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










Publication year range
1.
Aging Cell ; 23(5): e14128, 2024 05.
Article in English | MEDLINE | ID: mdl-38415292

ABSTRACT

Parkinson's disease (PD) is characterized by aggregation of α-synuclein (α-syn) into protein inclusions in degenerating brains. Increasing amounts of aggregated α-syn species indicate significant perturbation of cellular proteostasis. Altered proteostasis depends on α-syn protein levels and the impact of α-syn on other components of the proteostasis network. Budding yeast Saccharomyces cerevisiae was used as eukaryotic reference organism to study the consequences of α-syn expression on protein dynamics. To address this, we investigated the impact of overexpression of α-syn and S129A variant on the abundance and stability of most yeast proteins using a genome-wide yeast library and a tandem fluorescent protein timer (tFT) reporter as a measure for protein stability. This revealed that the stability of in total 377 cellular proteins was altered by α-syn expression, and that the impact on protein stability was significantly enhanced by phosphorylation at Ser129 (pS129). The proteasome assembly chaperone Rpn14 was identified as one of the top candidates for increased protein stability by expression of pS129 α-syn. Elevated levels of Rpn14 enhanced the growth inhibition by α-syn and the accumulation of ubiquitin conjugates in the cell. We found that Rpn14 interacts physically with α-syn and stabilizes pS129 α-syn. The expression of α-syn along with elevated levels of Rpn14 or its human counterpart PAAF1 reduced the proteasome activity in yeast and in human cells, supporting that pS129 α-syn negatively affects the 26S proteasome through Rpn14. This comprehensive study into the alternations of protein homeostasis highlights the critical role of the Rpn14/PAAF1 in α-syn-mediated proteasome dysfunction.


Subject(s)
Proteasome Endopeptidase Complex , Saccharomyces cerevisiae , alpha-Synuclein , alpha-Synuclein/metabolism , Proteasome Endopeptidase Complex/metabolism , Humans , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Molecular Chaperones/metabolism , Parkinson Disease/metabolism , Parkinson Disease/genetics , Parkinson Disease/pathology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics
2.
Nat Chem ; 16(3): 363-372, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38326645

ABSTRACT

The human enzyme p97 regulates various cellular pathways by unfolding hundreds of protein substrates in an ATP-dependent manner, making it an essential component of protein homeostasis and an impactful pharmacological target. The hexameric complex undergoes substantial conformational changes throughout its catalytic cycle. Here we elucidate the molecular motions that occur at the active site in the temporal window immediately before and after ATP hydrolysis by merging cryo-EM, NMR spectroscopy and molecular dynamics simulations. p97 populates a metastable reaction intermediate, the ADP·Pi state, which is poised between hydrolysis and product release. Detailed snapshots reveal that the active site is finely tuned to trap and eventually discharge the cleaved phosphate. Signalling pathways originating at the active site coordinate the action of the hexamer subunits and couple hydrolysis with allosteric conformational changes. Our multidisciplinary approach enables a glimpse into the sophisticated spatial and temporal orchestration of ATP handling by a prototype AAA+ protein.


Subject(s)
Adenosine Triphosphatases , Adenosine Triphosphate , Humans , Adenosine Triphosphate/chemistry , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Valosin Containing Protein , Molecular Dynamics Simulation
3.
Anal Chem ; 94(51): 17751-17756, 2022 12 27.
Article in English | MEDLINE | ID: mdl-36510358

ABSTRACT

Cross-linking mass spectrometry (XL-MS) has become an indispensable tool for the emerging field of systems structural biology over the recent years. However, the confidence in individual protein-protein interactions (PPIs) depends on the correct assessment of individual inter-protein cross-links. In this article, we describe a mono- and intralink filter (mi-filter) that is applicable to any kind of cross-linking data and workflow. It stipulates that only proteins for which at least one monolink or intra-protein cross-link has been identified within a given data set are considered for an inter-protein cross-link and therefore participate in a PPI. We show that this simple and intuitive filter has a dramatic effect on different types of cross-linking data ranging from individual protein complexes over medium-complexity affinity enrichments to proteome-wide cell lysates and significantly reduces the number of false-positive identifications for inter-protein links in all these types of XL-MS data.


