37 results on '"Oded Kleifeld"'
Search Results
2. Isolation and infection cycle of a polinton-like virus virophage in an abundant marine alga
- Author
-
Sheila Roitman, Andrey Rozenberg, Tali Lavy, Corina P. D. Brussaard, Oded Kleifeld, and Oded Béjà
- Subjects
Microbiology (medical) ,Immunology ,Genetics ,Cell Biology ,Applied Microbiology and Biotechnology ,Microbiology - Abstract
Virophages are small double stranded DNA (dsDNA) viruses that can only replicate in a host by co-infecting with another virus. Marine algae are commonly associated with virophage-like elements such as Polinton-like viruses (PLVs) that remain largely uncharacterized. Here we isolated a PLV that co-infects the alga Phaeocystis globosa with the Phaeocystis globosa virus-14T (PgV-14T), a close relative of the "Phaeocystis globosa virus-virophage" genomic sequence. We name this PLV ‘Gezel-14T. Gezel is phylogenetically distinct from the Lavidaviridae family where all known virophages belong. Gezel-14T co-infection decreases the fitness of its viral host by reducing burst sizes of PgV-14T, yet insufficiently to spare the cellular host population. Genomic screens show Gezel-14T-like PLVs integrated into Phaeocystis genomes, suggesting that these widespread viruses are capable of integration into cellular host genomes. This system presents an opportunity to better understand the evolution of eukaryotic dsDNA viruses as well as the complex dynamics and implications of viral parasitism.
- Published
- 2023
3. Phosphoproteomic characterization of the signaling network resulting from activation of the chemokine receptor CCR2
- Author
-
Martin J. Stone, Anup Shah, Oded Kleifeld, Ralf B. Schittenhelm, Meritxell Canals, Cheng Huang, and Simon R. Foster
- Subjects
Proteomics ,0301 basic medicine ,CCR2 ,Chemokine ,Cell signaling ,Receptors, CCR2 ,Biochemistry ,03 medical and health sciences ,Chemokine receptor ,Humans ,Phosphorylation ,Molecular Biology ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Phosphoproteomics ,Cell Biology ,Phosphoproteins ,Actin cytoskeleton ,Cell biology ,Gene Ontology ,HEK293 Cells ,030104 developmental biology ,biology.protein ,Signal transduction ,CC chemokine receptors ,Signal Transduction - Abstract
Leukocyte recruitment is a universal feature of tissue inflammation and regulated by the interactions of chemokines with their G protein–coupled receptors. Activation of CC chemokine receptor 2 (CCR2) by its cognate chemokine ligands, including CC chemokine ligand 2 (CCL2), plays a central role in recruitment of monocytes in several inflammatory diseases. In this study, we used phosphoproteomics to conduct an unbiased characterization of the signaling network resulting from CCL2 activation of CCR2. Using data-independent acquisition MS analysis, we quantified both the proteome and phosphoproteome in FlpIn-HEK293T cells stably expressing CCR2 at six time points after activation with CCL2. Differential expression analysis identified 699 significantly regulated phosphorylation sites on 441 proteins. As expected, many of these proteins are known to participate in canonical signal transduction pathways and in the regulation of actin cytoskeleton dynamics, including numerous guanine nucleotide exchange factors and GTPase-activating proteins. Moreover, we identified regulated phosphorylation sites in numerous proteins that function in the nucleus, including several constituents of the nuclear pore complex. The results of this study provide an unprecedented level of detail of CCR2 signaling and identify potential targets for regulation of CCR2 function.
- Published
- 2020
4. The 20S as a stand-alone proteasome in cells can degrade the ubiquitin tag
- Author
-
Cong Xu, Indrajit Sahu, Andrey Rozenberg, Roni Morag, Sumeet K. Singh, Yao Cong, Manisha Priyadarsini Sahoo, Sachitanand M. Mali, Michael H. Glickman, Zhanyu Ding, Sharleen Day, Prasad Sulkshane, Ashraf Brik, Oded Kleifeld, and Yifan Wang
- Subjects
Proteasome Endopeptidase Complex ,Ubiquitylation ,Cell Survival ,Protein Conformation ,Science ,Proteolysis ,medicine.medical_treatment ,General Physics and Astronomy ,Peptide ,General Biochemistry, Genetics and Molecular Biology ,Article ,Substrate Specificity ,Ubiquitylated proteins ,Ubiquitin ,Cryoelectron microscopy ,medicine ,Humans ,30S ,chemistry.chemical_classification ,Heart Failure ,Multidisciplinary ,Protease ,biology ,medicine.diagnostic_test ,Proteasome ,Chemistry ,Ubiquitination ,Substrate (chemistry) ,General Chemistry ,Ubiquitinated Proteins ,Cell Hypoxia ,Cell biology ,biology.protein ,Peptides ,Intracellular - Abstract
The proteasome, the primary protease for ubiquitin-dependent proteolysis in eukaryotes, is usually found as a mixture of 30S, 26S, and 20S complexes. These complexes have common catalytic sites, which makes it challenging to determine their distinctive roles in intracellular proteolysis. Here, we chemically synthesize a panel of homogenous ubiquitinated proteins, and use them to compare 20S and 26S proteasomes with respect to substrate selection and peptide-product generation. We show that 20S proteasomes can degrade the ubiquitin tag along with the conjugated substrate. Ubiquitin remnants on branched peptide products identified by LC-MS/MS, and flexibility in the 20S gate observed by cryo-EM, reflect the ability of the 20S proteasome to proteolyze an isopeptide-linked ubiquitin-conjugate. Peptidomics identifies proteasome-trapped ubiquitin-derived peptides and peptides of potential 20S substrates in Hi20S cells, hypoxic cells, and human failing-heart. Moreover, elevated levels of 20S proteasomes appear to contribute to cell survival under stress associated with damaged proteins., The 20S particle is part of the 26S proteasome, but also exists as a free complex. Here, the authors outline signature activities of the 20S and combine chemical, structural, functional and proteomic assays to show that the 20S can degrade ubiquitin tags along with conjugated substrates.
- Published
- 2021
- Full Text
- View/download PDF
5. Ubiquitination and receptor-mediated mitophagy converge to eliminate oxidation-damaged mitochondria during hypoxia
- Author
-
Oded Kleifeld, Prasad Sulkshane, Michael H. Glickman, Anita Thakur, Noa Reis, and Jonathan Ram
- Subjects
0301 basic medicine ,Medicine (General) ,PolyUb, polyubiquitin ,QH301-705.5 ,Clinical Biochemistry ,HIF-1α ,Mitochondrion ,Biochemistry ,Parkin ,03 medical and health sciences ,0302 clinical medicine ,R5-920 ,Ubiquitin ,Mitophagy ,MFN1 ,Humans ,OMM, Outer Mitochondrial Membrane ,OCR, oxygen consumption rate ,NAC, N-Acetyl Cysteine ,Biology (General) ,Hypoxia ,OXPHOS, Oxidative Phosphorylation ,MAD, Mitochondria-Associated Degradation ,biology ,Proteasome ,Chemistry ,Organic Chemistry ,Autophagy ,Ubiquitination ,Ub, Ubiquitin ,Ubiquitin ligase ,Cell biology ,Mitochondria ,Oxidative Stress ,030104 developmental biology ,biology.protein ,UPS, Ubiquitin Proteasome System ,PHD, Prolyl Hydroxylase ,MitoQ, Mitoquinone ,030217 neurology & neurosurgery ,Research Paper ,ROS, Reactive Oxygen Species ,HeLa Cells - Abstract
The contribution of the Ubiquitin-Proteasome System (UPS) to mitophagy has been largely attributed to the E3 ubiquitin ligase Parkin. Here we show that in response to the oxidative stress associated with hypoxia or the hypoxia mimic CoCl2, the damaged and fragmented mitochondria are removed by Parkin-independent mitophagy. Mitochondria isolated from hypoxia or CoCl2-treated cells exhibited extensive ubiquitination, predominantly Lysine 48-linked and involves the degradation of key mitochondrial proteins such as the mitofusins MFN1/2, or the import channel component TOM20. Reflecting the critical role of mitochondrial protein degradation, proteasome inhibition blocked CoCl2-induced mitophagy. The five conserved ubiquitin-binding autophagy receptors (p62, NDP52, Optineurin, NBR1, TAX1BP1) were dispensable for the ensuing mitophagy, suggesting that the mitophagy step itself was independent of ubiquitination. Instead, the expression of two ubiquitin-independent mitophagy receptor proteins BNIP3 and NIX was induced by hypoxia or CoCl2-treatment followed by their recruitment to the oxidation-damaged mitochondria. By employing BNIP3/NIX double knockout and DRP1-null cell lines, we confirmed that mitochondrial clearance relies on DRP1-dependent mitochondrial fragmentation and BNIP3/NIX-mediated mitophagy. General antioxidants such as N-Acetyl Cysteine (NAC) or the mitochondria-specific Mitoquinone prevented HIF-1α stabilization, ameliorated hypoxia-related mitochondrial oxidative stress, and suppressed mitophagy. We conclude that the UPS and receptor-mediated autophagy converge to eliminate oxidation-damaged mitochondria., Graphical abstract Sulkshane P. et al. show that oxidation-induced mitophagy entails two steps: degradation of outer mitochondrial membrane proteins in a ubiquitin-proteasome-dependent manner followed by ubiquitin-independent mitophagy.Image 1, Highlights • Mitochondria-derived ROS contributes to HIF-1α stabilization during hypoxia. • Oxidation-induced mitophagy entails ubiquitin-dependent and -independent steps. • PINK1/Parkin & Ub-binding receptors are dispensable for oxidation-induced mitophagy. • DRP1-dependent fragmentation facilitates oxidation-induced mitophagy. • BNIP3/NIX partake in hypoxia-induced mitophagy, independent of mitochondrial ubiquitination.
