Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Chem Biol ; 19(1): 55-63, 2023 01.
Article in English | MEDLINE | ID: mdl-36577875

ABSTRACT

Engineered destruction of target proteins by recruitment to the cell's degradation machinery has emerged as a promising strategy in drug discovery. The majority of molecules that facilitate targeted degradation do so via a select number of ubiquitin ligases, restricting this therapeutic approach to tissue types that express the requisite ligase. Here, we describe a new strategy of targeted protein degradation through direct substrate recruitment to the 26S proteasome. The proteolytic complex is essential and abundantly expressed in all cells; however, proteasomal ligands remain scarce. We identify potent peptidic macrocycles that bind directly to the 26S proteasome subunit PSMD2, with a 2.5-Å-resolution cryo-electron microscopy complex structure revealing a binding site near the 26S pore. Conjugation of this macrocycle to a potent BRD4 ligand enabled generation of chimeric molecules that effectively degrade BRD4 in cells, thus demonstrating that degradation via direct proteasomal recruitment is a viable strategy for targeted protein degradation.


Subject(s)
Nuclear Proteins , Transcription Factors , Nuclear Proteins/metabolism , Cryoelectron Microscopy , Transcription Factors/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism
2.
Curr Microbiol ; 80(2): 56, 2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36585971

ABSTRACT

The dynamic microflora associated within, and in the surrounding aquatic environment, has been found to be responsible for the functional properties of many aquatic plants. The aim of the current work was to evaluate the effectiveness of Lemnaceae-based wastewater treatment system under tropical conditions and investigate the changes in the aquatic microflora upon plant growth. A biological wastewater treatment system was designed and investigated using mixed Lemnaceae culture comprising Lemna minor and Spirodela polyrhiza in a batch mode. A significant reduction in total solids (31.8%), biochemical oxygen demand (93.5%), and chemical oxygen demand (73.2%) was observed after seven days of duckweed growth using a low inoculum. A preliminary study on the change in the microbial population diversity and functionality, in the wastewater before and after treatment, revealed an increase in the denitrifying microflora in wastewater post-Lemnaceae treatment. Dominance of 10 bacterial phyla, contributing for 98.3% of the total bacterial communities, was recorded, and ~ 50.6% loss of diversity post-treatment of wastewater was revealed by the Shannon Index. Among 16 bacterial families showing relative abundance of ≥ 1% in untreated wastewater, Methylobacteriaceae, Pseudomonadaceae, Brucellaceae, Rhodobacteraceae, and Acetobacteraceae prevailed in the water post-treatment by duckweeds. This is a novel work done on the dynamics of aquatic microflora associated with Lemnaceae under tropical Indian conditions. It confirms the application of Lemnaceae-based wastewater treatment system as effective biofilter and calls for further studies on the active involvement of the endophytic and aquatic microflora in the functions of these plant.


Subject(s)
Araceae , Water Purification , Humans , Wastewater , Araceae/chemistry , Araceae/microbiology , Plants , Bacteria , Population Dynamics
3.
J Am Chem Soc ; 143(28): 10571-10575, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34236858

ABSTRACT

We hypothesized that the proximity-driven ubiquitylation of E3-interacting small molecules could affect the degradation of E3 ubiquitin ligases. A series of XIAP BIR2 domain-binding small molecules was modified to append a nucleophilic primary amine. This modification transforms XIAP binders into inducers of XIAP degradation. The degradation of XIAP is E1- and proteasome-dependent, dependent on the ligase function of XIAP, and is rescued by subtle modifications of the small molecule that would obviate ubiquitylation. We demonstrate in vitro ubiquitylation of the small molecule that is dependent on its interaction with XIAP. Taken together, these results demonstrate the designed ubiquitylation of an engineered small molecule and a novel approach for the degradation of E3 ubiquitin ligases.


Subject(s)
Amines/pharmacology , Small Molecule Libraries/pharmacology , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Amines/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemistry , X-Linked Inhibitor of Apoptosis Protein/metabolism
4.
Cell Death Differ ; 28(2): 591-605, 2021 02.
Article in English | MEDLINE | ID: mdl-33432113

ABSTRACT

The ubiquitin system is complex, multifaceted, and is crucial for the modulation of a vast number of cellular processes. Ubiquitination is tightly regulated at different levels by a range of enzymes including E1s, E2s, and E3s, and an array of DUBs. The UPS directs protein degradation through the proteasome, and regulates a wide array of cellular processes including transcription and epigenetic factors as well as key oncoproteins. Ubiquitination is key to the dynamic regulation of programmed cell death. Notably, the TNF signaling pathway is controlled by competing ubiquitin conjugation and deubiquitination, which governs both proteasomal degradation and signaling complex formation. In the inflammatory response, ubiquitination is capable of both activating and dampening inflammasome activation through the control of either protein stability, complex formation, or, in some cases, directly affecting receptor activity. In this review, we discuss the enzymes and targets in the ubiquitin system that regulate fundamental cellular processes regulating cell death, and inflammation, as well as disease consequences resulting from their dysregulation. Finally, we highlight several pre-clinical and clinical compounds that regulate ubiquitin system enzymes, with the aim of restoring homeostasis and ameliorating diseases.