Subject(s)
Proteome , Mass Spectrometry , Proteome/chemistry , Cross-Linking Reagents/chemistry
4.
Nat Commun ; 13(1): 838, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35149681

ABSTRACT

The proteasome recognizes ubiquitinated proteins and can also edit ubiquitin marks, allowing substrates to be rejected based on ubiquitin chain topology. In yeast, editing is mediated by deubiquitinating enzyme Ubp6. The proteasome activates Ubp6, whereas Ubp6 inhibits the proteasome through deubiquitination and a noncatalytic effect. Here, we report cryo-EM structures of the proteasome bound to Ubp6, based on which we identify mutants in Ubp6 and proteasome subunit Rpt1 that abrogate Ubp6 activation. The Ubp6 mutations define a conserved region that we term the ILR element. The ILR is found within the BL1 loop, which obstructs the catalytic groove in free Ubp6. Rpt1-ILR interaction opens the groove by rearranging not only BL1 but also a previously undescribed network of three interconnected active-site-blocking loops. Ubp6 activation and noncatalytic proteasome inhibition are linked in that they are eliminated by the same mutations. Ubp6 and ubiquitin together drive proteasomes into a unique conformation associated with proteasome inhibition. Thus, a multicomponent allosteric switch exerts simultaneous control over both Ubp6 and the proteasome.


Subject(s)
Endopeptidases/chemistry , Endopeptidases/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Catalytic Domain , Cryoelectron Microscopy , Cytoplasm , Endopeptidases/genetics , Proteasome Endopeptidase Complex/genetics , Protein Conformation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin/metabolism , Ubiquitinated Proteins/metabolism
5.
Biochim Biophys Acta Proteins Proteom ; 1869(3): 140583, 2021 03.
Article in English | MEDLINE | ID: mdl-33321258

ABSTRACT

In eukaryotic cells, the ubiquitin-proteasome system serves to remove proteins that are either dysfunctional or no longer needed. The 26S proteasome is a 2.5 MDa multisubunit complex comprising the 20S core particle, where degradation is executed, and one or two regulatory particles which prepare substrates for degradation. Whereas the 20S core particles of several species had been studied extensively by X-ray crystallography, the 26S holocomplex structure had remained elusive for a long time. Recent advances in single-particle cryo-electron microscopy have changed the situation and provided atomic resolution models of this intriguing molecular machine and its dynamics. Besides, cryo-electron tomography enables structural studies in situ, providing molecular resolution images of macromolecules inside pristinely preserved cellular environments. This has greatly contributed to our understanding of proteasome dynamics in the context of cells.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Cryoelectron Microscopy , Crystallography, X-Ray , Proteasome Endopeptidase Complex/chemistry , Protein Conformation , Subcellular Fractions/metabolism
6.
Cell Rep ; 24(5): 1301-1315.e5, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30067984

ABSTRACT

The proteasome is the central protease for intracellular protein breakdown. Coordinated binding and hydrolysis of ATP by the six proteasomal ATPase subunits induces conformational changes that drive the unfolding and translocation of substrates into the proteolytic 20S core particle for degradation. Here, we combine genetic and biochemical approaches with cryo-electron microscopy and integrative modeling to dissect the relationship between individual nucleotide binding events and proteasome conformational dynamics. We demonstrate unique impacts of ATP binding by individual ATPases on the proteasome conformational distribution and report two conformational states of the proteasome suggestive of a rotary ATP hydrolysis mechanism. These structures, coupled with functional analyses, reveal key roles for the ATPases Rpt1 and Rpt6 in gating substrate entry into the core particle. This deepened knowledge of proteasome conformational dynamics reveals key elements of intersubunit communication within the proteasome and clarifies the regulation of substrate entry into the proteolytic chamber.


Subject(s)
Molecular Dynamics Simulation , Proteasome Endopeptidase Complex/chemistry , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism
7.
Mol Cell Proteomics ; 16(5): 840-854, 2017 05.
Article in English | MEDLINE | ID: mdl-28292943