- Published
- 2021
6. TRIM25 and DEAD-Box RNA Helicase DDX3X Cooperate to Regulate RIG-I-Mediated Antiviral Immunity
- Author
-
Steven M. Heaton, Oded Kleifeld, Michelle D Audsley, Natalie A. Borg, and Sarah C. Atkinson
- Subjects
viruses ,NS1 ,DEAD-box RNA Helicases ,Tripartite Motif Proteins ,Ubiquitin ,Interferon ,Receptors, Immunologic ,Biology (General) ,Promoter Regions, Genetic ,Spectroscopy ,E3 ligase ,0303 health sciences ,RIG-I ,030302 biochemistry & molecular biology ,General Medicine ,RNA Helicase A ,3. Good health ,Computer Science Applications ,Cell biology ,Ubiquitin ligase ,antiviral immunity ,RLR signalling ,Chemistry ,Influenza A virus ,DEAD Box Protein 58 ,DDX3X ,influenza ,Protein Binding ,Signal Transduction ,medicine.drug ,TRIM25 ,DEAD-box helicase ,DEAD box ,QH301-705.5 ,Ubiquitin-Protein Ligases ,Biology ,ubiquitination ,IFN ,Antiviral Agents ,Article ,Catalysis ,Cell Line ,Inorganic Chemistry ,03 medical and health sciences ,medicine ,Humans ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,030304 developmental biology ,Organic Chemistry ,Immunity ,RNA virus ,biology.organism_classification ,HEK293 Cells ,Gene Expression Regulation ,biology.protein ,Interferons ,Transcription Factors - Abstract
The cytoplasmic retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) initiate interferon (IFN) production and antiviral gene expression in response to RNA virus infection. Consequently, RLR signalling is tightly regulated by both host and viral factors. Tripartite motif protein 25 (TRIM25) is an E3 ligase that ubiquitinates multiple substrates within the RLR signalling cascade, playing both ubiquitination-dependent and -independent roles in RIG-I-mediated IFN induction. However, additional regulatory roles are emerging. Here, we show a novel interaction between TRIM25 and another protein in the RLR pathway that is essential for type I IFN induction, DEAD-box helicase 3X (DDX3X). In vitro assays and knockdown studies reveal that TRIM25 ubiquitinates DDX3X at lysine 55 (K55) and that TRIM25 and DDX3X cooperatively enhance IFNB1 induction following RIG-I activation, but the latter is independent of TRIM25’s catalytic activity. Furthermore, we found that the influenza A virus non-structural protein 1 (NS1) disrupts the TRIM25:DDX3X interaction, abrogating both TRIM25-mediated ubiquitination of DDX3X and cooperative activation of the IFNB1 promoter. Thus, our results reveal a new interplay between two RLR-host proteins that cooperatively enhance IFN-β production. We also uncover a new and further mechanism by which influenza A virus NS1 suppresses host antiviral defence.
- Published
- 2021
- Full Text
- View/download PDF
7. Activity-Based Probes Developed by Applying a Sequential Dehydroalanine Formation Strategy to Expressed Proteins Reveal a Potential α-Globin-Modulating Deubiquitinase
- Author
-
Ashraf Brik, Oded Kleifeld, Sachitanand M. Mali, and Roman Meledin
- Subjects
Alanine ,Deubiquitinating Enzymes ,Molecular Structure ,biology ,010405 organic chemistry ,Chemistry ,General Medicine ,General Chemistry ,010402 general chemistry ,01 natural sciences ,Catalysis ,α globin ,0104 chemical sciences ,Cell biology ,Deubiquitinating enzyme ,chemistry.chemical_compound ,alpha-Globins ,Ubiquitin ,Dehydroalanine ,Molecular Probes ,hemic and lymphatic diseases ,biology.protein ,Humans ,Deubiquitination - Abstract
We report a general and novel semisynthetic strategy for the preparation of ubiquitinated protein-activity-based probes on the basis of sequential dehydroalanine formation on expressed proteins. We applied this approach to construct a physiologically and therapeutically relevant ubiquitinated α-globin probe, which was used for the enrichment and proteomic identification of α-globin-modulating deubiquitinases. We found USP15 as a potential deubiquitinase for the modulation of α-globin, an excess of which aggravates β-thalassemia symptoms. This development opens new opportunities for activity-based-probe design to shed light on the important aspects underlying ubiquitination and deubiquitination in health and disease.
- Published
- 2018
8. Proteomic Identification of Interferon-Induced Proteins with Tetratricopeptide Repeats as Markers of M1 Macrophage Polarization
- Author
-
Martin J. Stone, Mingyu Zhu, Caitlin Lewis, Cheng Huang, Grant R Drummond, Henry Diep, Natalie A. Borg, Barbara K Kemp-Harper, Antony Vinh, Robert J. A. Goode, Meritxell Canals, Oded Kleifeld, and Ralf B. Schittenhelm
- Subjects
Proteomics ,0301 basic medicine ,THP-1 Cells ,Macrophage polarization ,Biology ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Interferon ,medicine ,Animals ,Humans ,Tetratricopeptide Repeat ,Macrophage ,Inflammation ,Mice, Knockout ,Macrophages ,Interferon-stimulated gene ,Proteins ,General Chemistry ,Atherosclerosis ,Up-Regulation ,3. Good health ,Cell biology ,Tetratricopeptide ,030104 developmental biology ,IRF1 ,Proteome ,Biomarkers ,Interferon Regulatory Factor-1 ,030215 immunology ,medicine.drug - Abstract
Macrophages, which accumulate in tissues during inflammation, may be polarized toward pro-inflammatory (M1) or tissue reparative (M2) phenotypes. The balance between these phenotypes can have a substantial influence on the outcome of inflammatory diseases such as atherosclerosis. Improved biomarkers of M1 and M2 macrophages would be beneficial for research, diagnosis, and monitoring the effects of trial therapeutics in such diseases. To identify novel biomarkers, we have characterized the global proteomes of THP-1 macrophages polarized to M1 and M2 states in comparison with unpolarized (M0) macrophages. M1 polarization resulted in increased expression of numerous pro-inflammatory proteins including the products of 31 genes under the transcriptional control of interferon regulatory factor 1 (IRF-1). In contrast, M2 polarization identified proteins regulated by components of the transcription factor AP-1. Among the most highly upregulated proteins under M1 conditions were the three interferon-induced proteins with tetratricopeptide repeats (IFITs: IFIT1, IFIT2, and IFIT3), which function in antiviral defense. Moreover, IFIT1, IFIT2, and IFIT3 mRNA were strongly upregulated in M1 polarized human primary macrophages and IFIT1 was also expressed in a subset of macrophages in aortic sinus and brachiocephalic artery sections from atherosclerotic ApoE
- Published
- 2018
9. Dynamic structure and localization of G protein-coupled receptor (GPCR) complexes determines unique signalling outcomes
- Author
-
Arisbel B. Gondin, Srgjan Chivchiristov, Michelle L. Halls, Oded Kleifeld, Ghizal Siddiqui, Meritxell Canals, and Darren J. Creek
- Subjects
Signalling ,Chemistry ,Applied Mathematics ,General Mathematics ,Cell biology ,G protein-coupled receptor - Published
- 2018
10. A novel recognition site for polyubiquitin and ubiquitin-like signals in an unexpected region of proteasomal subunit Rpn1
- Author
-
David Fushman, Andrew J. Boughton, Tali Lavy, Oded Kleifeld, and Leonard Liu
- Subjects
Ubp6 ,Amino Acid Motifs ,PRE, paramagnetic relaxation enhancement ,Protein Data Bank (RCSB PDB) ,K48-linked polyubiquitin ,Biochemistry ,chemistry.chemical_compound ,Ubiquitin ,MTSL, (1-oxyl-2,2,5,5-tetramethyl-3-pyrroline-3-methyl) methanesulfonate ,Polyubiquitin ,Ub, ubiquitin ,biology ,Chemistry ,K11-linked polyubiquitin ,computer.file_format ,Cell biology ,UBA, Ub-associated ,RP, regulatory particle ,PC, proteasome/cyclosome ,GuHCl, guanidinium chloride ,Research Article ,UPS, ubiquitin–proteasome system ,Proteasome Endopeptidase Complex ,NT, N-terminal to Toroid ,Saccharomyces cerevisiae Proteins ,TCEP, tris(2-carboxyethyl)phosphine ,MTSL ,Protein subunit ,Saccharomyces cerevisiae ,Rad23 ,Rpn1 ,PDB, Protein Data Bank ,ubiquitin ,Bpa, p-benzoyl-l-phenylalanine ,Rpn1214–355, Rpn1 encompassing residues 214 to 355 ,UBL, Ub-like ,Binding site ,Molecular Biology ,Mutagenesis ,Cell Biology ,CP, core particle ,Protein Data Bank ,Dsk2 ,CSP, chemical shift perturbation ,polyUb, polymeric Ub ,proteasome ,Proteasome ,biology.protein ,UBL domain ,computer - Abstract
The ubiquitin (Ub)-proteasome system is the primary mechanism for maintaining protein homeostasis in eukaryotes, yet the underlying signaling events and specificities of its components are poorly understood. Proteins destined for degradation are tagged with covalently linked polymeric Ub chains and subsequently delivered to the proteasome, often with the assistance of shuttle proteins that contain Ub-like domains. This degradation pathway is riddled with apparent redundancy-in the form of numerous polyubiquitin chains of various lengths and distinct architectures, multiple shuttle proteins, and at least three proteasomal receptors. Moreover, the largest proteasomal receptor, Rpn1, contains one known binding site for polyubiquitin and shuttle proteins, although several studies have recently proposed the existence of an additional uncharacterized site. Here, using a combination of NMR spectroscopy, photocrosslinking, mass spectrometry, and mutagenesis, we show that Rpn1 does indeed contain another recognition site that exhibits affinities and binding preferences for polyubiquitin and Ub-like signals comparable to those of the known binding site in Rpn1. Surprisingly, this novel site is situated in the N-terminal section of Rpn1, a region previously surmised to be devoid of functionality. We identified a stretch of adjacent helices as the location of this previously uncharacterized binding site, whose spatial proximity and similar properties to the known binding site in Rpn1 suggest the possibility of multivalent signal recognition across the solvent-exposed surface of Rpn1. These findings offer new mechanistic insights into signal recognition processes that are at the core of the Ub-proteasome system.
- Published
- 2021
11. Synthetic Uncleavable Ubiquitinated Proteins Dissect Proteasome Deubiquitination and Degradation, and Highlight Distinctive Fate of Tetraubiquitin
- Author
-
Sumeet K. Singh, Ashraf Brik, Michael H. Glickman, Oded Kleifeld, Sachitanand M. Mali, Hosahalli P. Hemantha, and Indrajit Sahu
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Proteasome Endopeptidase Complex ,Isopeptide bond ,Deubiquitinating Enzymes ,Molecular Structure ,Ubiquitin ,Chemistry ,Substrate (chemistry) ,General Chemistry ,Cleavage (embryo) ,Ubiquitinated Proteins ,Biochemistry ,Catalysis ,Cell biology ,03 medical and health sciences ,Chain length ,030104 developmental biology ,Colloid and Surface Chemistry ,Proteasome ,Humans ,Degradation (geology) ,Deubiquitination - Abstract
Various hypotheses have been proposed regarding how chain length, linkage type, position on substrate, and susceptibility to deubiquitinases (DUBs) affect processing of different substrates by proteasome. Here we report a new strategy for the chemical synthesis of ubiquitinated proteins to generate a set of well-defined conjugates bearing an oxime bond between the chain and the substrate. We confirmed that this isopeptide replacement is resistant to DUBs and to shaving by proteasome. Analyzing products generated by proteasomes ranked how chain length governed degradation outcome. Our results support that (1) the cleavage of the proximal isopeptide bond is not a prerequisite for proteasomal degradation, (2) by overcoming trimming at the proteasome, tetraUb is a fundamentally different signal than shorter chains, and (3) the tetra-ubiquitin chain can be degraded with the substrate. Together these results highlight the usefulness of chemistry to dissect the contribution of proteasome-associated DUBs and the complexity of the degradation process.