Subject(s)
Inflammation/metabolism , Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Apoptosis , Humans , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Ubiquitin/metabolism
5.
Nature ; 550(7677): 534-538, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29045385

ABSTRACT

The ubiquitin system regulates essential cellular processes in eukaryotes. Ubiquitin is ligated to substrate proteins as monomers or chains and the topology of ubiquitin modifications regulates substrate interactions with specific proteins. Thus ubiquitination directs a variety of substrate fates including proteasomal degradation. Deubiquitinase enzymes cleave ubiquitin from substrates and are implicated in disease; for example, ubiquitin-specific protease-7 (USP7) regulates stability of the p53 tumour suppressor and other proteins critical for tumour cell survival. However, developing selective deubiquitinase inhibitors has been challenging and no co-crystal structures have been solved with small-molecule inhibitors. Here, using nuclear magnetic resonance-based screening and structure-based design, we describe the development of selective USP7 inhibitors GNE-6640 and GNE-6776. These compounds induce tumour cell death and enhance cytotoxicity with chemotherapeutic agents and targeted compounds, including PIM kinase inhibitors. Structural studies reveal that GNE-6640 and GNE-6776 non-covalently target USP7 12 Å distant from the catalytic cysteine. The compounds attenuate ubiquitin binding and thus inhibit USP7 deubiquitinase activity. GNE-6640 and GNE-6776 interact with acidic residues that mediate hydrogen-bond interactions with the ubiquitin Lys48 side chain, suggesting that USP7 preferentially interacts with and cleaves ubiquitin moieties that have free Lys48 side chains. We investigated this idea by engineering di-ubiquitin chains containing differential proximal and distal isotopic labels and measuring USP7 binding by nuclear magnetic resonance. This preferential binding protracted the depolymerization kinetics of Lys48-linked ubiquitin chains relative to Lys63-linked chains. In summary, engineering compounds that inhibit USP7 activity by attenuating ubiquitin binding suggests opportunities for developing other deubiquitinase inhibitors and may be a strategy more broadly applicable to inhibiting proteins that require ubiquitin binding for full functional activity.


Subject(s)
Aminopyridines/chemistry , Aminopyridines/pharmacology , Indazoles/chemistry , Indazoles/pharmacology , Phenols/chemistry , Phenols/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitin/metabolism , Animals , Binding, Competitive , Cell Line, Tumor , Drug Synergism , Female , Humans , Mice , Mice, SCID , Models, Molecular , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/pathology , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Substrate Specificity , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin/chemistry , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/deficiency , Ubiquitin-Specific Peptidase 7/metabolism
6.
Proc Natl Acad Sci U S A ; 112(47): E6436-45, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26554011

ABSTRACT

Actin filament networks assemble on cellular membranes in response to signals that locally activate neural Wiskott-Aldrich-syndrome protein (N-WASP) and the Arp2/3 complex. An inactive conformation of N-WASP is stabilized by intramolecular contacts between the GTPase binding domain (GBD) and the C helix of the verprolin-homology, connector-helix, acidic motif (VCA) segment. Multiple SH3 domain-containing adapter proteins can bind and possibly activate N-WASP, but it remains unclear how such binding events relieve autoinhibition to unmask the VCA segment and activate the Arp2/3 complex. Here, we have used purified components to reconstitute a signaling cascade driven by membrane-localized Src homology 3 (SH3) adapters and N-WASP, resulting in the assembly of dynamic actin networks. Among six SH3 adapters tested, Nck was the most potent activator of N-WASP-driven actin assembly. We identify within Nck a previously unrecognized activation motif in a linker between the first two SH3 domains. This linker sequence, reminiscent of bacterial virulence factors, directly engages the N-WASP GBD and competes with VCA binding. Our results suggest that animals, like pathogenic bacteria, have evolved peptide motifs that allosterically activate N-WASP, leading to localized actin nucleation on cellular membranes.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , src Homology Domains , Actins/metabolism , Allosteric Regulation , Amino Acid Motifs , Amino Acid Sequence , Animals , Humans , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Mutant Proteins/chemistry , Protein Binding , Protein Structure, Secondary , Rats , Structure-Activity Relationship
7.
Nat Commun ; 2: 192, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21304521