ABSTRACT

The 26S proteasome is the macromolecular machine responsible for ATP/ubiquitin dependent degradation. As aberration in proteasomal degradation has been implicated in many human diseases, structural analysis of the human 26S proteasome complex is essential to advance our understanding of its action and regulation mechanisms. In recent years, cross-linking mass spectrometry (XL-MS) has emerged as a powerful tool for elucidating structural topologies of large protein assemblies, with its unique capability of studying protein complexes in cells. To facilitate the identification of cross-linked peptides, we have previously developed a robust amine reactive sulfoxide-containing MS-cleavable cross-linker, disuccinimidyl sulfoxide (DSSO). To better understand the structure and regulation of the human 26S proteasome, we have established new DSSO-based in vivo and in vitro XL-MS workflows by coupling with HB-tag based affinity purification to comprehensively examine protein-protein interactions within the 26S proteasome. In total, we have identified 447 unique lysine-to-lysine linkages delineating 67 interprotein and 26 intraprotein interactions, representing the largest cross-link dataset for proteasome complexes. In combination with EM maps and computational modeling, the architecture of the 26S proteasome was determined to infer its structural dynamics. In particular, three proteasome subunits Rpn1, Rpn6, and Rpt6 displayed multiple conformations that have not been previously reported. Additionally, cross-links between proteasome subunits and 15 proteasome interacting proteins including 9 known and 6 novel ones have been determined to demonstrate their physical interactions at the amino acid level. Our results have provided new insights on the dynamics of the 26S human proteasome and the methodologies presented here can be applied to study other protein complexes.


Subject(s)
Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Cell Line , Humans , Models, Molecular , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps , Reproducibility of Results , Tandem Mass Spectrometry
8.
Proc Natl Acad Sci U S A ; 114(6): 1305-1310, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28115689

ABSTRACT

In eukaryotic cells, the ubiquitin-proteasome system (UPS) is responsible for the regulated degradation of intracellular proteins. The 26S holocomplex comprises the core particle (CP), where proteolysis takes place, and one or two regulatory particles (RPs). The base of the RP is formed by a heterohexameric AAA+ ATPase module, which unfolds and translocates substrates into the CP. Applying single-particle cryo-electron microscopy (cryo-EM) and image classification to samples in the presence of different nucleotides and nucleotide analogs, we were able to observe four distinct conformational states (s1 to s4). The resolution of the four conformers allowed for the construction of atomic models of the AAA+ ATPase module as it progresses through the functional cycle. In a hitherto unobserved state (s4), the gate controlling access to the CP is open. The structures described in this study allow us to put forward a model for the 26S functional cycle driven by ATP hydrolysis.


Subject(s)
Adenosine Triphosphatases/chemistry , Models, Molecular , Proteasome Endopeptidase Complex/chemistry , Cryoelectron Microscopy , Nucleotides/chemistry , Proteasome Endopeptidase Complex/ultrastructure , Protein Conformation
9.
Int J Biochem Cell Biol ; 79: 437-442, 2016 10.
Article in English | MEDLINE | ID: mdl-27498189

ABSTRACT

There is growing appreciation for the fundamental role of structural dynamics in the function of macromolecules. In particular, the 26S proteasome, responsible for selective protein degradation in an ATP dependent manner, exhibits dynamic conformational changes that enable substrate processing. Recent cryo-electron microscopy (cryo-EM) work has revealed the conformational dynamics of the 26S proteasome and established the function of the different conformational states. Technological advances such as direct electron detectors and image processing algorithms allowed resolving the structure of the proteasome at atomic resolution. Here we will review those studies and discuss their contribution to our understanding of proteasome function.


Subject(s)
Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Animals , Humans
10.
Proc Natl Acad Sci U S A ; 113(28): 7816-21, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27342858

ABSTRACT

Protein degradation in eukaryotic cells is performed by the Ubiquitin-Proteasome System (UPS). The 26S proteasome holocomplex consists of a core particle (CP) that proteolytically degrades polyubiquitylated proteins, and a regulatory particle (RP) containing the AAA-ATPase module. This module controls access to the proteolytic chamber inside the CP and is surrounded by non-ATPase subunits (Rpns) that recognize substrates and deubiquitylate them before unfolding and degradation. The architecture of the 26S holocomplex is highly conserved between yeast and humans. The structure of the human 26S holocomplex described here reveals previously unidentified features of the AAA-ATPase heterohexamer. One subunit, Rpt6, has ADP bound, whereas the other five have ATP in their binding pockets. Rpt6 is structurally distinct from the other five Rpt subunits, most notably in its pore loop region. For Rpns, the map reveals two main, previously undetected, features: the C terminus of Rpn3 protrudes into the mouth of the ATPase ring; and Rpn1 and Rpn2, the largest proteasome subunits, are linked by an extended connection. The structural features of the 26S proteasome observed in this study are likely to be important for coordinating the proteasomal subunits during substrate processing.