- Published
- 2016
12. Remodeling Membrane Binding by Mono-Ubiquitylation
- Author
-
Neta Tanner, Iftach Nachman, Gali Prag, and Oded Kleifeld
- Subjects
Models, Molecular ,phosphatidylinositol phosphate ,Epsin ,Saccharomyces cerevisiae Proteins ,lcsh:QR1-502 ,Vesicular Transport Proteins ,Endocytosis ,Biochemistry ,lcsh:Microbiology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Protein Domains ,Live cell imaging ,Receptor ,ubiquitylation ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,biology ,Chemistry ,Cell Membrane ,Ubiquitination ,In vitro ,Cell biology ,Membrane protein ,biology.protein ,Ub receptor ,030217 neurology & neurosurgery ,Function (biology) ,Protein Binding - Abstract
Ubiquitin (Ub) receptors respond to ubiquitylation signals. They bind ubiquitylated substrates and exert their activity in situ. Intriguingly, Ub receptors themselves undergo rapid ubiquitylation and deubiquitylation. Here we asked what is the function of ubiquitylation of Ub receptors? We focused on yeast epsin, a Ub receptor that decodes the ubiquitylation signal of plasma membrane proteins into an endocytosis response. Using mass spectrometry, we identified lysine-3 as the major ubiquitylation site in the epsin plasma membrane binding domain. By projecting this ubiquitylation site onto our crystal structure, we hypothesized that this modification would compete with phosphatidylinositol-4,5-bisphosphate (PIP2) binding and dissociate epsin from the membrane. Using an E. coli-based expression of an authentic ubiquitylation apparatus, we purified ubiquitylated epsin. We demonstrated in vitro that in contrast to apo epsin, the ubiquitylated epsin does not bind to either immobilized PIPs or PIP2-enriched liposomes. To test this hypothesis in vivo, we mimicked ubiquitylation by the fusion of Ub at the ubiquitylation site. Live cell imaging demonstrated that the mimicked ubiquitylated epsin dissociates from the membrane. Our findings suggest that ubiquitylation of the Ub receptors dissociates them from their products to allow binding to a new ubiquitylated substrates, consequently promoting cyclic activity of the Ub receptors.
- Published
- 2019
- Full Text
- View/download PDF
13. Structural Insight into Regulation of the Proteasome Ub-Receptor Rpn10
- Author
-
Ilan Attali, Gali Prag, Oded Kleifeld, Olga Levin-Kravets, Tal Keren-Kaplan, and Shay Ben-Aroya
- Subjects
0303 health sciences ,03 medical and health sciences ,0302 clinical medicine ,Proteasome ,Chemistry ,InformationSystems_INFORMATIONSTORAGEANDRETRIEVAL ,Receptor ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,030217 neurology & neurosurgery ,030304 developmental biology ,Cell biology - Published
- 2019
14. Signature activities of 20S proteasome include degradation of the ubiquitin-tag with the protein under hypoxia
- Author
-
Zhanyu Ding, Cong Xu, Indrajit Sahu, Sumeet K. Singh, Andrey Rozenberg, Manisha Priyadarsini Sahoo, Yifan Wang, Prasad Sulkshane, Sachitanand M. Mali, Roni Morag, Yao Cong, Michael H. Glickman, Sharleen Day, Ashraf Brik, and Oded Kleifeld
- Subjects
0303 health sciences ,Protease ,biology ,medicine.diagnostic_test ,Chemistry ,Proteolysis ,medicine.medical_treatment ,Substrate (chemistry) ,Protein degradation ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,biology.protein ,medicine ,Target protein ,030217 neurology & neurosurgery ,Function (biology) ,Intracellular ,030304 developmental biology - Abstract
Careful removal of unwanted proteins is necessary for cell survival. The primary constitutive intracellular protease is the 26S proteasome complex, often found in equilibrium with its free catalytic subcomplex– the 20S core particle. Protein degradation by 26S is tightly regulated by prior ubiquitination of substrates, whereas 20S is amenable to substrates with an unstructured segment. Differentiating their contributions to intracellular proteolysis is challenging due to their common catalytic sites. Here, by chemically synthesizing a synoptic set of homogenous ubiquitinated proteins, we ascribe signature features to 20S function and demonstrate a unique property: degrading the ubiquitin-tag along with the target protein. Cryo-EM confirms that a ubiquitinated substrate can induce asymmetric conformational changes to 20S. Mass-spectrometry of intracellular peptidome under hypoxia and in human failing heart identifies the signature properties of 20S in cells. Moreover, the ability of 20S proteasome to clear toxic proteins rapidly, contributes to better survival under these conditions.
- Published
- 2019
- Full Text
- View/download PDF
15. Integration of Two In-depth Quantitative Proteomics Approaches Determines the Kallikrein-related Peptidase 7 (KLK7) Degradome in Ovarian Cancer Cell Secretome
- Author
-
Thomas Stoll, Lakmali Munasinghage Silva, Christine Hoogland, Marcus L. Hastie, Judith A. Clements, Viktor Magdolen, Oded Kleifeld, Thomas Kryza, Jeffrey J. Gorman, Ying Dong, and Carson R. Stephens
- Subjects
Proteomics ,Proteome ,Quantitative proteomics ,Biochemistry ,Analytical Chemistry ,Substrate Specificity ,Thrombospondin 1 ,03 medical and health sciences ,Matrix Metalloproteinase 10 ,Stable isotope labeling by amino acids in cell culture ,Cell Line, Tumor ,KLK7 ,medicine ,Chymotrypsin ,Humans ,Amino Acid Sequence ,Cell adhesion ,Molecular Biology ,030304 developmental biology ,Ovarian Neoplasms ,0303 health sciences ,Chemistry ,Hydrolysis ,Research ,030302 biochemistry & molecular biology ,Cancer ,Terminal amine isotopic labeling of substrates ,medicine.disease ,Cell biology ,Enzyme Activation ,Gene Ontology ,Culture Media, Conditioned ,Proteolysis ,Female ,Kallikreins ,Benjamin Cravatt III ,Peptides - Abstract
Kallikrein-related peptidase 7 (KLK7) is a serine peptidase that is over expressed in ovarian cancer. In vitro functional analyses have suggested KLK7 to play a cancer progressive role, although monitoring of KLK7 expression has suggested a contradictory protective role for KLK7 in ovarian cancer patients. In order to help delineate its mechanism of action and thereby the functional roles, information on its substrate repertoire is crucial. Therefore, in this study a quantitative proteomics approach-PROtein TOpography and Migration Analysis Platform (PROTOMAP)-coupled with SILAC was used for in-depth analysis of putative KLK7 substrates from a representative ovarian cancer cell line, SKOV-3, secreted proteins. The Terminal Amine Isotopic Labeling of Substrates (TAILS) approach was used to determine the exact cleavage sites and to validate qPROTOMAP-identified putative substrates. By employing these two technically divergent approaches, exact cleavage sites on 16 novel putative substrates and two established substrates, matrix metalloprotease (MMP) 2 and insulin growth factor binding protein 3 (IGFBP3), were identified in the SKOV-3 secretome. Eight of these substrates were also identified on TAILS analysis of another ovarian cancer cell (OVMZ-6) secretome, with a further seven OVMZ-6 substrates common to the SKOV-3 qPROTOMAP profile. Identified substrates were significantly associated with the common processes of cell adhesion, extracellular matrix remodeling and cell migration according to the gene ontology (GO) biological process analysis. Biochemical validation supports a role for KLK7 in directly activating pro-MMP10, hydrolysis of IGFBP6 and cleavage of thrombospondin 1 with generation of a potentially bioactive N-terminal fragment. Overall, this study constitutes the most comprehensive analysis of the putative KLK7 degradome in any cancer to date, thereby opening new avenues for KLK7 research.
- Published
- 2018
16. N-Terminomics TAILS Identifies Host Cell Substrates of Poliovirus and Coxsackievirus B3 3C Proteinases That Modulate Virus Infection
- Author
-
Jayachandran N. Kizhakkedathu, Oded Kleifeld, Christopher M. Overall, Antoine Dufour, Nestor Solis, Eric Jan, Honglin Luo, Julienne Jagdeo, and Theo Klein
- Subjects
0301 basic medicine ,Proteases ,viruses ,Immunology ,Biology ,medicine.disease_cause ,Proteomics ,Microbiology ,Virus ,Substrate Specificity ,Heterogeneous-Nuclear Ribonucleoprotein K ,Viral Proteins ,03 medical and health sciences ,proteomics ,Virology ,medicine ,Humans ,coxsackievirus ,poliovirus ,030102 biochemistry & molecular biology ,enterovirus ,plus-strand RNA virus ,Poliovirus ,Endoplasmic reticulum ,3C Viral Proteases ,RNA ,Terminal amine isotopic labeling of substrates ,Virus-Cell Interactions ,Enterovirus B, Human ,3. Good health ,Cell biology ,Cysteine Endopeptidases ,030104 developmental biology ,Viral replication ,Isotope Labeling ,Insect Science ,RNA replication ,proteases ,proteinase ,Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor ,HeLa Cells - Abstract
Enteroviruses encode proteinases that are essential for processing of the translated viral polyprotein. In addition, viral proteinases also target host proteins to manipulate cellular processes and evade innate antiviral responses to promote replication and infection. Although some host protein substrates of enterovirus proteinases have been identified, the full repertoire of targets remains unknown. We used a novel quantitative in vitro proteomics-based approach, termed t erminal a mine i sotopic l abeling of s ubstrates (TAILS), to identify with high confidence 72 and 34 new host protein targets of poliovirus and coxsackievirus B3 (CVB3) 3C proteinases (3C pro s) in HeLa cell and cardiomyocyte HL-1 cell lysates, respectively. We validated a subset of candidate substrates that are targets of poliovirus 3C pro in vitro including three common protein targets, phosphoribosylformylglycinamidine synthetase (PFAS), hnRNP K, and hnRNP M, of both proteinases. 3C pro -targeted substrates were also cleaved in virus-infected cells but not noncleavable mutant proteins designed from the TAILS-identified cleavage sites. Knockdown of TAILS-identified target proteins modulated infection both negatively and positively, suggesting that cleavage by 3C pro promotes infection. Indeed, expression of a cleavage-resistant mutant form of the endoplasmic reticulum (ER)-Golgi vesicle-tethering protein p115 decreased viral replication and yield. As the first comprehensive study to identify and validate functional enterovirus 3C pro substrates in vivo , we conclude that N-terminomics by TAILS is an effective strategy to identify host targets of viral proteinases in a nonbiased manner. IMPORTANCE Enteroviruses are positive-strand RNA viruses that encode proteases that cleave the viral polyprotein into the individual mature viral proteins. In addition, viral proteases target host proteins in order to modulate cellular pathways and block antiviral responses in order to facilitate virus infection. Although several host protein targets have been identified, the entire list of proteins that are targeted is not known. In this study, we used a novel unbiased proteomics approach to identify ∼100 novel host targets of the enterovirus 3C protease, thus providing further insights into the network of cellular pathways that are modulated to promote virus infection.