ABSTRACT

Rad23 is an adaptor protein that binds to both ubiquitinated substrates and to the proteasome. Despite its association with the proteasome, Rad23 escapes degradation. Here we show that Rad23 remains stable because it lacks an effective initiation region at which the proteasome can engage the protein and unfold it. Rad23 contains several internal, unstructured loops, but these are too short to act as initiation regions. Experiments with model proteins show that internal loops must be surprisingly long to engage the proteasome and support degradation. These length requirements are not specific to Rad23 and reflect a general property of the proteasome.


Subject(s)
DNA-Binding Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Unfolding , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Cytochromes c1/genetics , DNA Primers/genetics , DNA-Binding Proteins/genetics , Linear Models , Molecular Sequence Data , Saccharomyces cerevisiae Proteins/genetics
8.
Nat Chem Biol ; 7(3): 161-7, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21278740

ABSTRACT

The eukaryotic 26S proteasome controls cellular processes by degrading specific regulatory proteins. Most proteins are targeted for degradation by a signal or degron that consists of two parts: a proteasome-binding tag, typically covalently attached polyubiquitin chains, and an unstructured region that serves as the initiation region for proteasomal proteolysis. Here we have characterized how the arrangement of the two degron parts in a protein affects degradation. We found that a substrate is degraded efficiently only when its initiation region is of a certain minimal length and is appropriately separated in space from the proteasome-binding tag. Regions that are located too close or too far from the proteasome-binding tag cannot access the proteasome and induce degradation. These spacing requirements are different for a polyubiquitin chain and a ubiquitin-like domain. Thus, the arrangement and location of the proteasome initiation region affect a protein's fate and are important in selecting proteins for proteasome-mediated degradation.


Subject(s)
Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae/cytology , Binding Sites , Catalysis , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Humans , Immunoglobulins/chemistry , Immunoglobulins/metabolism , Neurospora crassa/metabolism , Polyubiquitin/chemistry , Proteasome Endopeptidase Complex/chemistry , Saccharomyces cerevisiae/metabolism , Substrate Specificity
9.
Indian J Occup Environ Med ; 15(3): 113-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22412289

ABSTRACT

INTRODUCTION: Increasing demands, exacting management, poor ergonomics, and intense competition within and without are likely to contribute to stress among the railway engine pilots. This excess of stress and its consequences cost very high to both the organization and the consumers. AIMS AND OBJECTIVES: This study aims to identify the particular stressors affecting the railway engine pilots and their level of occupational stress in order to improve the safety, efficiency, and overall productivity of the railways and to propose the remedies. MATERIALS AND METHODS: A cross-sectional study conducted at Central Hospital, SECR, Bilaspur, from 20/09/10 to 20/11/10, involving a study sample of 100 healthy Loco Pilots and 100 controls with similar safety category meeting the set inclusion and exclusion criteria, subjected to cross-sectional interviews and questionnaires. RESULTS: Job stress correlated significantly with fatigue (P<0.001), ergonomics of work place (P<0.05) (particularly the postural discomfort and cab space), management pressure (P<0.01), high job demand (P<0.001), low control and low support at work (P<0.01), biological functions (P<0.05), and absenteeism (P<0.001). Top ten stressors have been identified and postural discomfort tops the list. The study also identifies minimal efforts from administration to reduce stress of its employees. CONCLUSION: The high demand, low control, and low support at the work with difficult work environment and inadequate recreation at the place of intermediary rest corroborates with development of stress affecting the normal biological functions leading to either avoidance of duty or making the railway engine pilots susceptible to fatigue and drowsiness, neglect, injuries, and accidents.

10.
J Biol Chem ; 284(28): 18674-84, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19383601

ABSTRACT

ATP-dependent proteases control the concentrations of hundreds of regulatory proteins and remove damaged or misfolded proteins from cells. They select their substrates primarily by recognizing sequence motifs or covalent modifications. Once a substrate is bound to the protease, it has to be unfolded and translocated into the proteolytic chamber to be degraded. Some proteases appear to be promiscuous, degrading substrates with poorly defined targeting signals, which suggests that selectivity may be controlled at additional levels. Here we compare the abilities of representatives from all classes of ATP-dependent proteases to unfold a model substrate protein and find that the unfolding abilities range over more than 2 orders of magnitude. We propose that these differences in unfolding abilities contribute to the fates of substrate proteins and may act as a further layer of selectivity during protein destruction.