Subject(s)
Models, Molecular , Proteasome Endopeptidase Complex/chemistry , Humans , Microscopy, Electron, Transmission , Proteasome Endopeptidase Complex/isolation & purification , Proteasome Endopeptidase Complex/metabolism , Protein Conformation , Yeasts
11.
Proc Natl Acad Sci U S A ; 112(28): 8626-31, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26130806

ABSTRACT

In eukaryotic cells, the 26S proteasome is responsible for the regulated degradation of intracellular proteins. Several cofactors interact transiently with this large macromolecular machine and modulate its function. The deubiquitylating enzyme ubiquitin C-terminal hydrolase 6 [Ubp6; ubiquitin-specific protease (USP) 14 in mammals] is the most abundant proteasome-interacting protein and has multiple roles in regulating proteasome function. Here, we investigate the structural basis of the interaction between Ubp6 and the 26S proteasome in the presence and absence of the inhibitor ubiquitin aldehyde. To this end we have used single-particle electron cryomicroscopy in combination with cross-linking and mass spectrometry. Ubp6 binds to the regulatory particle non-ATPase (Rpn) 1 via its N-terminal ubiquitin-like domain, whereas its catalytic USP domain is positioned variably. Addition of ubiquitin aldehyde stabilizes the binding of the USP domain in a position where it bridges the proteasome subunits Rpn1 and the regulatory particle triple-A ATPase (Rpt) 1. The USP domain binds to Rpt1 in the immediate vicinity of the Ubp6 active site, which may effect its activation. The catalytic triad is positioned in proximity to the mouth of the ATPase module and to the deubiquitylating enzyme Rpn11, strongly implying their functional linkage. On the proteasome side, binding of Ubp6 favors conformational switching of the 26S proteasome into an intermediate-energy conformational state, in particular upon the addition of ubiquitin aldehyde. This modulation of the conformational space of the 26S proteasome by Ubp6 explains the effects of Ubp6 on the kinetics of proteasomal degradation.


Subject(s)
Endopeptidases/metabolism , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Catalytic Domain , Cryoelectron Microscopy , Endopeptidases/chemistry , Proteasome Endopeptidase Complex/chemistry , Protein Binding , Protein Conformation , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry
12.
FEBS Lett ; 588(23): 4422-30, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25448680

ABSTRACT

Ataxin-3, which is encoded by a gene that has been associated with Machado-Joseph disease, contains a catalytic N-terminal Josephin domain with deubiquitinase activity. Here, we show that the Josephin domain of ataxin 3 catalyzes endo-type cleavage of Lys48-linked polyubiquitin. Furthermore, NMR data obtained following site-specific paramagnetic spin labeling of Lys48-linked di-ubiquitin revealed that both ubiquitin units interact with the Josephin domain, with the C-terminal Gly76 of the proximal unit being situated in the vicinity of the catalytic triad of Josephin domain. Our results help to elucidate how the substrate is recognized by the Josephin domain and properly positioned for an endo-type deubiquitination reaction.


Subject(s)
Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Ubiquitin-Specific Proteases/metabolism , Ataxin-3 , Humans , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Ubiquitin/chemistry , Ubiquitin/metabolism
13.
Nat Commun ; 5: 3396, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24598877

ABSTRACT

The 26S proteasome is a 2.5-MDa multisubunit protease complex that degrades polyubiquitylated proteins. Although its functions and structure have been extensively characterized, little is known about its dynamics in living cells. Here, we investigate the absolute concentration, spatio-temporal dynamics and complex formation of the proteasome in living cells using fluorescence correlation spectroscopy. We find that the 26S proteasome complex is highly mobile, and that almost all proteasome subunits throughout the cell are stably incorporated into 26S proteasomes. The interaction between 19S and 20S particles is stable even in an importin-α mutant, suggesting that the 26S proteasome is assembled in the cytoplasm. Furthermore, a genetically stabilized 26S proteasome mutant is able to enter the nucleus. These results suggest that the 26S proteasome completes its assembly process in the cytoplasm and translocates into the nucleus through the nuclear pore complex as a holoenzyme.