- Published
- 2018
17. Outer membrane vesicles from Neisseria gonorrhoeae target PorB to mitochondria and induce apoptosis
- Author
-
Kirstin Elgass, Iain D. Hay, Jhih-Hang Jiang, P. Deo, Seong Hoong Chow, Gordon Dougan, Oded Kleifeld, Kipros Gabriel, Georg Ramm, Eva Heinz, Thomas Naderer, Adam Costin, Trevor Lithgow, Kubori, T, Deo, Pankaj [0000-0002-6947-5317], Chow, Seong H [0000-0002-4392-3863], Hay, Iain D [0000-0001-8797-6038], Kleifeld, Oded [0000-0003-3091-7154], Costin, Adam [0000-0001-5334-557X], Jiang, Jhih-Hang [0000-0002-1543-1634], Ramm, Georg [0000-0003-3596-2288], Naderer, Thomas [0000-0003-2691-0283], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Sexually transmitted disease ,Cell Membranes ,Apoptosis ,Pathology and Laboratory Medicine ,medicine.disease_cause ,Biochemistry ,White Blood Cells ,Gonorrhea ,Mice ,Animal Cells ,Medicine and Health Sciences ,Biology (General) ,Energy-Producing Organelles ,Staining ,Membrane Potential, Mitochondrial ,Cell Death ,Chemistry ,Bacterial Pathogens ,Mitochondria ,3. Good health ,Cell biology ,Membrane Staining ,Medical Microbiology ,Cell Processes ,Porin ,Pathogens ,Cellular Structures and Organelles ,Cellular Types ,Bacterial outer membrane ,Neisseria ,Research Article ,Programmed cell death ,QH301-705.5 ,Immune Cells ,Immunology ,Porins ,Bioenergetics ,Research and Analysis Methods ,Microbiology ,03 medical and health sciences ,Virology ,Genetics ,medicine ,Animals ,Humans ,Vesicles ,Microbial Pathogens ,Molecular Biology ,Blood Cells ,Innate immune system ,Bacteria ,Macrophages ,Cell Membrane ,Organisms ,Biology and Life Sciences ,Membrane Proteins ,Cell Biology ,RC581-607 ,Outer Membrane Proteins ,Neisseria gonorrhoeae ,Mice, Inbred C57BL ,030104 developmental biology ,Membrane protein ,Specimen Preparation and Treatment ,Parasitology ,Immunologic diseases. Allergy - Abstract
Neisseria gonorrhoeae causes the sexually transmitted disease gonorrhoea by evading innate immunity. Colonizing the mucosa of the reproductive tract depends on the bacterial outer membrane porin, PorB, which is essential for ion and nutrient uptake. PorB is also targeted to host mitochondria and regulates apoptosis pathways to promote infections. How PorB traffics from the outer membrane of N. gonorrhoeae to mitochondria and whether it modulates innate immune cells, such as macrophages, remains unclear. Here, we show that N. gonorrhoeae secretes PorB via outer membrane vesicles (OMVs). Purified OMVs contained primarily outer membrane proteins including oligomeric PorB. The porin was targeted to mitochondria of macrophages after exposure to purified OMVs and wild type N. gonorrhoeae. This was associated with loss of mitochondrial membrane potential, release of cytochrome c, activation of apoptotic caspases and cell death in a time-dependent manner. Consistent with this, OMV-induced macrophage death was prevented with the pan-caspase inhibitor, Q-VD-PH. This shows that N. gonorrhoeae utilizes OMVs to target PorB to mitochondria and to induce apoptosis in macrophages, thus affecting innate immunity., Author summary Neisseria gonorrhoeae causes the sexually transmitted disease gonorrhoea in more than 100 million people worldwide every year. The bacteria replicate in the reproductive tract by evading innate and adaptive immunity. In the absence of effective vaccines and the rise of antibiotic resistance, understanding the molecular interactions between innate immune cells and N. gonorrhoeae may lead to new strategies to combat bacterial growth and the symptoms of gonorrhoea. It has long been known that the N. gonorrhoeae porin, PorB, promotes bacterial survival but also targets host mitochondria in infections. The mechanism by which PorB traffics form the bacterial outer membrane to host mitochondria remains unclear. Here, we utilized proteomics and super-resolution microscopy to show that N. gonorrhoeae secretes PorB via outer membrane vesicles. These vesicles are taken up by macrophages and deliver PorB to mitochondria. Macrophages treated with N. gonorrhoeae vesicles contained damaged mitochondria and active caspase-3. A caspase inhibitor prevented apoptosis of macrophages treated with N. gonorrhoeae vesicles. This suggests that N. gonorrhoeae secretes membrane vesicles, which are readily detectable in gonorrhoea patients, to target macrophages and to promote infections.
- Published
- 2018
18. Granzyme B Promotes Cytotoxic Lymphocyte Transmigration via Basement Membrane Remodeling
- Author
-
Paulus Mrass, Stephen J. Turner, Bosco K. Ho, Marcia A. Munoz, Matthew R. Olson, Philip L. Tong, Monica Devi Prakash, Aulikki Koskinen, Phillip I. Bird, Rohit Jain, Oded Kleifeld, Wolfgang Weninger, and Matthias Regner
- Subjects
Ectromelia virus ,Lymphocyte ,Immunology ,Basement Membrane ,Granzymes ,GZMB ,Extracellular matrix ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Ectromelia, Infectious ,Cells, Cultured ,030304 developmental biology ,Mice, Knockout ,Basement membrane ,Extracellular Matrix Proteins ,0303 health sciences ,biology ,Transendothelial and Transepithelial Migration ,biology.organism_classification ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,Granzyme B ,Infectious Diseases ,medicine.anatomical_structure ,Granzyme ,030220 oncology & carcinogenesis ,Proteolysis ,biology.protein ,Chemokines ,T-Lymphocytes, Cytotoxic - Abstract
SummaryGranzyme B (GzmB) is a protease with a well-characterized intracellular role in targeted destruction of compromised cells by cytotoxic lymphocytes. However, GzmB also cleaves extracellular matrix components, suggesting that it influences the interplay between cytotoxic lymphocytes and their environment. Here, we show that GzmB-null effector T cells and natural killer (NK) cells exhibited a cell-autonomous homing deficit in mouse models of inflammation and Ectromelia virus infection. Intravital imaging of effector T cells in inflamed cremaster muscle venules revealed that GzmB-null cells adhered normally to the vessel wall and could extend lamellipodia through it but did not cross it efficiently. In vitro migration assays showed that active GzmB was released from migrating cytotoxic lymphocytes and enabled chemokine-driven movement through basement membranes. Finally, proteomic analysis demonstrated that GzmB cleaved basement membrane constituents. Our results highlight an important role for GzmB in expediting cytotoxic lymphocyte diapedesis via basement membrane remodeling.
- Published
- 2014
19. Structure-function analyses of a pertussis-like toxin from pathogenic Escherichia coli reveal a distinct mechanism of inhibition of trimeric G-proteins
- Author
-
Mark A. Schembri, Roger J. Summers, Jamie Rossjohn, Martina Kocan, Oded Kleifeld, James C. Paton, Adrienne W. Paton, Sheng Y. Ang, Travis Clarke Beddoe, Dene R. Littler, Mai T. Tran, Danilo Gomes Moriel, and Matthew D. Johnson
- Subjects
Models, Molecular ,0301 basic medicine ,Bordetella pertussis ,media_common.quotation_subject ,Bacterial Toxins ,030106 microbiology ,Clostridium difficile toxin B ,medicine.disease_cause ,Pertussis toxin ,Biochemistry ,Microbiology ,Structure-Activity Relationship ,03 medical and health sciences ,AB toxin ,Chlorocebus aethiops ,Escherichia coli ,medicine ,Animals ,Humans ,Editors' Picks ,Internalization ,Vero Cells ,Molecular Biology ,Cell Proliferation ,media_common ,Dose-Response Relationship, Drug ,biology ,Cholera toxin ,Epithelial Cells ,Cell Biology ,biology.organism_classification ,Heterotrimeric GTP-Binding Proteins ,3. Good health ,HEK293 Cells ,Pertussis Toxin ,ADP-ribosylation - Abstract
Pertussis-like toxins are secreted by several bacterial pathogens during infection. They belong to the AB5 virulence factors, which bind to glycans on host cell membranes for internalization. Host cell recognition and internalization are mediated by toxin B subunits sharing a unique pentameric ring-like assembly. Although the role of pertussis toxin in whooping cough is well-established, pertussis-like toxins produced by other bacteria are less studied, and their mechanisms of action are unclear. Here, we report that some extra-intestinal Escherichia coli pathogens (i.e. those that reside in the gut but can spread to other bodily locations) encode a pertussis-like toxin that inhibits mammalian cell growth in vitro. We found that this protein, EcPlt, is related to toxins produced by both nontyphoidal and typhoidal Salmonella serovars. Pertussis-like toxins are secreted as disulfide-bonded heterohexamers in which the catalytic ADP-ribosyltransferase subunit is activated when exposed to the reducing environment in mammalian cells. We found here that the reduced EcPlt exhibits large structural rearrangements associated with its activation. We noted that inhibitory residues tethered within the NAD+-binding site by an intramolecular disulfide in the oxidized state dissociate upon the reduction and enable loop restructuring to form the nucleotide-binding site. Surprisingly, although pertussis toxin targets a cysteine residue within the α subunit of inhibitory trimeric G-proteins, we observed that activated EcPlt toxin modifies a proximal lysine/asparagine residue instead. In conclusion, our results reveal the molecular mechanism underpinning activation of pertussis-like toxins, and we also identified differences in host target specificity.
