial Agents/pharmacology , Autophagy/drug effects , Bacteria , Cells, Cultured , Child , Child, Preschool , Chlorpromazine/pharmacology , Crohn Disease/complications , Crohn Disease/pathology , Dopamine Antagonists/pharmacology , Female , Genetic Diseases, X-Linked/genetics , Gentamicins/pharmacology , Granuloma/pathology , Humans , Imidazoles/pharmacology , Leukocytes, Mononuclear , Lysosomes , Macrophages/physiology , Male , Mutation , Niemann-Pick Disease, Type C/complications , Nod2 Signaling Adaptor Protein/metabolism , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , X-Linked Inhibitor of Apoptosis Protein/deficiency , X-Linked Inhibitor of Apoptosis Protein/metabolism , Young Adult
10.
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
11.
Mol Cell ; 62(6): 918-928, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27264873

ABSTRACT

Ubiquitin conjugation is an essential process modulating protein function in eukaryotic cells. Surprisingly, little is known about how the progressive assembly of ubiquitin chains is managed by the responsible enzymes. Only recently has ubiquitin binding activity emerged as an important factor in chain formation. The Ubc7 activator Cue1 carries a ubiquitin binding CUE domain that substantially stimulates K48-linked polyubiquitination mediated by Ubc7. Our results from NMR-based analysis and in vitro ubiquitination reactions point out that two parameters accelerate ubiquitin chain assembly: the increasing number of CUE binding sites and the position of CUE binding within a growing chain. In particular, interactions with a ubiquitin moiety adjacent to the acceptor ubiquitin facilitate chain elongation. These data indicate a mechanism for ubiquitin binding in which Cue1 positions Ubc7 and the distal acceptor ubiquitin for rapid polyubiquitination. Disrupting this mechanism results in dysfunction of the ERAD pathway by a delayed turnover of substrates.


Subject(s)
Carrier Proteins/metabolism , Endoplasmic Reticulum-Associated Degradation , Membrane Proteins/metabolism , Polyubiquitin/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , Carrier Proteins/chemistry , Carrier Proteins/genetics , Hydrophobic and Hydrophilic Interactions , Kinetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Proteolysis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Spectrometry, Fluorescence , Structure-Activity Relationship , Substrate Specificity , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics
12.
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
13.
Mol Cell ; 50(4): 528-39, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23665229

ABSTRACT

Ubiquitin-binding domains (UBDs) differentially recognize ubiquitin (ub) modifications. Some of them specifically bind mono-ub, as has been shown for the CUE domain. Interestingly, so far no significant ubiquitin binding has been observed for the CUE domain of yeast Cue1p. Cue1p is receptor and activator of the ubiquitin-conjugating enzyme Ubc7p. It integrates Ubc7p into endoplasmic reticulum (ER) membrane-bound ubiquitin ligase complexes, and thus, it is crucial for ER-associated protein degradation (ERAD). Here we show that the CUE domain of Cue1p binds ubiquitin chains, which is pivotal for the efficient formation of K48-linked polyubiquitin chains in vitro. Mutations that abolish ubiquitin binding by Cue1p affect the turnover of ERAD substrates in vivo. Our data strongly imply that the CUE domain facilitates substrate ubiquitylation by stabilizing growing ubiquitin chains at the ERAD ubiquitin ligases. Hence, we demonstrate an unexpected function of a UBD in the regulation of ubiquitin chain synthesis.


Subject(s)
Carrier Proteins/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Binding Sites/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Lysine/genetics , Lysine/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Mutation , Polyubiquitin/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Ubiquitin/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
14.
Biochim Biophys Acta ; 1808(3): 925-36, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20599420

ABSTRACT

Protein folding within the endoplasmic reticulum (ER) of eukaryotic cells is erroneous and often results in the formation of terminally malfolded species. A quality control system retards such molecules in the ER and eventually initiates their dislocation into the cytosol for proteolysis by 26S proteasomes. This process is termed ER associated protein degradation (ERAD). The spatial separation of ER based quality control and cytosolic proteolysis poses the need for a machinery that promotes the extraction of substrates from the ER. Due to the heterogeneous nature of the client proteins this transport system displays several unique features. Selective recognition of ERAD substrates does not involve transferable transport signals in the primary sequence and thus must follow other principles than established for proteins designated for the import into organelles. Moreover, an ER dislocation system must be capable to ship polypeptides, which may be at least partly folded and are in most cases covalently modified with bulky and hydrophilic glycans, through a membrane without disrupting the integrity of the ER. In this review we present current ideas on the highly dynamic and flexible nature of the dislocation apparatus and speculate on the mechanism that removes aberrant polypeptides from the ER in the course of ERAD. This article is part of a Special Issue entitled Protein translocation across or insertion into membranes.


Subject(s)
Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Animals , Humans , Protein Transport
15.
Curr Biol ; 18(21): R1019-21, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-19000801

ABSTRACT

The accumulation of misfolded cytosolic or aggregation-prone proteins leads to cellular stress. To protect the cell, damaged or aggregated proteins are actively sequestered in two newly discovered quality control compartments, JUNQ and IPOD, which are highly conserved in evolution.


Subject(s)
Cell Compartmentation , Protein Folding , Proteins/metabolism , Stress, Physiological , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...