Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 401
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 174(6): 1507-1521.e16, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30100183

ABSTRACT

The hetero-oligomeric chaperonin of eukarya, TRiC, is required to fold the cytoskeletal protein actin. The simpler bacterial chaperonin system, GroEL/GroES, is unable to mediate actin folding. Here, we use spectroscopic and structural techniques to determine how TRiC promotes the conformational progression of actin to the native state. We find that actin fails to fold spontaneously even in the absence of aggregation but populates a kinetically trapped, conformationally dynamic state. Binding of this frustrated intermediate to TRiC specifies an extended topology of actin with native-like secondary structure. In contrast, GroEL stabilizes bound actin in an unfolded state. ATP binding to TRiC effects an asymmetric conformational change in the chaperonin ring. This step induces the partial release of actin, priming it for folding upon complete release into the chaperonin cavity, mediated by ATP hydrolysis. Our results reveal how the unique features of TRiC direct the folding pathway of an obligate eukaryotic substrate.


Subject(s)
Actins/metabolism , Chaperonin 10/metabolism , Chaperonin 60/metabolism , Actins/chemistry , Adenosine Triphosphate/metabolism , Animals , Cattle , Chaperonin 10/chemistry , Chaperonin 60/chemistry , Cryoelectron Microscopy , Deoxyribonuclease I/chemistry , Deoxyribonuclease I/metabolism , Deuterium Exchange Measurement , Humans , Protein Binding , Protein Folding , Protein Structure, Tertiary
2.
Proc Natl Acad Sci U S A ; 120(50): e2308933120, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38064510

ABSTRACT

The bacterial chaperonin GroEL-GroES promotes protein folding through ATP-regulated cycles of substrate protein binding, encapsulation, and release. Here, we have used cryoEM to determine structures of GroEL, GroEL-ADP·BeF3, and GroEL-ADP·AlF3-GroES all complexed with the model substrate Rubisco. Our structures provide a series of snapshots that show how the conformation and interactions of non-native Rubisco change as it proceeds through the GroEL-GroES reaction cycle. We observe specific charged and hydrophobic GroEL residues forming strong initial contacts with non-native Rubisco. Binding of ATP or ADP·BeF3 to GroEL-Rubisco results in the formation of an intermediate GroEL complex displaying striking asymmetry in the ATP/ADP·BeF3-bound ring. In this ring, four GroEL subunits bind Rubisco and the other three are in the GroES-accepting conformation, suggesting how GroEL can recruit GroES without releasing bound substrate. Our cryoEM structures of stalled GroEL-ADP·AlF3-Rubisco-GroES complexes show Rubisco folding intermediates interacting with GroEL-GroES via different sets of residues.


Subject(s)
Adenosine Triphosphate , Ribulose-Bisphosphate Carboxylase , Ribulose-Bisphosphate Carboxylase/metabolism , Adenosine Triphosphate/metabolism , Chaperonin 60/metabolism , Chaperonin 10/chemistry , Protein Folding , Protein Binding
3.
Subcell Biochem ; 101: 213-246, 2023.
Article in English | MEDLINE | ID: mdl-36520309

ABSTRACT

Co-chaperonins function together with chaperonins to mediate ATP-dependent protein folding in a variety of cellular compartments. Chaperonins are evolutionarily conserved and form two distinct classes, namely, group I and group II chaperonins. GroEL and its co-chaperonin GroES form part of group I and are the archetypal members of this family of protein folding machines. The unique mechanism used by GroEL and GroES to drive protein folding is embedded in the complex architecture of double-ringed complexes, forming two central chambers that undergo conformational rearrangements that enable protein folding to occur. GroES forms a lid over the chamber and in doing so dislodges bound substrate into the chamber, thereby allowing non-native proteins to fold in isolation. GroES also modulates allosteric transitions of GroEL. Group II chaperonins are functionally similar to group I chaperonins but differ in structure and do not require a co-chaperonin. A significant number of bacteria and eukaryotes house multiple chaperonin and co-chaperonin proteins, many of which have acquired additional intracellular and extracellular biological functions. In some instances, co-chaperonins display contrasting functions to those of chaperonins. Human HSP60 (HSPD) continues to play a key role in the pathogenesis of many human diseases, in particular autoimmune diseases and cancer. A greater understanding of the fascinating roles of both intracellular and extracellular Hsp10 on cellular processes will accelerate the development of techniques to treat diseases associated with the chaperonin family.