- Published
- 2017
20. Oncogenic ZEB2 activation drives sensitivity toward KDM1A inhibition in T-cell acute lymphoblastic leukemia
- Author
-
Steven Goossens, Pieter Van Vlierberghe, Wouter Van Loocke, Magdaline Costa, Niels Vandamme, David J. Curtis, Thao Nguyen, Dieter Deforce, Jody J. Haigh, Katharina Haigh, Sofie Peirs, Filip Matthijssens, Catherine Carmichael, Oded Kleifeld, Stefan Eugen Sonderegger, Filip Van Nieuwerburgh, Jueqiong Wang, Mina Takawy, and Geert Berx
- Subjects
0301 basic medicine ,Cyclopropanes ,T cell ,Immunology ,Mice, SCID ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Biochemistry ,Benzoates ,Article ,03 medical and health sciences ,Mice ,Mice, Inbred NOD ,Cell Line, Tumor ,Demethylase activity ,medicine ,Animals ,Humans ,Protein Interaction Maps ,Zinc Finger E-box Binding Homeobox 2 ,Zinc finger ,Histone Demethylases ,Homeodomain Proteins ,Gene Expression Regulation, Leukemic ,Myeloid leukemia ,KDM1A ,Cell Biology ,Hematology ,medicine.disease ,Up-Regulation ,Repressor Proteins ,Leukemia ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,biology.protein ,Demethylase - Abstract
Elevated expression of the Zinc finger E-box binding homeobox transcription factor-2 (ZEB2) is correlated with poor prognosis and patient outcome in a variety of human cancer subtypes. Using a conditional gain-of-function mouse model, we recently demonstrated that ZEB2 is an oncogenic driver of immature T-cell acute lymphoblastic leukemia (T-ALL), a heterogenic subgroup of human leukemia characterized by a high incidence of remission failure or hematological relapse after conventional chemotherapy. Here, we identified the lysine-specific demethylase KDM1A as a novel interaction partner of ZEB2 and demonstrated that mouse and human T-ALLs with increased ZEB2 levels critically depend on KDM1A activity for survival. Therefore, targeting the ZEB2 protein complex through direct disruption of the ZEB2-KDM1A interaction or pharmacological inhibition of the KDM1A demethylase activity itself could serve as a novel therapeutic strategy for this aggressive subtype of human leukemia and possibly other ZEB2-driven malignancies.
- Published
- 2017
21. Studying Protein Ubiquitylation in Yeast
- Author
-
Sébastien Léon, Michel Becuwe, Michael H. Glickman, Junie Hovsepian, Oded Kleifeld, Institut Jacques Monod (IJM (UMR_7592)), Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Department of Biology, Technion Israel institute of Technology, Technion - Israel Institute of Technology [Haifa], and Faculty of Biology, Technion
- Subjects
0301 basic medicine ,biology ,Chemistry ,Saccharomyces cerevisiae ,Substrate (chemistry) ,[SDV.BC.BC]Life Sciences [q-bio]/Cellular Biology/Subcellular Processes [q-bio.SC] ,Subcellular localization ,biology.organism_classification ,Yeast ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Protein ubiquitylation ,Ubiquitin ,Posttranslational modification ,biology.protein ,Moiety ,ComputingMilieux_MISCELLANEOUS - Abstract
Ubiquitylation is a reversible posttranslational modification that is critical for most, if not all, cellular processes and essential for viability. Ubiquitin conjugates to substrate proteins either as a single moiety (monoubiquitylation) or as polymers composed of ubiquitin molecules linked to each other with various topologies and structures (polyubiquitylation). This contributes to an elaborate ubiquitin code that is decrypted by specific ubiquitin-binding proteins. Indeed, these different types of ubiquitylation have different functional outcomes, notably affecting the stability of the substrate, its interactions, its activity, or its subcellular localization. In this chapter, we describe protocols to determine whether a protein is ubiquitylated, to identify the site that is ubiquitylated, and provide direction to study the topology of the ubiquitin modification, in the yeast Saccharomyces cerevisiae.
- Published
- 2016
22. Physicochemical properties that control protein aggregation also determine whether a protein is retained or released from necrotic cells
- Author
-
Oded Kleifeld, Mark J. Smyth, Amanda E. Au, Andre L. Samson, Bosco K. Ho, Stephen P. Bottomley, Robert L. Medcalf, and Simone M. Schoenwaelder
- Subjects
0301 basic medicine ,Proteomics ,Cell type ,immune tolerance ,Cytoplasm ,Fas Ligand Protein ,Proteome ,Cell Survival ,Immunology ,Apoptosis ,Protein aggregation ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Immune tolerance ,protein aggregation ,necrosis ,Cell Line ,03 medical and health sciences ,Jurkat Cells ,Protein Aggregates ,parasitic diseases ,Humans ,lcsh:QH301-705.5 ,RNA-binding protein FUS ,Etoposide ,Cell Nucleus ,urogenital system ,General Neuroscience ,Research ,Staurosporine ,Cell biology ,030104 developmental biology ,lcsh:Biology (General) ,Biochemistry ,embryonic structures ,Liberation ,Intracellular ,RNA-Binding Protein FUS ,Research Article ,disulfide - Abstract
Amyloidogenic protein aggregation impairs cell function and is a hallmark of many chronic degenerative disorders. Protein aggregation is also a major event during acute injury; however, unlike amyloidogenesis, the process of injury-induced protein aggregation remains largely undefined. To provide this insight, we profiled the insoluble proteome of several cell types after acute injury. These experiments show that the disulfide-driven process of nucleocytoplasmic coagulation (NCC) is the main form of injury-induced protein aggregation. NCC is mechanistically distinct from amyloidogenesis, but still broadly impairs cell function by promoting the aggregation of hundreds of abundant and essential intracellular proteins. A small proportion of the intracellular proteome resists NCC and is instead released from necrotic cells. Notably, the physicochemical properties of NCC-resistant proteins are contrary to those of NCC-sensitive proteins. These observations challenge the dogma that liberation of constituents during necrosis is anarchic. Rather, inherent physicochemical features including cysteine content, hydrophobicity and intrinsic disorder determine whether a protein is released from necrotic cells. Furthermore, as half of the identified NCC-resistant proteins are known autoantigens, we propose that physicochemical properties that control NCC also affect immune tolerance and other host responses important for the restoration of homeostasis after necrotic injury.
- Published
- 2016
23. Structure of ubiquitylated-Rpn10 provides insight into its autoregulation mechanism
- Author
-
Lee Zeev Peters, Ori Zucker, Shay Ben-Aroya, Tal Keren-Kaplan, Olga Levin-Kravets, Oded Kleifeld, Gali Prag, Ilan Attali, Noa Shohat, Shay Artzi, Inbar Pilzer, Noa Reis, and Michael H. Glickman
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Saccharomyces cerevisiae Proteins ,Protein Conformation ,Protein subunit ,Science ,Protein domain ,Molecular Conformation ,General Physics and Astronomy ,Plasma protein binding ,Saccharomyces cerevisiae ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Open Reading Frames ,Protein structure ,Ubiquitin ,Protein Domains ,Gene Expression Regulation, Fungal ,Protein Interaction Mapping ,Receptor ,Multidisciplinary ,Wild type ,Ubiquitination ,General Chemistry ,DNA ,Surface Plasmon Resonance ,3. Good health ,Cell biology ,Microscopy, Electron ,030104 developmental biology ,Biochemistry ,Proteasome ,biology.protein ,Protein Binding - Abstract
Ubiquitin receptors decode ubiquitin signals into many cellular responses. Ubiquitin receptors also undergo coupled monoubiquitylation, and rapid deubiquitylation has hampered the characterization of the ubiquitylated state. Using bacteria that express a ubiquitylation apparatus, we purified and determined the crystal structure of the proteasomal ubiquitin-receptor Rpn10 in its ubiquitylated state. The structure shows a novel ubiquitin-binding patch that directs K84 ubiquitylation. Superimposition of ubiquitylated-Rpn10 onto electron-microscopy models of proteasomes indicates that the Rpn10-conjugated ubiquitin clashes with Rpn9, suggesting that ubiquitylation might be involved in releasing Rpn10 from the proteasome. Indeed, ubiquitylation on immobilized proteasomes dissociates the modified Rpn10 from the complex, while unmodified Rpn10 mainly remains associated. In vivo experiments indicate that contrary to wild type, Rpn10-K84R is stably associated with the proteasomal subunit Rpn9. Similarly Rpn10, but not ubiquitylated-Rpn10, binds Rpn9 in vitro. Thus we suggest that ubiquitylation functions to dissociate modified ubiquitin receptors from their targets, a function that promotes cyclic activity of ubiquitin receptors., Ubiquitin (Ub) receptors are responsible for the recognition of ubiquitylated proteins. Here the authors describe the crystal structure of the ubiquitylated form of the Ub-receptor Rpn10, which suggest that ubiquitylation of Rpn10 promotes its dissociation from the proteasome.
- Published
- 2015
24. Nuclear import of an intact preassembled proteasome particle
- Author
-
Anca F. Savulescu, Ilana Cohen, Rita Gruber, Oded Kleifeld, Amnon Harel, Michael H. Glickman, and Hagai Shorer
- Subjects
Cytoplasm ,Proteasome Endopeptidase Complex ,Nucleoplasm ,Nuclear Envelope ,Nuclear Functions ,Active Transport, Cell Nucleus ,Cell Biology ,Articles ,Biology ,Protein degradation ,Recombinant Proteins ,Cell biology ,Protein Subunits ,Xenopus laevis ,ran GTP-Binding Protein ,Ran ,Nuclear Pore ,Oocytes ,Animals ,Nucleoporin ,Nuclear pore ,Nuclear transport ,Nuclear protein ,Molecular Biology ,Lamin - Abstract
Nuclear targeting of intact proteasome particles was tested in the Xenopus egg extract system. Both the 26S proteasome holoenzyme and the 20S core particle were targeted to the nuclear envelope but could not enter the nucleus. A novel proteolytically active 20S+ particle was actively imported into the nucleoplasm in a Ran-independent fashion., The 26S proteasome is a conserved 2.5 MDa protein degradation machine that localizes to different cellular compartments, including the nucleus. Little is known about the specific targeting mechanisms of proteasomes in eukaryotic cells. We used a cell-free nuclear reconstitution system to test for nuclear targeting and import of distinct proteasome species. Three types of stable, proteolytically active proteasomes particles were purified from Xenopus egg cytosol. Two of these, the 26S holoenzyme and the 20S core particle, were targeted to the nuclear periphery but did not reach the nucleoplasm. This targeting depends on the presence of mature nuclear pore complexes (NPCs) in the nuclear envelope. A third, novel form, designated here as 20S+, was actively imported through NPCs. The 20S+ proteasome particle resembles recently described structural intermediates from other systems. Nuclear import of this particle requires functional NPCs, but it is not directly regulated by the Ran GTPase cycle. The mere presence of the associated “+” factors is sufficient to reconstitute nuclear targeting and confer onto isolated 20S core particles the ability to be imported. Stable 20S+ particles found in unfertilized eggs may provide a means for quick mobilization of existing proteasome particles into newly formed nuclear compartments during early development.