Subject(s)
Cytoplasm/metabolism , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Cryoelectron Microscopy , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Mutation , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Time Factors , Time-Lapse Imaging/methods , Red Fluorescent Protein
14.
J Cell Biol ; 201(5): 725-40, 2013 May 27.
Article in English | MEDLINE | ID: mdl-23712261

ABSTRACT

Synaptic vesicles are embedded in a complex filamentous network at the presynaptic terminal. Before fusion, vesicles are linked to the active zone (AZ) by short filaments (tethers). The identity of the molecules that form and regulate tethers remains unknown, but Rab3-interacting molecule (RIM) is a prominent candidate, given its central role in AZ organization. In this paper, we analyzed presynaptic architecture of RIM1α knockout (KO) mice by cryo-electron tomography. In stark contrast to previous work on dehydrated, chemically fixed samples, our data show significant alterations in vesicle distribution and AZ tethering that could provide a structural basis for the functional deficits of RIM1α KO synapses. Proteasome inhibition reversed these structural defects, suggesting a functional recovery confirmed by electrophysiological recordings. Altogether, our results not only point to the ubiquitin-proteasome system as an important regulator of presynaptic architecture and function but also show that the tethering machinery plays a critical role in exocytosis, converging into a structural model of synaptic vesicle priming by RIM1α.


Subject(s)
GTP-Binding Proteins/physiology , Synaptic Vesicles/metabolism , Animals , Exocytosis/drug effects , GTP-Binding Proteins/genetics , Leupeptins/pharmacology , Membrane Fusion/drug effects , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/physiology , Proteasome Inhibitors/pharmacology , Synaptic Vesicles/drug effects , Synaptic Vesicles/ultrastructure , Tomography/methods
15.
Biochem Biophys Res Commun ; 435(2): 250-4, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23643786

ABSTRACT

The ubiquitin-proteasome system is responsible for regulated protein degradation in the cell with the 26S proteasome acting as its executive arm. The molecular architecture of this 2.5 MDa complex has been established recently, with the notable exception of the small acidic subunit Sem1. Here, we localize the C-terminal helix of Sem1 binding to the PCI domain of the subunit Rpn7 using cryo-electron microscopy single particle reconstruction of proteasomes purified from yeast cells with sem1 deletion. The approximate position of the N-terminal region of Sem1 bridging the cleft between Rpn7 and Rpn3 was inferred based on site-specific cross-linking data of the 26S proteasome. Our structural studies indicate that Sem1 can assume different conformations in different contexts, which supports the idea that Sem1 functions as a molecular glue stabilizing the Rpn3/Rpn7 heterodimer.


Subject(s)
Proteasome Endopeptidase Complex/chemistry , Regulatory Elements, Transcriptional , Saccharomyces cerevisiae Proteins/chemistry , Amino Acid Sequence , Molecular Sequence Data , Protein Subunits/chemistry
16.
Biochem Biophys Res Commun ; 432(1): 141-5, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23376067

ABSTRACT

A major form of proteasome activator PA28 is a heteroheptamer composed of interferon-γ-inducible α and ß subunits, which share approximately 50% amino acid identity and possess distinct insert loops. This activator forms a complex with the 20S proteasome and thereby stimulates proteasomal degradation of peptides in an ATP-independent manner, giving rise to smaller antigenic peptides presented by major histocompatibility complex class I molecules. In this study, we performed biophysical and biochemical characterization of the structure and function of the PA28 hetero-oligomer. Deuteration-assisted small-angle neutron scattering demonstrated three α and four ß subunits are alternately arranged in the heptameric ring. In this arrangement, PA28 loops surround the central pore of the heptameric ring (site for peptide entry). Activating the 20S proteasome with a PA28 mutant that lacked the α subunit loops cleaved model substrates longer than a nonapeptide with better efficiency when compared to wild-type PA28. Based on these data, we hypothesize that the flexible PA28 loops act as gatekeepers, which function to select the length of peptide substrates to be transported between the proteolytic chamber and the extra-proteasomal medium.


Subject(s)
Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Animals , Mice , Peptides/chemistry , Protein Conformation , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/metabolism , Proteolysis
17.
Proc Natl Acad Sci U S A ; 109(37): 14870-5, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22927375

ABSTRACT

The 26S proteasome operates at the executive end of the ubiquitin-proteasome pathway. Here, we present a cryo-EM structure of the Saccharomyces cerevisiae 26S proteasome at a resolution of 7.4 Å or 6.7 Å (Fourier-Shell Correlation of 0.5 or 0.3, respectively). We used this map in conjunction with molecular dynamics-based flexible fitting to build a near-atomic resolution model of the holocomplex. The quality of the map allowed us to assign α-helices, the predominant secondary structure element of the regulatory particle subunits, throughout the entire map. We were able to determine the architecture of the Rpn8/Rpn11 heterodimer, which had hitherto remained elusive. The MPN domain of Rpn11 is positioned directly above the AAA-ATPase N-ring suggesting that Rpn11 deubiquitylates substrates immediately following commitment and prior to their unfolding by the AAA-ATPase module. The MPN domain of Rpn11 dimerizes with that of Rpn8 and the C-termini of both subunits form long helices, which are integral parts of a coiled-coil module. Together with the C-terminal helices of the six PCI-domain subunits they form a very large coiled-coil bundle, which appears to serve as a flexible anchoring device for all the lid subunits.