Subject(s)
Chaperonin 10 , Chaperonins , Humans , Chaperonin 10/chemistry , Chaperonins/chemistry , Chaperonins/metabolism , Chaperonin 60/chemistry , Protein Folding , Group II Chaperonins/metabolism , Adenosine Triphosphate/metabolism
4.
FASEB J ; 36(3): e22198, 2022 03.
Article in English | MEDLINE | ID: mdl-35199390

ABSTRACT

GroES/GroEL is the only bacterial chaperone essential under all conditions, making it a potential antibiotic target. Rationally targeting ESKAPE GroES/GroEL as an antibiotic strategy necessitates studying their structure and function. Herein, we outline the structural similarities between Escherichia coli and ESKAPE GroES/GroEL and identify significant differences in intra- and inter-ring cooperativity, required in the refolding cycle of client polypeptides. Previously, we observed that one-half of ESKAPE GroES/GroEL family members could not support cell viability when each was individually expressed in GroES/GroEL-deficient E. coli cells. Cell viability was found to be dependent on the allosteric compatibility between ESKAPE and E. coli subunits within mixed (E. coli and ESKAPE) tetradecameric GroEL complexes. Interestingly, differences in allostery did not necessarily result in differences in refolding rate for a given homotetradecameric chaperonin. Characterization of ESKAPE GroEL allostery, ATPase, and refolding rates in this study will serve to inform future studies focused on inhibitor design and mechanism of action studies.


Subject(s)
Allosteric Site , Escherichia coli Proteins/chemistry , Heat-Shock Proteins/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Allosteric Regulation , Chaperonin 10/chemistry , Chaperonin 10/genetics , Chaperonin 10/metabolism , Escherichia coli , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism
5.
Int J Mol Sci ; 24(9)2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37175554

ABSTRACT

Similar to its bacterial homolog GroEL, Hsp60 in oligomeric conformation is known to work as a folding machine, with the assistance of co-chaperonin Hsp10 and ATP. However, recent results have evidenced that Hsp60 can stabilize aggregation-prone molecules in the absence of Hsp10 and ATP by a different, "holding-like" mechanism. Here, we investigated the relationship between the oligomeric conformation of Hsp60 and its ability to inhibit fibrillization of the Ab40 peptide. The monomeric or tetradecameric form of the protein was isolated, and its effect on beta-amyloid aggregation was separately tested. The structural stability of the two forms of Hsp60 was also investigated using differential scanning calorimetry (DSC), light scattering, and circular dichroism. The results showed that the protein in monomeric form is less stable, but more effective against amyloid fibrillization. This greater functionality is attributed to the disordered nature of the domains involved in subunit contacts.


Subject(s)
Adenosine Triphosphate , Chaperonin 60 , Chaperonin 60/metabolism , Adenosine Triphosphate/metabolism , Chaperonin 10/chemistry , Protein Folding
6.
J Biol Chem ; 296: 100744, 2021.
Article in English | MEDLINE | ID: mdl-33957121

ABSTRACT

This review contains a personal account of the role played by the PDB in the development of the field of molecular chaperones and protein homeostasis, from the viewpoint of someone who experienced the concurrent advances in the structural biology, electron microscopy, and chaperone fields. The emphasis is on some key structures, including those of Hsp70, GroEL, Hsp90, and small heat shock proteins, that were determined as the molecular chaperone concept and systems for protein quality control were emerging. These structures were pivotal in demonstrating how seemingly nonspecific chaperones could assist the specific folding pathways of a variety of substrates. Moreover, they have provided mechanistic insights into the ATPase machinery of complexes such as GroEL/GroES that promote unfolding and folding and the disaggregases that extract polypeptides from large aggregates and disassemble amyloid fibers. The PDB has provided a framework for the current success in curating, evaluating, and distributing structural biology data, through both the PDB and the EMDB.