- Published
- 2011
25. Extraproteasomal Rpn10 Restricts Access of the Polyubiquitin-Binding Protein Dsk2 to Proteasome
- Author
-
Woong Kim, Oded Kleifeld, Arun Dakshinamurthy, Yulia Matiuhin, Inbal Ziv, Michael H. Glickman, Noa Reis, Donald S. Kirkpatrick, and Steven P. Gygi
- Subjects
Proteasome Endopeptidase Complex ,Saccharomyces cerevisiae Proteins ,Proteolysis ,Cell Cycle Proteins ,Saccharomyces cerevisiae ,Ubiquitin-conjugating enzyme ,Deubiquitinating enzyme ,Ubiquitin ,medicine ,Humans ,Polyubiquitin ,Ubiquitins ,Molecular Biology ,biology ,medicine.diagnostic_test ,RNA-Binding Proteins ,Cell Biology ,Ubiquitin ligase ,Kinetics ,Biochemistry ,Proteasome ,biology.protein ,Polyubiquitin binding ,Protein Binding - Abstract
Polyubiquitin is a diverse signal both in terms of chain length and linkage type. Lysine 48-linked ubiquitin is essential for marking targets for proteasomal degradation, but the significance and relative abundance of different linkages remain ambiguous. Here we dissect the relationship of two proteasome-associated polyubiquitin-binding proteins, Rpn10 and Dsk2, and demonstrate how Rpn10 filters Dsk2 interactions, maintaining proper function of the ubiquitin-proteasome system. Using quantitative mass spectrometry of ubiquitin, we found that in S. cerevisiae under normal growth conditions the majority of conjugated ubiquitin was linked via lysine 48 and lysine 63. In contrast, upon DSK2 induction, conjugates accumulated primarily in the form of lysine 48 linkages correlating with impaired proteolysis and cytotoxicity. By restricting Dsk2 access to the proteasome, extraproteasomal Rpn10 was essential for alleviating the cellular stress associated with Dsk2. This work highlights the importance of polyubiquitin shuttles such as Rpn10 and Dsk2 in controlling the ubiquitin landscape.
- Published
- 2008
26. Base-CP proteasome can serve as a platform for stepwise lid formation
- Author
-
Michael H. Glickman, Mark A. Nakasone, David Fushman, Carlos A. Castañeda, Noa Reis, Oded Kleifeld, Zanlin Yu, Wissam Mansour, Elah Pick, Nurit Livnat-Levanon, and Emma K. Dixon
- Subjects
Models, Molecular ,Proteasome Endopeptidase Complex ,TPP, trans proteomic pipeline ,Saccharomyces cerevisiae Proteins ,medicine.medical_treatment ,Protein domain ,Mutant ,MPN ,polyUb, polyubiquitin ,lcsh:Life ,lcsh:QR1-502 ,Biophysics ,medicine.disease_cause ,Bioinformatics ,Biochemistry ,lcsh:Microbiology ,Holoenzymes ,Protein Domains ,19S regulatory particle ,Endopeptidases ,medicine ,Gene silencing ,base ,Gene Silencing ,Molecular Biology ,26S proteasome ,Ub, ubiquitin ,Mutation ,Original Paper ,Protease ,Chemistry ,rpn11-m1 ,CP, 20S core particle ,DUB, deubiquitinase ,PCI ,Cell Biology ,WCE, whole cell extract ,WT, wild-type ,Cell biology ,lcsh:QH501-531 ,Proteasome ,20S core particle ,Proteasome assembly ,lid ,RP, 19S regulatory particle - Abstract
26S proteasome, a major regulatory protease in eukaryotes, consists of a 20S proteolytic core particle (CP) capped by a 19S regulatory particle (RP). The 19S RP is divisible into base and lid sub-complexes. Even within the lid, subunits have been demarcated into two modules: module 1 (Rpn5, Rpn6, Rpn8, Rpn9 and Rpn11), which interacts with both CP and base sub-complexes and module 2 (Rpn3, Rpn7, Rpn12 and Rpn15) that is attached mainly to module 1. We now show that suppression of RPN11 expression halted lid assembly yet enabled the base and 20S CP to pre-assemble and form a base-CP. A key role for Regulatory particle non-ATPase 11 (Rpn11) in bridging lid module 1 and module 2 subunits together is inferred from observing defective proteasomes in rpn11–m1, a mutant expressing a truncated form of Rpn11 and displaying mitochondrial phenotypes. An incomplete lid made up of five module 1 subunits attached to base-CP was identified in proteasomes isolated from this mutant. Re-introducing the C-terminal portion of Rpn11 enabled recruitment of missing module 2 subunits. In vitro, module 1 was reconstituted stepwise, initiated by Rpn11–Rpn8 heterodimerization. Upon recruitment of Rpn6, the module 1 intermediate was competent to lock into base-CP and reconstitute an incomplete 26S proteasome. Thus, base-CP can serve as a platform for gradual incorporation of lid, along a proteasome assembly pathway. Identification of proteasome intermediates and reconstitution of minimal functional units should clarify aspects of the inner workings of this machine and how multiple catalytic processes are synchronized within the 26S proteasome holoenzymes., Defective proteasome 19S regulatory particles (RPs) were identified in rpn11f–m1, a proteasomal mutant with mitochondrial phenotypes. The Rpn11 subunit initiates assembly of a five-subunit lid module competent to integrate into pre-assembled base-20S core particle (CP), with subsequent recruitment of remaining lid subunits.
- Published
- 2015
27. Disassembly of Lys11 and Mixed Linkage Polyubiquitin Conjugates Provides Insights into Function of Proteasomal Deubiquitinases Rpn11 and Ubp6*
- Author
-
Michael H. Glickman, Maximilian von Delbrück, Thomas Sommer, Noa Reis, Wissam Mansour, Zanlin Yu, David Fushman, Oded Kleifeld, Mark A. Nakasone, and Daria Krutauz
- Subjects
Proteasome Endopeptidase Complex ,Saccharomyces cerevisiae Proteins ,Proteolysis ,macromolecular substances ,Saccharomyces cerevisiae ,Biochemistry ,environment and public health ,Ubiquitin ,Endopeptidases ,medicine ,Polyubiquitin ,Molecular Biology ,biology ,medicine.diagnostic_test ,Chemistry ,C-terminus ,Lysine ,Cell Biology ,Cysteine protease ,Protein Structure, Tertiary ,Proteasome ,Protein Synthesis and Degradation ,biology.protein ,Biophysics ,Function (biology) ,Conjugate ,Deubiquitination - Abstract
Protein homeostasis is largely dependent on proteolysis by the ubiquitin-proteasome system. Diverse polyubiquitin modifications are reported to target cellular proteins to the proteasome. At the proteasome, deubiquitination is an essential preprocessing event that contributes to degradation efficiency. We characterized the specificities of two proteasome-associated deubiquitinases (DUBs), Rpn11 and Ubp6, and explored their impact on overall proteasome DUB activity. This was accomplished by constructing a panel of well defined ubiquitin (Ub) conjugates, including homogeneous linkages of varying lengths as well as a heterogeneously modified target. Rpn11 and Ubp6 processed Lys(11) and Lys(63) linkages with comparable efficiencies that increased with chain length. In contrast, processing of Lys(48) linkages by proteasome was inversely correlated to chain length. Fluorescently labeled tetra-Ub chains revealed endo-chain preference for Ubp6 acting on Lys(48) and random action for Rpn11. Proteasomes were more efficient at deconjugating identical substrates than their constituent DUBs by roughly 2 orders of magnitude. Incorporation into proteasomes significantly enhanced enzymatic efficiency of Rpn11, due in part to alleviation of the autoinhibitory role of its C terminus. The broad specificity of Rpn11 could explain how proteasomes were more effective at disassembling a heterogeneously modified conjugate compared with homogeneous Lys(48)-linked chains. The reduced ability to disassemble homogeneous Lys(48)-linked chains longer than 4 Ub units may prolong residency time on the proteasome.
- Published
- 2014
28. Oxidation of an Exposed Methionine Instigates the Aggregation of Glyceraldehyde-3-phosphate Dehydrogenase*
- Author
-
Stephen P. Bottomley, Emilia M. Marijanovic, Itamar Kass, Victoria A Hughes, Christopher J. Lupton, Andre L. Samson, Ashley M. Buckle, Robert L. Medcalf, Oded Kleifeld, and Anja S Knaupp
- Subjects
Models, Molecular ,Protein Folding ,Mutation, Missense ,Context (language use) ,Dehydrogenase ,Protein aggregation ,medicine.disease_cause ,Biochemistry ,Protein Aggregation, Pathological ,chemistry.chemical_compound ,Methionine ,stomatognathic system ,medicine ,Humans ,Molecular Biology ,Glyceraldehyde 3-phosphate dehydrogenase ,Mutation ,biology ,Glyceraldehyde-3-Phosphate Dehydrogenases ,Cell Biology ,chemistry ,Amino Acid Substitution ,Protein Structure and Folding ,biology.protein ,Protein folding ,Leucine ,Oxidation-Reduction ,Protein Processing, Post-Translational - Abstract
Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is a ubiquitous and abundant protein that participates in cellular energy production. GAPDH normally exists in a soluble form; however, following necrosis, GAPDH and numerous other intracellular proteins convert into an insoluble disulfide-cross-linked state via the process of "nucleocytoplasmic coagulation." Here, free radical-induced aggregation of GAPDH was studied as an in vitro model of nucleocytoplasmic coagulation. Despite the fact that disulfide cross-linking is a prominent feature of GAPDH aggregation, our data show that it is not a primary rate-determining step. To identify the true instigating event of GAPDH misfolding, we mapped the post-translational modifications that arise during its aggregation. Solvent accessibility and energy calculations of the mapped modifications within the context of the high resolution native GAPDH structure suggested that oxidation of methionine 46 may instigate aggregation. We confirmed this by mutating methionine 46 to leucine, which rendered GAPDH highly resistant to free radical-induced aggregation. Molecular dynamics simulations suggest that oxidation of methionine 46 triggers a local increase in the conformational plasticity of GAPDH that likely promotes further oxidation and eventual aggregation. Hence, methionine 46 represents a "linchpin" whereby its oxidation is a primary event permissive for the subsequent misfolding, aggregation, and disulfide cross-linking of GAPDH. A critical role for linchpin residues in nucleocytoplasmic coagulation and other forms of free radical-induced protein misfolding should now be investigated. Furthermore, because disulfide-cross-linked aggregates of GAPDH arise in many disorders and because methionine 46 is irrelevant to native GAPDH function, mutation of methionine 46 in models of disease should allow the unequivocal assessment of whether GAPDH aggregation influences disease progression.