Subject(s)
Adenosine Triphosphate/chemistry , Bacillus/enzymology , ATP-Dependent Proteases/chemistry , Bacterial Proteins , Biochemistry/methods , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Kinetics , Protease La/chemistry , Protein Binding , Protein Denaturation , Protein Folding , Protein Structure, Tertiary , Ribonucleases/chemistry , Thermodynamics
11.
Nat Chem Biol ; 5(1): 29-36, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19029916

ABSTRACT

The proteasome controls the turnover of many cellular proteins. Two structural features are typically required for proteins to be degraded: covalently attached ubiquitin polypeptides that allow binding to the proteasome and an unstructured region in the targeted protein that initiates proteolysis. Here, we have tested the degradation of model proteins to further explore how the proteasome selects its substrates. Using purified yeast proteasome and mammalian proteasome in cell lysate, we have demonstrated that the two structural features can act in trans when separated onto different proteins in a multisubunit complex. In such complexes, the location of the unstructured initiation site and its chemical properties determine which subunit is degraded. Thus, our findings reveal the molecular basis of subunit specificity in the degradation of protein complexes. In addition, our data provide a plausible explanation for how adaptor proteins can bind to otherwise stable proteins and target them for degradation.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteins/metabolism , Bacterial Proteins/metabolism , Catalytic Domain , Gene Expression Regulation , Models, Molecular , Protein Conformation , Protein Subunits/metabolism , Ribonucleases/metabolism , Saccharomyces cerevisiae , Substrate Specificity , Ubiquitination
12.
J Am Chem Soc ; 130(12): 4081-8, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18321107

ABSTRACT

Protein-protein pore interaction is a fundamental and ubiquitous process in biology and medical biotechnology. Here, we employed high-resolution time-resolved single-channel electrical recording along with protein engineering to examine a protein-protein pore interaction at single-molecule resolution. The pore was formed by Staphylococcus aureus alpha-hemolysin (alphaHL) protein and contained electrostatic traps formed by rings of seven aspartic acid residues placed at two different positions within the pore lumen. The protein analytes were positively charged presequences (pb2) of varying length fused to the small ribonuclease barnase (Ba). The presence of the electrostatic traps greatly enhanced the interaction of the pb2-Ba protein with the alphaHL protein pore. This study demonstrates the high sensitivity of the nanopore technique to an array of factors that govern the protein-protein pore interaction, including the length of the pb2 presequence, the position of the electrostatic traps within the pore lumen, the ionic strength of the aqueous phase, and the transmembrane potential. Alterations in the functional properties of the pb2-Ba protein and the alphaHL protein pore and systematic changes of the experimental parameters revealed the balance between forces driving the pb2-Ba protein into the pore and forces driving it out.


Subject(s)
Bacterial Toxins/chemistry , Hemolysin Proteins/chemistry , Ribonucleases/chemistry , Aspartic Acid/chemistry , Lipid Bilayers/chemistry , Protein Binding , Protein Engineering , Static Electricity , Time Factors
13.
Trends Cell Biol ; 17(9): 419-21, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17900906

ABSTRACT

The proteasome controls many cellular processes by degrading a large number of regulatory proteins. Most proteins are targeted to the proteasome through covalent tagging by a chain consisting of several copies of the small protein ubiquitin. Finley and coworkers have now discovered two proteins, Hul5 and Ubp6, which regulate degradation further, when bound to the proteasome. Hul5 promotes degradation by extending the number of ubiquitin moieties in the tag on substrates, whereas Ubp6 antagonizes degradation by trimming ubiquitin from the tag. The balance between these two opposing activities might control the substrate specificity of the proteasome and adjusting the balance would provide a new level of degradation control.


Subject(s)
Polyubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Endopeptidases/metabolism , Models, Biological , Saccharomyces cerevisiae Proteins/metabolism , Substrate Specificity , Ubiquitin-Protein Ligases/metabolism
14.
Trends Biochem Sci ; 29(11): 593-600, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15501678

ABSTRACT

Protein unfolding is an important step in several cellular processes such as protein degradation by ATP-dependent proteases and protein translocation across some membranes. Recent studies have shown that the mechanisms of protein unfolding in vivo differ from those of the spontaneous unfolding in vitro measured by solvent denaturation. Proteases and translocases pull at a substrate polypeptide chain and thereby catalyze unraveling by changing the unfolding pathway of that protein. The unfoldases move along the polypeptide chains of their protein substrates. The resistance of a protein to unfolding is then determined by the stability of the region of its structure that is first encountered by the unfoldase. Because unfolding is a necessary step in protein degradation and translocation, the susceptibility of a substrate protein to unfolding contributes to the specificity of these pathways.