Subject(s)
Chaperonin 10 , Chaperonin 60 , Databases, Protein , HSP70 Heat-Shock Proteins , HSP90 Heat-Shock Proteins , Proteolysis , Animals , Chaperonin 10/chemistry , Chaperonin 10/genetics , Chaperonin 10/metabolism , Chaperonin 60/chemistry , Chaperonin 60/genetics , Chaperonin 60/metabolism , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans
7.
J Am Chem Soc ; 144(6): 2667-2678, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35107280

ABSTRACT

Chaperonins are nanomachines that harness ATP hydrolysis to power and catalyze protein folding, a chemical action that is directly linked to the maintenance of cell function through protein folding/refolding and assembly. GroEL and the GroEL-GroES complex are archetypal examples of such protein folding machines. Here, variable-temperature electrospray ionization (vT-ESI) native mass spectrometry is used to delineate the effects of solution temperature and ATP concentrations on the stabilities of GroEL and GroEL-GroES complexes. The results show clear evidence for destabilization of both GroEL14 and GroES7 at temperatures of 50 and 45 °C, respectively, substantially below the previously reported melting temperature (Tm ∼ 70 °C). This destabilization is accompanied by temperature-dependent reaction products that have previously unreported stoichiometries, viz. GroEL14-GroESy-ATPn, where y = 1, 2, 8 and n = 0, 1, 2, 8, that are also dependent on Mg2+ and ATP concentrations. Variable-temperature native mass spectrometry reveals new insights about the stability of GroEL in response to temperature effects: (i) temperature-dependent ATP binding to GroEL; (ii) effects of temperature as well as Mg2+ and ATP concentrations on the stoichiometry of the GroEL-GroES complex, with Mg2+ showing greater effects compared to ATP; and (iii) a change in the temperature-dependent stoichiometries of the GroEL-GroES complex (GroEL14-GroES7 vs GroEL14-GroES8) between 24 and 40 °C. The similarities between results obtained by using native MS and cryo-EM [Clare et al. An expanded protein folding cage in the GroEL-gp31 complex. J. Mol. Biol. 2006, 358, 905-911; Ranson et al. Allosteric signaling of ATP hydrolysis in GroEL-GroES complexes.Nat. Struct. Mol. Biol. 2006, 13, 147-152] underscore the utility of native MS for investigations of molecular machines as well as identification of key intermediates involved in the chaperonin-assisted protein folding cycle.


Subject(s)
Adenosine Triphosphate/metabolism , Chaperonin 10/metabolism , Chaperonin 60/metabolism , Magnesium/metabolism , Chaperonin 10/chemistry , Chaperonin 60/chemistry , Escherichia coli/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Ligands , Mass Spectrometry , Protein Binding , Protein Conformation , Protein Stability , Protein Unfolding , Temperature
8.
Biochemistry ; 60(6): 460-464, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33464880

ABSTRACT

The Escherichia coli ATP-consuming chaperonin machinery, a complex between GroEL and GroES, has evolved to facilitate folding of substrate proteins (SPs) that cannot do so spontaneously. A series of kinetic experiments show that the SPs are encapsulated in the GroEL/ES nanocage for a short duration. If confinement of the SPs is the mechanism by which GroEL/ES facilitates folding, it follows that the assisted folding rate, relative to the bulk value, should always be enhanced. Here, we show that this is not the case for the folding of rhodanese in the presence of the full machinery of GroEL/ES and ATP. The assisted folding rate of rhodanese decreases. On the basis of our finding and those reported in other studies, we suggest that the ATP-consuming chaperonin machinery has evolved to optimize the product of the folding rate and the yield of the folded SPs on the biological time scale. Neither the rate nor the yield is separately maximized.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 60/chemistry , Protein Folding , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Chaperonin 10/metabolism , Chaperonin 60/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Kinetics , Protein Conformation
9.
J Struct Biol ; 213(2): 107727, 2021 06.
Article in English | MEDLINE | ID: mdl-33753204

ABSTRACT

Cryo-electron tomography provides the opportunity for unsupervised discovery of endogenous complexes in situ. This process usually requires particle picking, clustering and alignment of subtomograms to produce an average structure of the complex. When applied to heterogeneous samples, template-free clustering and alignment of subtomograms can potentially lead to the discovery of structures for unknown endogenous complexes. However, such methods require scoring functions to measure and accurately rank the quality of aligned subtomogram clusters, which can be compromised by contaminations from misclassified complexes and alignment errors. Here, we provide the first study to assess the effectiveness of more than 15 scoring functions for evaluating the quality of subtomogram clusters, which differ in the amount of structural misalignments and contaminations due to misclassified complexes. We assessed both experimental and simulated subtomograms as ground truth data sets. Our analysis showed that the robustness of scoring functions varies largely. Most scores were sensitive to the signal-to-noise ratio of subtomograms and often required Gaussian filtering as preprocessing for improved performance. Two scoring functions, Spectral SNR-based Fourier Shell Correlation and Pearson Correlation in the Fourier domain with missing wedge correction, showed a robust ranking of subtomogram clusters without any preprocessing and irrespective of SNR levels of subtomograms. Of these two scoring functions, Spectral SNR-based Fourier Shell Correlation was fastest to compute and is a better choice for handling large numbers of subtomograms. Our results provide a guidance for choosing an accurate scoring function for template-free approaches to detect complexes from heterogeneous samples.