- Published
- 2014
29. Conformational changes during pore formation by the perforin-related protein pleurotolysin
- Author
-
James C. Whisstock, Rodney K. Tweten, Michelle A. Dunstone, Oded Kleifeld, Helen R. Saibil, Daouda A K Traore, S.C. Kondos, Bradley A. Spicer, Ruby H. P. Law, Cyril F. Reboul, Joseph A. Trapani, Katherine V. Oliver, Maya Topf, Ilia Voskoboinik, Tamas Zsolt Hatfaludi, Natalya Lukoyanova, Eileen M. Hotze, Irene Farabella, Susan M. Ekkel, and Tom T. Caradoc-Davies
- Subjects
Models, Molecular ,Conformational change ,Protein Folding ,Erythrocytes ,QH301-705.5 ,Recombinant Fusion Proteins ,Gene Expression ,Complement Membrane Attack Complex ,Biology ,Cholesterol-dependent cytolysin ,Q1 ,bcs ,Crystallography, X-Ray ,Pleurotus ,General Biochemistry, Genetics and Molecular Biology ,Protein Structure, Secondary ,Fungal Proteins ,Hemolysin Proteins ,Protein structure ,Escherichia coli ,Animals ,Protein Isoforms ,Biology (General) ,Integral membrane protein ,Fungal protein ,MACPF ,Sheep ,General Immunology and Microbiology ,General Neuroscience ,QH ,Cell Membrane ,Cryoelectron Microscopy ,Transmembrane protein ,Cell biology ,Protein Structure, Tertiary ,Biophysics ,Protein folding ,General Agricultural and Biological Sciences ,Protein Binding ,Research Article - Abstract
Membrane attack complex/perforin-like (MACPF) proteins comprise the largest superfamily of pore-forming proteins, playing crucial roles in immunity and pathogenesis. Soluble monomers assemble into large transmembrane pores via conformational transitions that remain to be structurally and mechanistically characterised. Here we present an 11 Å resolution cryo-electron microscopy (cryo-EM) structure of the two-part, fungal toxin Pleurotolysin (Ply), together with crystal structures of both components (the lipid binding PlyA protein and the pore-forming MACPF component PlyB). These data reveal a 13-fold pore 80 Å in diameter and 100 Å in height, with each subunit comprised of a PlyB molecule atop a membrane bound dimer of PlyA. The resolution of the EM map, together with biophysical and computational experiments, allowed confident assignment of subdomains in a MACPF pore assembly. The major conformational changes in PlyB are a ∼70° opening of the bent and distorted central β-sheet of the MACPF domain, accompanied by extrusion and refolding of two α-helical regions into transmembrane β-hairpins (TMH1 and TMH2). We determined the structures of three different disulphide bond-trapped prepore intermediates. Analysis of these data by molecular modelling and flexible fitting allows us to generate a potential trajectory of β-sheet unbending. The results suggest that MACPF conformational change is triggered through disruption of the interface between a conserved helix-turn-helix motif and the top of TMH2. Following their release we propose that the transmembrane regions assemble into β-hairpins via top down zippering of backbone hydrogen bonds to form the membrane-inserted β-barrel. The intermediate structures of the MACPF domain during refolding into the β-barrel pore establish a structural paradigm for the transition from soluble monomer to pore, which may be conserved across the whole superfamily. The TMH2 region is critical for the release of both TMH clusters, suggesting why this region is targeted by endogenous inhibitors of MACPF function., Author Summary Animals, plants, fungi, and bacteria all use pore-forming proteins of the membrane attack complex-perforin (MACPF) family as lethal, cell-killing weapons. These proteins are able to insert into the plasma membranes of target cells, creating large pores that short circuit the natural separation between the intracellular and extracellular milieu, with catastrophic results. However, the pore-forming proteins must undergo a substantial transformation from soluble precursors to a large barrel-shaped transmembrane complex as they punch their way into cells. Using a combination of X-ray crystallography and cryo electron microscopy, we have visualized, for the first time, the mechanism of action of one of these pore-forming proteins—pleurotolysin, a MACPF protein from the edible oyster mushroom. This enabled us to propose a model of the pleurotolysin pore by fitting the crystallographic structures of the pore proteins into a three-dimensional map of the pore obtained by cryo electron microscopy. We then designed a set of double mutants that allowed us to chemically trap intermediate states along the trajectory of the pore formation process, and to determine their structures too. By combining these data we proposed a detailed molecular mechanism for pore formation. The pleurotolysin first assembles into rings of 13 subunits, each of which then opens up by about 70° during pore formation. This process is accompanied by refolding and extrusion of two compact regions from each subunit into long hairpins that then zipper together to form an 80-Å wide barrel-shaped channel through the membrane., A combination of structural methods reveals the complex process by which the perforin-like fungal toxin Pleurotolysin rearranges its structure to form a pore that punches a hole in target cell membranes.
- Published
- 2014
30. Structural and mechanistic insight into alkane hydroxylation by Pseudomonas putida AlkB
- Author
-
Yu C. Liu, Hernan Alonso, Jeanette E. Stok, James J. De Voss, Oded Kleifeld, Adva Yeheskel, Anna Roujeinikova, and Poh C. Ong
- Subjects
Models, Molecular ,Stereochemistry ,AlkB ,Hydroxylation ,Biochemistry ,chemistry.chemical_compound ,Tandem Mass Spectrometry ,Catalytic Domain ,Alkanes ,Molecular Biology ,Integral membrane protein ,Alanine ,biology ,Pseudomonas putida ,Lysine ,Mutagenesis ,Active site ,Membrane Proteins ,Cell Biology ,biology.organism_classification ,Transmembrane domain ,chemistry ,Alkynes ,biology.protein ,Cytochrome P-450 CYP4A ,Hydrophobic and Hydrophilic Interactions - Abstract
Pseudomonas putida GPo1 alkane hydroxylase (AlkB) is an integral membrane protein that catalyses the hydroxylation of medium-chain alkanes (C3–C12). 1-Octyne irreversibly inhibits this non-haem di-iron mono-oxygenase under turnover conditions, suggesting that it acts as a mechanism-based inactivator. Upon binding to the active site, 1-octyne is postulated to be oxidized to an oxirene that rapidly rearranges to a reactive ketene which covalently acylates nearby residues, resulting in enzyme inactivation. In analysis of inactivated AlkB by LC–MS/MS, several residues exhibited a mass increase of 126.1 Da, corresponding to the octanoyl moiety derived from oxidative activation of 1-octyne. Mutagenesis studies of conserved acylated residues showed that Lys18 plays a critical role in enzyme function, as a single-point mutation of Lys18 to alanine (K18A) completely abolished enzymatic activity. Finally, we present a computational 3D model structure of the transmembrane domain of AlkB, which revealed the overall packing arrangement of the transmembrane helices within the lipid bilayer and the location of the active site mapped by the 1-octyne modifications.
- Published
- 2014
31. Activated platelets rescue apoptotic cells via paracrine activation of EGFR and DNA-dependent protein kinase
- Author
-
Robert K. Andrews, Oded Kleifeld, Elizabeth E. Gardiner, Andre L. Samson, Maithili Sashindranath, Amanda E. Au, Rachael Jade Borg, and Robert L. Medcalf
- Subjects
Adult ,Blood Platelets ,Male ,Cancer Research ,Adolescent ,Immunology ,Apoptosis ,DNA-Activated Protein Kinase ,Biology ,Cellular and Molecular Neuroscience ,Paracrine signalling ,Young Adult ,Cell Line, Tumor ,Animals ,Humans ,Platelet ,Platelet activation ,Epidermal growth factor receptor ,Protein kinase A ,Cells, Cultured ,Aged ,Neurons ,Kinase ,Activator (genetics) ,Brain ,Cell Biology ,Middle Aged ,Platelet Activation ,Cytoprotection ,Cell biology ,ErbB Receptors ,Mice, Inbred C57BL ,Biochemistry ,Brain Injuries ,biology.protein ,Female ,Original Article - Abstract
Platelet activation is a frontline response to injury, not only essential for clot formation but also important for tissue repair. Indeed, the reparative influence of platelets has long been exploited therapeutically where application of platelet concentrates expedites wound recovery. Despite this, the mechanisms of platelet-triggered cytoprotection are poorly understood. Here, we show that activated platelets accumulate in the brain to exceptionally high levels following injury and release factors that potently protect neurons from apoptosis. Kinomic microarray and subsequent kinase inhibitor studies showed that platelet-based neuroprotection relies upon paracrine activation of the epidermal growth factor receptor (EGFR) and downstream DNA-dependent protein kinase (DNA-PK). This same anti-apoptotic cascade stimulated by activated platelets also provided chemo-resistance to several cancer cell types. Surprisingly, deep proteomic profiling of the platelet releasate failed to identify any known EGFR ligand, indicating that activated platelets release an atypical activator of the EGFR. This study is the first to formally associate platelet activation to EGFR/DNA-PK – an endogenous cytoprotective cascade.
- Published
- 2014
32. LysargiNase mirrors trypsin for protein C-terminal and methylation-site identification
- Author
-
Theodoros Goulas, Nestor Solis, Oded Kleifeld, Philipp F. Lange, Lindsay D. Rogers, Ulrich Eckhard, Christopher M. Overall, Pitter F. Huesgen, F. Xavier Gomis-Rüth, Alexander von Humboldt Foundation, Canadian Institutes of Health Research, Michael Smith Foundation for Health Research, Canada Foundation for Innovation, Ministerio de Ciencia y Tecnología (España), and Ministerio de Economía y Competitividad (España)
- Subjects
Proteomics ,Proteome ,Arginine ,Lysine ,Biology ,Methylation Site ,Methylation ,complex mixtures ,Biochemistry ,Substrate Specificity ,medicine ,Trypsin ,Amino Acid Sequence ,Methanosarcina acetivorans ,Molecular Biology ,Peptide sequence ,Metalloproteinase ,Cell Biology ,biology.organism_classification ,Molecular biology ,Methanosarcina ,Metalloproteases ,bacteria ,Protein Processing, Post-Translational ,Biotechnology ,medicine.drug - Abstract
To improve proteome coverage and protein C-terminal identification, we characterized the Methanosarcina acetivorans thermophilic proteinase LysargiNase, which cleaves before lysine and arginine up to 55 °C. Unlike trypsin, LysargiNase-generated peptides had N-terminal lysine or arginine residues and fragmented with b ion-dominated spectra. This improved protein C terminal-peptide identification and several arginine-rich phosphosite assignments. Notably, cleavage also occurred at methylated or dimethylated lysine and arginine, facilitating detection of these epigenetic modifications., The German Academic Exchange Service (DAAD) and the Michael Smith Foundation for Health Research (MSFHR) supported P.F.H. P.F.L. was supported by the Alexander von Humboldt Foundation, Breast Cancer Society of Canada and MSFHR; L.D.R. was supported by the Canadian Institutes for Health Research (CIHR) and MSFHR; and U.E. was supported by the MSFHR. This work was supported by a Canada Research Chair in Metalloproteinase Proteomics and Systems Biology (C.M.O.), a grant from the CIHR (grant number M.O.P. 111055) as well as by an Infrastructure Grant from MSFHR and the Canada Foundations for Innovation (C.M.O.). Further support was provided by the European Union FP7 program; Consolider Program of the Spanish Ministry of Science and Technology; and State Plan for Research in Science, Technology and Innovation of the Spanish Ministry of Economy and Competitiveness (F.X.G.-R.)