Subject(s)
Protein Folding , Proteins/metabolism , ATP-Dependent Proteases/chemistry , ATP-Dependent Proteases/metabolism , Adenosine Triphosphate/metabolism , Animals , Humans , Intracellular Membranes/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/metabolism , Models, Biological , Models, Molecular , Molecular Chaperones/metabolism , Protein Denaturation , Protein Transport , Proteins/chemistry , Substrate Specificity
15.
Nat Struct Mol Biol ; 11(9): 830-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15311270

ABSTRACT

The proteasome is the main ATP-dependent protease in eukaryotic cells and controls the concentration of many regulatory proteins in the cytosol and nucleus. Proteins are targeted to the proteasome by the covalent attachment of polyubiquitin chains. The ubiquitin modification serves as the proteasome recognition element but by itself is not sufficient for efficient degradation of folded proteins. We report that proteolysis of tightly folded proteins is accelerated greatly when an unstructured region is attached to the substrate. The unstructured region serves as the initiation site for degradation and is hydrolyzed first, after which the rest of the protein is digested sequentially. These results identify the initiation site as a novel component of the targeting signal, which is required to engage the proteasome unfolding machinery efficiently. The proteasome degrades a substrate by first binding to its ubiquitin modification and then initiating unfolding at an unstructured region.


Subject(s)
Adenosine Triphosphate/chemistry , Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Animals , Bacillus/metabolism , Binding Sites , Catalysis , Cell Nucleus/metabolism , Cytosol/metabolism , Electrophoresis, Polyacrylamide Gel , Endopeptidases/metabolism , Escherichia coli/metabolism , Hydrolysis , Models, Biological , Promoter Regions, Genetic , Proteasome Endopeptidase Complex , Protein Binding , Protein Folding , Protein Structure, Tertiary , Rabbits , Temperature , Time Factors , Ubiquitin/metabolism
16.
Mol Cell ; 11(3): 659-69, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12667449

ABSTRACT

FtsH, a member of the AAA family of proteins, is the only membrane ATP-dependent protease universally conserved in prokaryotes, and the only essential ATP-dependent protease in Escherichia coli. We investigated the mechanism of degradation by FtsH. Other well-studied ATP-dependent proteases use ATP to unfold their substrates. In contrast, both in vitro and in vivo studies indicate that degradation by FtsH occurs efficiently only when the substrate is a protein of low intrinsic thermodynamic stability. Because FtsH lacks robust unfoldase activity, it is able to use the protein folding state of substrates as a criterion for degradation. This feature may be key to its role in the cell and account for its ubiquitous distribution among prokaryotic organisms.


Subject(s)
Bacterial Proteins/chemistry , Membrane Proteins/chemistry , Sigma Factor , ATP-Dependent Proteases , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Escherichia coli/metabolism , Escherichia coli Proteins , Green Fluorescent Proteins , Heat-Shock Proteins/metabolism , Kinetics , Luminescent Proteins/metabolism , Membrane Proteins/metabolism , Molecular Sequence Data , Plasmids/metabolism , Protein Binding , Protein Folding , Protein Structure, Tertiary , RNA, Bacterial/metabolism , Recombinant Fusion Proteins/metabolism , Ribonucleases/metabolism , Substrate Specificity , Temperature , Thermodynamics , Time Factors , Transcription Factors/metabolism
17.
J Biol Chem ; 277(38): 34760-5, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12080075

ABSTRACT

The proteasome can actively unfold proteins by sequentially unraveling their substrates from the attachment point of the degradation signal. To investigate the steric constraints imposed on substrate proteins during their degradation by the proteasome, we constructed a model protein in which specific parts of the polypeptide chain were covalently connected through disulfide bridges. The cross-linked model proteins were fully degraded by the proteasome, but two or more cross-links retarded the degradation slightly. These results suggest that the pore of the proteasome allows the concurrent passage of at least three stretches of a polypeptide chain. A degradation channel that can tolerate some steric bulk may reconcile the two opposing needs for degradation that is compartmentalized to avoid aberrant proteolysis yet able to handle a range of substrates of various sizes.


Subject(s)
Cysteine Endopeptidases/metabolism , Multienzyme Complexes/metabolism , Electrophoresis, Polyacrylamide Gel , Hydrolysis , Proteasome Endopeptidase Complex , Protein Transport , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...