Subject(s)
Cryoelectron Microscopy/methods , Electron Microscope Tomography/methods , Imaging, Three-Dimensional/methods , Chaperonin 10/chemistry , Chaperonin 60/chemistry , Databases, Protein , Normal Distribution , Ribosomes/chemistry , Signal-To-Noise Ratio
10.
Trends Biochem Sci ; 41(1): 62-76, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26422689

ABSTRACT

The bacterial chaperonin GroEL and its cofactor GroES constitute the paradigmatic molecular machine of protein folding. GroEL is a large double-ring cylinder with ATPase activity that binds non-native substrate protein (SP) via hydrophobic residues exposed towards the ring center. Binding of the lid-shaped GroES to GroEL displaces the bound protein into an enlarged chamber, allowing folding to occur unimpaired by aggregation. GroES and SP undergo cycles of binding and release, regulated allosterically by the GroEL ATPase. Recent structural and functional studies are providing insights into how the physical environment of the chaperonin cage actively promotes protein folding, in addition to preventing aggregation. Here, we review different models of chaperonin action and discuss issues of current debate.


Subject(s)
Chaperonin 10/metabolism , Chaperonin 60/metabolism , Mitochondrial Proteins/metabolism , Nanostructures , Protein Folding , Chaperonin 10/chemistry , Chaperonin 60/chemistry , Humans , Mitochondrial Proteins/chemistry , Models, Molecular
11.
Phys Chem Chem Phys ; 22(6): 3734-3743, 2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32010904

ABSTRACT

The chaperonin system GroEL-GroES is present in all kingdoms of life and rescues proteins from improper folding and aggregation upon internal and external stress conditions, including high temperatures and pressures. Here, we set out to explore the thermo- and piezostability of GroEL, GroES and the GroEL-GroES complex in the presence of cosolvents, nucleotides and salts employing quantitative FTIR spectroscopy and small-angle X-ray scattering. Owing to its high biological relevance and lack of data, our focus was especially on the effect of pressure on the chaperonin system. The experimental results reveal that the GroEL-GroES complex is remarkably temperature stable with an unfolding temperature beyond 70 °C, which can still be slightly increased by compatible cosolutes like TMAO. Conversely, the pressure stability of GroEL and hence the GroEL-GroES complex is rather limited and much less than that of monomeric proteins. Whereas GroES is pressure stable up to ∼5 kbar, GroEl and the GroEl-GroES complex undergo minor structural changes already beyond 1 kbar, which can be attributed to a dissociation-induced conformational drift. Quite unexpectedly, no significant unfolding of GroEL is observed even up to 10 kbar, however, i.e., the subunits themselves are very pressure stable. As for the physiological relevance, the structural integrity of the chaperonin system is retained in a relatively narrow pressure range, from about 1 to 1000 bar, which is just the pressure range encountered by life on Earth.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 60/chemistry , Environment , Pressure , Protein Stability , Temperature
12.
Proc Natl Acad Sci U S A ; 114(34): 9104-9109, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28784759