- Published
- 2014
33. X-ray Absorption Studies of Human Matrix Metalloproteinase-2 (MMP-2) Bound to a Highly Selective Mechanism-based Inhibitor
- Author
-
Stephen Brown, Lakshmi P. Kotra, Oded Kleifeld, Shahriar Mobashery, Rafael Fridman, M. Margarida Bernardo, David C. Gervasi, and Irit Sagi
- Subjects
chemistry.chemical_classification ,Gelatinases ,biology ,chemistry.chemical_element ,Active site ,Cell Biology ,Zinc ,Matrix metalloproteinase ,Biochemistry ,Extracellular matrix ,Enzyme activator ,Enzyme ,chemistry ,biology.protein ,Metalloprotein ,Molecular Biology - Abstract
Malignant tumors express high levels of zinc-dependent endopeptidases called matrix metalloproteinases (MMPs), which are thought to facilitate tumor metastasis and angiogenesis by hydrolyzing components of the extracellular matrix. Of these enzymes, gelatinases A (MMP-2) and B (MMP-9), have especially been implicated in malignant processes, and thus, they have been a target for drugs designed to block their activity. Therefore, understanding their molecular structure is key for a rational approach to inhibitor design. Here, we have conducted x-ray absorption spectroscopy of the full-length human MMP-2 in its latent, active, and inhibited states and report the structural changes at the zinc ion site upon enzyme activation and inhibition. We have also examined the molecular structure of MMP-2 in complex with SB-3CT, a recently reported novel mechanism-based synthetic inhibitor that was designed to be highly selective in gelatinases (1). It is shown that SB-3CT directly binds the catalytic zinc ion of MMP-2. Interestingly, the novel mode of binding of the inhibitor to the catalytic zinc reconstructs the conformational environment around the active site metal ion back to that of the proenzyme.
- Published
- 2001
34. Reversible 26S proteasome disassembly upon mitochondrial stress
- Author
-
Thorsten Hoppe, Michael H. Glickman, Oded Kleifeld, Zoi Erpapazoglou, Daria Krutauz, Nurit Livnat-Levanon, Mickael M. Cohen, Alexandra Segref, Éva Kevei, Noa Reis, Teresa Rinaldi, Technion - Israel Institute of Technology [Haifa], Universität zu Köln, Monash University [Clayton], Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome], Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes (LBMCE), Université Pierre et Marie Curie - Paris 6 (UPMC)-Centre National de la Recherche Scientifique (CNRS), Universität zu Köln = University of Cologne, and Università degli Studi di Roma 'La Sapienza' = Sapienza University [Rome] (UNIROMA)
- Subjects
Proteasome Endopeptidase Complex ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Saccharomyces cerevisiae ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Antioxidants ,Cell Line ,stress ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Ubiquitinated Protein Degradation ,Holoenzymes ,In vivo ,Cricetinae ,medicine ,Animals ,Cysteine ,Caenorhabditis elegans ,Molecular Biology ,lcsh:QH301-705.5 ,Cysteine metabolism ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Reactive oxygen species ,Ubiquitin ,proteasome ,mitochondria ,Cell Biology ,In vitro ,Cell biology ,Mitochondria ,Oxidative Stress ,Proteasome ,chemistry ,lcsh:Biology (General) ,Molecular Medicine ,Protein Multimerization ,Reactive Oxygen Species ,030217 neurology & neurosurgery ,Oxidative stress - Abstract
International audience; In eukaryotic cells, proteasomes exist primarily as 26S holoenzymes, the most efficient configuration for ubiquitinated protein degradation. Here, we show that acute oxidative stress caused by environmental insults or mitochondrial defects results in rapid disassembly of 26S proteasomes into intact 20S core and 19S regulatory particles. Consequently, polyubiquitinated substrates accumulate, mito-chondrial networks fragment, and cellular reactive oxygen species (ROS) levels increase. Oxidation of cysteine residues is sufficient to induce proteasome disassembly, and spontaneous reassembly from existing components is observed both in vivo and in vitro upon reduction. Ubiquitin-dependent sub-strate turnover also resumes after treatment with antioxidants. Reversible attenuation of 26S protea-some activity induced by acute mitochondrial or oxidative stress may be a short-term response distinct from adaptation to long-term ROS exposure or changes during aging.
- Published
- 2013
35. Structural Characterization of the Catalytic Active Site in the Latent and Active Natural Gelatinase B from Human Neutrophils
- Author
-
Feng Cheng, Anatoly I. Frenkel, Oded Kleifeld, Ghislain Opdenakker, Philippe E. Van den Steen, Irit Sagi, and Hua-Liang Jiang
- Subjects
Binding Sites ,biology ,Neutrophils ,Protein Conformation ,Chemistry ,Active site ,chemistry.chemical_element ,Cell Biology ,Zinc ,Matrix metalloproteinase ,Biochemistry ,Catalysis ,Enzyme activator ,Protein structure ,Matrix Metalloproteinase 9 ,Catalytic Domain ,biology.protein ,Biophysics ,Humans ,Thermodynamics ,Gelatinase ,Binding site ,Molecular Biology ,Electron Probe Microanalysis - Abstract
Matrix metalloproteinases are endopeptidases that have a leading role in the catabolism of the macromolecular components of the extracellular matrix in a variety of normal and pathological processes. Human gelatinase B is a zinc-dependent proteinase and a member of the matrix metalloproteinase family that is involved in inflammation, tissue remodeling, and cancer. We have conducted x-ray absorption spectroscopy, atomic emission, and quantum mechanics studies of natural and activated human gelatinase B. Our results show that the natural enzyme contains one catalytic zinc ion that is central to catalysis. In addition, upon enzyme activation, the catalytic zinc site exhibits a conformation change that results in the expansion of the bond distances around the zinc ion and the replacement of one sulfur with oxygen. Interestingly, quantum mechanics calculations show that oxygen ligation at the catalytic zinc ion exhibits a greater affinity to the binding of an oxygen from an amino acid residue rather than from an external water molecule. These results suggest that the catalytic zinc ion plays a key role in both substrate binding and catalysis.
- Published
- 2000
36. Dual function of Rpn5 in two PCI complexes, the 26S proteasome and COP9 signalosome
- Author
-
Kay Hofmann, Avigail Lande-Atir, Oded Kleifeld, Daria Krutauz, Maya Kleiman, Elah Pick, Maisa Bsoul, Noa Reis, Zanlin Yu, Richard D. Vierstra, Michael H. Glickman, and Adam J. Book
- Subjects
Proteasome Endopeptidase Complex ,Saccharomyces cerevisiae Proteins ,Protein subunit ,Saccharomyces cerevisiae ,Biosynthesis and Biodegradation ,Biology ,NEDD8 ,Eukaryotic translation ,Initiation factor ,COP9 signalosome ,Molecular Biology ,COP9 Signalosome Complex ,Cell Biology ,Articles ,biology.organism_classification ,Cullin Proteins ,eye diseases ,Molecular Weight ,Protein Subunits ,Phenotype ,Proteasome ,Biochemistry ,Multiprotein Complexes ,Peptide Hydrolases - Abstract
Functional redundancy of Rpn5 in budding yeast allows its participation and function in two distinct but structurally related PCI complexes: the proteasome lid and the CSN. As a lid subunit, Rpn5 stabilizes proteasome integrity; as a CSN subunit, Rpn5 is required for enzymatic hydrolysis of Rub1/Nedd8 from cullins., Subunit composition and architectural structure of the 26S proteasome lid is strictly conserved between all eukaryotes. This eight-subunit complex bears high similarity to the eukaryotic translation initiation factor 3 and to the COP9 signalosome (CSN), which together define the proteasome CSN/COP9/initiation factor (PCI) troika. In some unicellular eukaryotes, the latter two complexes lack key subunits, encouraging questions about the conservation of their structural design. Here we demonstrate that, in Saccharomyces cerevisiae, Rpn5 plays dual roles by stabilizing proteasome and CSN structures independently. Proteasome and CSN complexes are easily dissected, with Rpn5 the only subunit in common. Together with Rpn5, we identified a total of six bona fide subunits at roughly stoichiometric ratios in isolated, affinity-purified CSN. Moreover, the copy of Rpn5 associated with the CSN is required for enzymatic hydrolysis of Rub1/Nedd8 conjugated to cullins. We propose that multitasking by a single subunit, Rpn5 in this case, allows it to function in different complexes simultaneously. These observations demonstrate that functional substitution of subunits by paralogues is feasible, implying that the canonical composition of the three PCI complexes in S. cerevisiae is more robust than hitherto appreciated.
- Published
- 2011
37. The eIF3 interactome reveals the translasome, a supercomplex linking protein synthesis and degradation machineries
- Author
-
Rodrigo Cabrera, Eric C. Chang, Laurence M. Brill, Judith S. Scheliga, Oded Kleifeld, Dieter A. Wolf, Michael H. Glickman, and Zhe Sha
- Subjects
Models, Molecular ,Proteasome Endopeptidase Complex ,Eukaryotic Initiation Factor-3 ,Active Transport, Cell Nucleus ,Ribosome biogenesis ,Biology ,Interactome ,Article ,Eukaryotic translation ,Ribosomal protein ,Tandem Mass Spectrometry ,Protein Interaction Mapping ,Schizosaccharomyces ,Ribosome Subunits ,Initiation factor ,Molecular Biology ,Cell Biology ,beta Karyopherins ,Cell biology ,Enzymes ,Elongation factor ,Actin Cytoskeleton ,Multiprotein Complexes ,Protein Biosynthesis ,Transfer RNA ,Schizosaccharomyces pombe Proteins - Abstract
Summary eIF3 promotes translation initiation, but relatively little is known about its full range of activities in the cell. Here, we employed affinity purification and highly sensitive LC-MS/MS to decipher the fission yeast eIF3 interactome, which was found to contain 230 proteins. eIF3 assembles into a large supercomplex, the translasome, which contains elongation factors, tRNA synthetases, 40S and 60S ribosomal proteins, chaperones, and the proteasome. eIF3 also associates with ribosome biogenesis factors and the importins-β Kap123p and Sal3p. Our genetic data indicated that the binding to both importins-β is essential for cell growth, and photobleaching experiments revealed a critical role for Sal3p in the nuclear import of one of the translasome constituents, the proteasome. Our data reveal the breadth of the eIF3 interactome and suggest that factors involved in translation initiation, ribosome biogenesis, translation elongation, quality control, and transport are physically linked to facilitate efficient protein synthesis.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.