Subject(s)
Endopeptidases/chemistry , Models, Molecular , Proteasome Endopeptidase Complex/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Cryoelectron Microscopy , Molecular Dynamics Simulation , Protein Structure, Tertiary
18.
Proc Natl Acad Sci U S A ; 109(5): 1479-84, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22215586

ABSTRACT

Two canonical subunits of the 26S proteasome, Rpn10 and Rpn13, function as ubiquitin (Ub) receptors. The mutual arrangement of these subunits--and all other non-ATPase subunits--in the regulatory particle is unknown. Using electron cryomicroscopy, we calculated difference maps between wild-type 26S proteasome from Saccharomyces cerevisiae and deletion mutants (rpn10Δ, rpn13Δ, and rpn10Δrpn13Δ). These maps allowed us to localize the two Ub receptors unambiguously. Rpn10 and Rpn13 mapped to the apical part of the 26S proteasome, above the N-terminal coiled coils of the AAA-ATPase heterodimers Rpt4/Rpt5 and Rpt1/Rpt2, respectively. On the basis of the mutual positions of Rpn10 and Rpn13, we propose a model for polyubiquitin binding to the 26S proteasome.


Subject(s)
Cryoelectron Microscopy/methods , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Animals , Drosophila melanogaster , Mass Spectrometry , Models, Molecular
19.
Biomol NMR Assign ; 6(2): 177-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22127525

ABSTRACT

HOIL-1L and its binding partner, HOIL-1L interacting protein (HOIP), are essential components of linear ubiquitin (Ub) chain assembly complex (LUBAC), a 600-kDa enzyme complex catalyzing elongation of a tandemly connected Ub chain, which serve as a regulator of NF-κB activation. Specific interaction between the N-terminal Ub-like domain (UBL) of HOIL-1L and the Ub-associated domain (UBA) located at the central region of HOIP is shown to be involved in the formation of LUBAC. For better understanding of the mechanisms underlying the generation of the linear Ub chains by LUBAC, it is necessary to characterize the UBL-UBA interaction on the basis of structural data, which, however, is not available to date. Here we report backbone and side chain NMR assignments of the UBL of human HOIL-1L. By inspection of chemical shift index, it was predicted that HOIL-1L-UBL assumes a Ub fold followed by an α-helical segment, offering the basis for determination its 3D structure and interaction with HOIP-UBA in solution.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular , Protons , Ubiquitin-Protein Ligases/chemistry , Ubiquitin/chemistry , Ubiquitin/metabolism , Amino Acid Sequence , Carbon Isotopes , Humans , Molecular Sequence Data , Nitrogen Isotopes , Protein Structure, Tertiary , Transcription Factors
20.
Mol Cell ; 42(5): 637-49, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21658604

ABSTRACT

The 26S proteasome is a 2.5 MDa macromolecular machine responsible for targeted protein degradation. Recently, four chaperones were identified that promote the assembly of the 19S regulatory particle (RP). Here, we probe the dynamic architecture of the proteasome by applying quantitative proteomics and mass spectrometry (MS) of intact complexes to provide a detailed characterization of how Ubp6 assists this assembly process. Our MS data demonstrate stoichiometric binding of chaperones and Ubp6 to the basal part of the RP. Genetic interactions of Ubp6 with Hsm3, but not with the other chaperones, indicate a functional overlay with Hsm3. Our biochemical data identified Ubp6 as an additional member of the Hsm3 module. Deletions of ubp6 with hsm3 perturb 26S proteasome assembly, which we attribute to an accumulation of ubiquitylated substrates on these assembly precursors. We therefore propose that Ubp6 facilitates proteasomal assembly by clearing ubiquitylated substrates from assembly precursors by its deubiquitylating activity.


Subject(s)
Endopeptidases/physiology , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/metabolism , Adenosine Triphosphatases/metabolism , Endopeptidases/chemistry , Endopeptidases/metabolism , Gene Deletion , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/isolation & purification , Proteasome Endopeptidase Complex/metabolism , Protein Interaction Mapping , Proteomics/methods , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...