ABSTRACT

We have studied the interaction of the prototypical chaperonin GroEL with the prion domain of the Het-s protein using solution and solid-state NMR, electron and atomic force microscopies, and EPR. While GroEL accelerates Het-s protofibril formation by several orders of magnitude, the rate of appearance of fibrils is reduced. GroEL remains bound to Het-s throughout the aggregation process and densely decorates the fibrils at a regular spacing of ∼200 Å. GroEL binds to the Het-s fibrils via its apical domain located at the top of the large open ring. Thus, apo GroEL and bullet-shaped GroEL/GroES complexes in which only a single ring is capped by GroES interact with the Het-s fibrils; no evidence is seen for any interaction with football-shaped GroEL/GroES complexes in which both rings are capped by GroES. EPR spectroscopy shows that rotational motion of a nitroxide spin label, placed at the N-terminal end of the first ß-strand of Het-s fibrils, is significantly reduced in both Het-s/GroEL aggregates and Het-s fibrils, but virtually completely eliminated in Het-s/GroEL fibrils, suggesting that in the latter, GroEL may come into close proximity to the nitroxide label. Solid-state NMR measurements indicate that GroEL binds to the mobile regions of the Het-s fibril comprising the N-terminal tail and a loop connecting ß-strands 4 and 5, consistent with interactions involving GroEL binding consensus sequences located therein.


Subject(s)
Amyloid/chemistry , Chaperonin 60/chemistry , Fungal Proteins/chemistry , Prion Proteins/chemistry , Amino Acid Sequence , Amyloid/metabolism , Amyloid/ultrastructure , Chaperonin 10/chemistry , Chaperonin 10/genetics , Chaperonin 10/metabolism , Chaperonin 60/genetics , Chaperonin 60/metabolism , Electron Spin Resonance Spectroscopy , Fungal Proteins/genetics , Fungal Proteins/metabolism , Magnetic Resonance Spectroscopy , Microscopy, Atomic Force , Microscopy, Electron , Models, Molecular , Mutation , Prion Proteins/genetics , Prion Proteins/metabolism , Protein Binding , Protein Conformation
13.
J Biol Chem ; 292(50): 20583-20591, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29066625

ABSTRACT

The GroE chaperonin system in Escherichia coli comprises GroEL and GroES and facilitates ATP-dependent protein folding in vivo and in vitro Proteins with very similar sequences and structures can differ in their dependence on GroEL for efficient folding. One potential but unverified source for GroEL dependence is frustration, wherein not all interactions in the native state are optimized energetically, thereby potentiating slow folding and misfolding. Here, we chose enhanced green fluorescent protein as a model system and subjected it to random mutagenesis, followed by screening for variants whose in vivo folding displays increased or decreased GroEL dependence. We confirmed the altered GroEL dependence of these variants with in vitro folding assays. Strikingly, mutations at positions predicted to be highly frustrated were found to correlate with decreased GroEL dependence. Conversely, mutations at positions with low frustration were found to correlate with increased GroEL dependence. Further support for this finding was obtained by showing that folding of an enhanced green fluorescent protein variant designed computationally to have reduced frustration is indeed less GroEL-dependent. Our results indicate that changes in local frustration also affect partitioning in vivo between spontaneous and chaperonin-mediated folding. Hence, the design of minimally frustrated sequences can reduce chaperonin dependence and improve protein expression levels.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 60/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/metabolism , Green Fluorescent Proteins/chemistry , Heat-Shock Proteins/chemistry , Models, Molecular , Amino Acid Substitution , Chaperonin 10/genetics , Chaperonin 10/metabolism , Chaperonin 60/genetics , Chaperonin 60/metabolism , Computational Biology , Crystallography, X-Ray , Databases, Protein , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Kinetics , Mutation , Protein Conformation , Protein Engineering , Protein Folding , Protein Refolding , Protein Stability , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Solubility , Structural Homology, Protein
14.
Biochim Biophys Acta Gen Subj ; 1862(7): 1576-1583, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29627450

ABSTRACT

GroEL along with ATP and its co-chaperonin GroES has been demonstrated to significantly enhance the folding of newly translated G-protein-coupled receptors (GPCRs). This work extends the previous studies to explore the guest capture and release processes in GroEL-assisted folding of GPCRs, by the reduced approach of employing CXCR4 transmembrane peptides as model substrates. Each of the CXCR4-derived peptides exhibited high affinity for GroEL with a binding stoichiometry near seven. It is found that the peptides interact with the paired α helices in the apical domain of the chaperonin which are similar with the binding sites of SBP (strongly binding peptide: SWMTTPWGFLHP). Complementary binding study with a single-ring version of GroEL indicates that each of the two chaperonin rings is competent for accommodating all the seven CXCR4 peptides bound to GroEL under saturation condition. Meanwhile, the binding kinetics of CXCR4 peptides with GroEL was also examined; ATP alone, or in combination of GroES evidently promoted the release of the peptide substrates from the chaperonin. The results obtained would be beneficial to understand the thermodynamic and kinetic nature of GroEL-GPCRs interaction which is the central molecular event in the assisted folding process.


Subject(s)
Chaperonin 60/metabolism , Receptors, CXCR4/metabolism , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Binding Sites , Binding, Competitive , Chaperonin 10/chemistry , Chaperonin 10/metabolism , Chaperonin 60/chemistry , Chaperonin 60/genetics , Humans , Kinetics , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Domains , Protein Folding , Protein Interaction Mapping , Protein Structure, Secondary , Receptors, CXCR4/chemistry , Thermodynamics
15.
Subcell Biochem ; 83: 483-504, 2017.
Article in English | MEDLINE | ID: mdl-28271487

ABSTRACT

Chaperonin is categorized as a molecular chaperone and mediates the formation of the native conformation of proteins by first preventing folding during synthesis or membrane translocation and subsequently by mediating the step-wise ATP-dependent release that result in proper folding. In the GroEL-GroES complex, a single heptameric GroEL ring binds one GroES ring in the presence of ATP/ADP, in this vein, the double ring GroEL tetradecamer is present in two distinct types of GroEL-GroES complexes: asymmetric 1:1 "bullet"-shaped GroEL:GroES and symmetric 1:2 "football" (American football)-shaped GroEL:GroES2. There have been debates as to which complex is critical to the productive protein folding mediated by the GroEL-GroES complex, and how GroES coordinates with GroEL in the chaperonin reaction cycle in association with regulation by adenine nucleotides and through the interplay of substrate proteins. A lot of knowledge on chaperonins has been accumulating as if expanding as ripples spread around the GroEL-GroES from Escherichia coli. In this article, an overview is presented on GroEL and the GroEL-GroES complex, with emphasis on their morphological variations, and some potential applications to the fabrication of nanocomposites using GroEL as a nano-block. In parallel, a guideline is presented that supports the recognition that the E. coli and its GroEL-GroES complex do not always receive in standard literature because the biochemical features of chaperonins derived from others special, such as mammals, are not always the same as those confirmed using GroEL-GroES derived from E. coli.


Subject(s)
Chaperonin 10/metabolism , Chaperonin 60/metabolism , Escherichia coli/metabolism , Adenosine Triphosphate/metabolism , Animals , Chaperonin 10/chemistry , Chaperonin 60/chemistry , Escherichia coli/chemistry , Escherichia coli/enzymology , Protein Binding , Protein Folding
16.
Proc Natl Acad Sci U S A ; 112(19): 6044-9, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25918392

ABSTRACT

Human mitochondria harbor a single type I chaperonin system that is generally thought to function via a unique single-ring intermediate. To date, no crystal structure has been published for any mammalian type I chaperonin complex. In this study, we describe the crystal structure of a football-shaped, double-ring human mitochondrial chaperonin complex at 3.15 Å, which is a novel intermediate, likely representing the complex in an early stage of dissociation. Interestingly, the mitochondrial chaperonin was captured in a state that exhibits subunit asymmetry within the rings and nucleotide symmetry between the rings. Moreover, the chaperonin tetradecamers show a different interring subunit arrangement when compared to GroEL. Our findings suggest that the mitochondrial chaperonins use a mechanism that is distinct from the mechanism of the well-studied Escherichia coli system.


Subject(s)
Chaperonins/chemistry , Mitochondria/chemistry , Mitochondrial Proteins/chemistry , Adenosine Triphosphate/chemistry , Animals , Chaperonin 10/chemistry , Chaperonin 60/chemistry , Crystallography, X-Ray , Escherichia coli/metabolism , Humans , Hydrolysis , Mice , Models, Molecular , Nucleotides/chemistry , Protein Binding , Protein Folding , Protein Structure, Tertiary
17.
Int J Mol Sci ; 19(2)2018 Feb 06.
Article in English | MEDLINE | ID: mdl-29415503

ABSTRACT

The E. coli GroEL/GroES chaperonin complex acts as a folding cage by producing a bullet-like asymmetric complex, and GroEL exists as double rings regardless of the presence of adenosine triphosphate (ATP). Its mammalian chaperonin homolog, heat shock protein, HSP60, and co-chaperonin, HSP10, play an essential role in protein folding by capturing unfolded proteins in the HSP60/HSP10 complex. However, the structural transition in ATPase-dependent reaction cycle has remained unclear. We found nucleotide-dependent association and dissociation of the HSP60/HSP10 complex using various analytical techniques under near physiological conditions. Our results showed that HSP60 exist as a significant number of double-ring complexes (football- and bullet-type complexes) and a small number of single-ring complexes in the presence of ATP and HSP10. HSP10 binds to HSP60 in the presence of ATP, which increased the HSP60 double-ring formation. After ATP is hydrolyzed to Adenosine diphosphate (ADP), HSP60 released the HSP10 and the dissociation of the double-ring to single-rings occurred. These results indicated that HSP60/HSP10 undergoes an ATP-dependent transition between the single- and double-rings in their system that is highly distinctive from the GroEL/GroES system particularly in the manner of complex formation and the roles of ATP binding and hydrolysis in the reaction cycle.


Subject(s)
Chaperonin 60/chemistry , Chaperonin 60/metabolism , Chemical Phenomena , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Chaperonin 10/chemistry , Chaperonin 10/metabolism , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Structure , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Protein Binding
18.
Mol Biol (Mosk) ; 52(5): 879-897, 2018.
Article in Russian | MEDLINE | ID: mdl-30363062

ABSTRACT

The Hsp60/Hsp10 chaperonin system is one of the most studied systems of cell emergency responses to stresses associated with changes in environmental conditions. In this regard, we have performed a bioinformatics analysis of 164 amino acid sequences of Hsp60 and 125 amino acid sequences of Hsp10 in five classes of chordata. This enabled uncovering the relationship between the identity of the amino acid composition of Hsp60/Hsp10 and the evolutionary distance between classes of chordata. In the course of the study of the chaperonin crystal structure, potentially significant amino acid motifs responsible for the oligomerization of Hsp60 and Hsp10 monomers and the association/dissociation of the Hsp60 and Hsp10 heterodimer have been identified. In addition, we have established that Hsp60 and Hsp10 can form amyloid fibrils due to structural features through the alternative using of the oligomerization sites of monomers as well as association/dissociation sites.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 60/chemistry , Chordata , Amino Acid Motifs , Animals , Computational Biology
19.
Mol Biol (Mosk) ; 52(1): 82-87, 2018.
Article in Russian | MEDLINE | ID: mdl-29512639

ABSTRACT

GroES is a heptameric partner of tetradecameric molecular chaperone GroEL, which ensures the correct folding and assembly of numerous cellular proteins both in vitro and in vivo. This work demonstrates the results of a study of structural aspects of GroES that affect its interaction with GroEL and reassembly. The effect of limited trypsinolysis of GroES on these processes has been studied. It has been shown that limited trypsinolysis of GroES is only strongly pronounced outside the complex with GroEL and results in the cleavage of the peptide bond between Lys20 and Ser21. The N-terminal fragment (~2 kDa) is retained in the GroES particle, which maintains its heptaoligomeric structure but loses the ability to interact with GroEL and dissociates upon a change in the pH from 7 to 8. Trypsin-nicked GroES cannot reassemble after urea-induced unfolding, while the urea-induced unfolding of intact GroES is fully reversible. The reported results indicate the important role of the N-terminal part of GroES subunit in the assembly of its heptameric structure and the interaction with GroEL.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 60/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Trypsin/chemistry , Protein Folding
20.
Mol Biol (Mosk) ; 52(1): 120-124, 2018.
Article in Russian | MEDLINE | ID: mdl-29512644

ABSTRACT

The products of the reassembly reaction of tetradecameric two-ring quaternary structure of GroEL chaperonin under the pressure of its heptameric co-chaperonin GroES have been visualized by electron microscopy. It has been shown that one-ring heptameric oligomers of GroEL have been formed at the beginning (after ~5 min) of the reaction, while at the final stage of the reaction (after ~70 min), both one-ring heptamers in complex with one GroES and two-rings tetradecamers in complexes with one (asymmetrical complex) or two (symmetrical complex) GroES heptamers are present. The relationship between the data of light scattering, native electrophoresis, and electron microscopy obtained earlier has been discussed.


Subject(s)
Chaperonin 10/chemistry , Chaperonin 60/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Chaperonin 10/ultrastructure , Chaperonin 60/ultrastructure , Escherichia coli Proteins/ultrastructure , Microscopy, Electron , Protein Binding , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL