Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(48): e2123238119, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36409905

ABSTRACT

The 70 kDa heat shock proteins (Hsp70s) are highly versatile molecular chaperones that assist in a wide variety of protein-folding processes. They exert their functions by continuously cycling between states of low and high affinity for client polypeptides, driven by ATP-binding and hydrolysis. This cycling is tuned by cochaperones and clients. Although structures for the high and low client affinity conformations of Hsp70 and Hsp70 domains in complex with various cochaperones and peptide clients are available, it is unclear how structural rearrangements in the presence of cochaperones and clients are orchestrated in space and time. Here, we report insights into the conformational dynamics of the prokaryotic model Hsp70 DnaK throughout its adenosine-5'-triphosphate hydrolysis (ATPase) cycle using proximity-induced fluorescence quenching. Our data suggest that ATP and cochaperone-induced structural rearrangements in DnaK occur in a sequential manner and resolve hitherto unpredicted cochaperone and client-induced structural rearrangements. Peptides induce large conformational changes in DnaK·ATP prior to ATP hydrolysis, whereas a protein client induces significantly smaller changes but is much more effective in stimulating ATP hydrolysis. Analysis of the enthalpies of activation for the ATP-induced opening of the DnaK lid in the presence of clients indicates that the lid does not exert an enthalpic pulling force onto bound clients, suggesting entropic pulling as a major mechanism for client unfolding. Our data reveal important insights into the mechanics, allostery, and dynamics of Hsp70 chaperones. We established a methodology for understanding the link between dynamics and function, Hsp70 diversity, and activity modulation.


Subject(s)
Adenosine Triphosphatases , Escherichia coli Proteins , Humans , Adenosine Triphosphatases/metabolism , Escherichia coli Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Adenosine Triphosphate/metabolism
2.
Mol Cell ; 74(4): 831-843.e4, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31027880

ABSTRACT

The activity of the tumor suppressor p53 has to be timed and balanced closely to prevent untimely induction of cell death. The stability of p53 depends on the ubiquitin ligase Mdm2 but also on Hsp70 and Hsp90 chaperones that interact with its DNA binding domain (DBD). Using hydrogen exchange mass spectrometry and biochemical methods, we analyzed conformational states of wild-type p53-DBD at physiological temperatures and conformational perturbations in three frequent p53 cancer mutants. We demonstrate that the Hsp70/Hdj1 system shifts the conformational equilibrium of p53 toward a flexible, more mutant-like, DNA binding inactive state by binding to the DNA binding loop. The analyzed cancer mutants are likewise destabilized by interaction with the Hsp70/Hdj1 system. In contrast, Hsp90 protects the DBD of p53 wild-type and mutant proteins from unfolding. We propose that the Hsp70 and Hsp90 chaperone systems assume complementary functions to optimally balance conformational plasticity with conformational stability.


Subject(s)
HSP40 Heat-Shock Proteins/chemistry , Neoplasms/genetics , Protein Conformation , Tumor Suppressor Protein p53/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HSP40 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/genetics , Humans , Mass Spectrometry , Molecular Chaperones , Neoplasms/pathology , Protein Domains/genetics , Protein Unfolding , Tumor Suppressor Protein p53/genetics
3.
Mol Cell ; 70(3): 545-552.e9, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29706537

ABSTRACT

Protein folding in the cell requires ATP-driven chaperone machines such as the conserved Hsp70 and Hsp90. It is enigmatic how these machines fold proteins. Here, we show that Hsp90 takes a key role in protein folding by breaking an Hsp70-inflicted folding block, empowering protein clients to fold on their own. At physiological concentrations, Hsp70 stalls productive folding by binding hydrophobic, core-forming segments. Hsp90 breaks this deadlock and restarts folding. Remarkably, neither Hsp70 nor Hsp90 alters the folding rate despite ensuring high folding yields. In fact, ATP-dependent chaperoning is restricted to the early folding phase. Thus, the Hsp70-Hsp90 cascade does not fold proteins, but instead prepares them for spontaneous, productive folding. This stop-start mechanism is conserved from bacteria to man, assigning also a general function to bacterial Hsp90, HtpG. We speculate that the decreasing hydrophobicity along the Hsp70-Hsp90 cascade may be crucial for enabling spontaneous folding.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Adenosine Triphosphate/metabolism , Animals , Escherichia coli/metabolism , Fireflies/metabolism , Humans , Protein Folding , Saccharomyces cerevisiae/metabolism
4.
Nat Struct Mol Biol ; 25(1): 83-89, 2018 01.
Article in English | MEDLINE | ID: mdl-29323280

ABSTRACT

Protein quality control depends on the tight regulation of interactions between molecular chaperones and polypeptide substrates. Substrate release from the chaperone Hsp70 is triggered by nucleotide-exchange factors (NEFs) that control folding and degradation fates via poorly understood mechanisms. We found that the armadillo-type NEFs budding yeast Fes1 and its human homolog HspBP1 employ flexible N-terminal release domains (RDs) with substrate-mimicking properties to ensure the efficient release of persistent substrates from Hsp70. The RD contacts the substrate-binding domain of the chaperone, competes with peptide substrate for binding and is essential for proper function in yeast and mammalian cells. Thus, the armadillo domain engages Hsp70 to trigger nucleotide exchange, whereas the RD safeguards the release of substrates. Our findings provide fundamental mechanistic insight into the functional specialization of Hsp70 NEFs and have implications for the understanding of proteostasis-related disorders, including Marinesco-Sjögren syndrome.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , HSP70 Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Adenosine Triphosphate/chemistry , DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Molecular Chaperones/metabolism , Nucleotides/metabolism , Phenotype , Protein Denaturation , Protein Folding , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism
5.
Mol Cell ; 69(2): 227-237.e4, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29290615

ABSTRACT

Efficient targeting of Hsp70 chaperones to substrate proteins depends on J-domain cochaperones, which in synergism with substrates trigger ATP hydrolysis in Hsp70s and concomitant substrate trapping. We present the crystal structure of the J-domain of Escherichia coli DnaJ in complex with the E. coli Hsp70 DnaK. The J-domain interacts not only with DnaK's nucleotide-binding domain (NBD) but also with its substrate-binding domain (SBD) and packs against the highly conserved interdomain linker. Mutational replacement of contacts between J-domain and SBD strongly reduces the ability of substrates to stimulate ATP hydrolysis in the presence of DnaJ and compromises viability at heat shock temperatures. Our data demonstrate that the J-domain and the substrate do not deliver completely independent signals for ATP hydrolysis, but the J-domain, in addition to its direct influence on Hsp70s catalytic center, makes Hsp70 more responsive for the hydrolysis-inducing signal of the substrate, resulting in efficient substrate trapping.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli Proteins/ultrastructure , HSP40 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/ultrastructure , HSP70 Heat-Shock Proteins/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Binding Sites , Escherichia coli/metabolism , HSP70 Heat-Shock Proteins/physiology , HSP70 Heat-Shock Proteins/ultrastructure , Heat-Shock Proteins/metabolism , Hydrolysis , Kinetics , Models, Molecular , Molecular Chaperones/metabolism , Protein Domains/physiology
6.
Nat Commun ; 7: 13695, 2016 12 05.
Article in English | MEDLINE | ID: mdl-27917864

ABSTRACT

Hsp70 chaperones assist de novo folding of newly synthesized proteins in all cells. In yeast, the specialized Hsp70 Ssb directly binds to ribosomes. The structural basis and functional mode of recruitment of Ssb to ribosomes is not understood. Here, we present the molecular details underlying ribosome binding of Ssb in Saccharomyces cerevisiae. This interaction is multifaceted, involving the co-chaperone RAC and two specific regions within Ssb characterized by positive charges. The C-terminus of Ssb mediates the key contact and a second attachment point is provided by a KRR-motif in the substrate binding domain. Strikingly, ribosome binding of Ssb is not essential. Autonomous ribosome attachment becomes necessary if RAC is absent, suggesting a dual mode of Ssb recruitment to nascent chains. We propose, that the multilayered ribosomal interaction allows positioning of Ssb in an optimal orientation to the tunnel exit guaranteeing an efficient nascent polypeptide interaction.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Peptides/metabolism , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Computer Simulation , Conserved Sequence , Genetic Complementation Test , Genetic Pleiotropy , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/genetics , Models, Molecular , Mutation/genetics , Phenotype , Protein Binding , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Substrate Specificity
7.
Nature ; 539(7629): 448-451, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27783598

ABSTRACT

The Hsp70 system is a central hub of chaperone activity in all domains of life. Hsp70 performs a plethora of tasks, including folding assistance, protection against aggregation, protein trafficking, and enzyme activity regulation, and interacts with non-folded chains, as well as near-native, misfolded, and aggregated proteins. Hsp70 is thought to achieve its many physiological roles by binding peptide segments that extend from these different protein conformers within a groove that can be covered by an ATP-driven helical lid. However, it has been difficult to test directly how Hsp70 interacts with protein substrates in different stages of folding and how it affects their structure. Moreover, recent indications of diverse lid conformations in Hsp70-substrate complexes raise the possibility of additional interaction mechanisms. Addressing these issues is technically challenging, given the conformational dynamics of both chaperone and client, the transient nature of their interaction, and the involvement of co-chaperones and the ATP hydrolysis cycle. Here, using optical tweezers, we show that the bacterial Hsp70 homologue (DnaK) binds and stabilizes not only extended peptide segments, but also partially folded and near-native protein structures. The Hsp70 lid and groove act synergistically when stabilizing folded structures: stabilization is abolished when the lid is truncated and less efficient when the groove is mutated. The diversity of binding modes has important consequences: Hsp70 can both stabilize and destabilize folded structures, in a nucleotide-regulated manner; like Hsp90 and GroEL, Hsp70 can affect the late stages of protein folding; and Hsp70 can suppress aggregation by protecting partially folded structures as well as unfolded protein chains. Overall, these findings in the DnaK system indicate an extension of the Hsp70 canonical model that potentially affects a wide range of physiological roles of the Hsp70 system.


Subject(s)
HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , Protein Folding , Adenosine Triphosphate/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Models, Biological , Optical Tweezers , Protein Aggregates , Protein Binding , Protein Conformation , Protein Denaturation , Protein Refolding , Protein Stability , Single Molecule Imaging , Substrate Specificity
8.
Front Mol Biosci ; 2: 58, 2015.
Article in English | MEDLINE | ID: mdl-26539440

ABSTRACT

Hsp70s chaperone an amazing number and variety of cellular protein folding processes. Key to their versatility is the recognition of a short degenerate sequence motif, present in practically all polypeptides, and a bidirectional allosteric intramolecular regulation mechanism linking their N-terminal nucleotide binding domain (NBD) and their C-terminal polypeptide substrate binding domain (SBD). Through this interdomain communication ATP binding to the NBD and ATP hydrolysis control the affinity of the SBD for polypeptide substrates and substrate binding to the SBD triggers ATP hydrolysis. Genetic screens for defective variants of Hsp70s and systematic analysis of available structures of the isolated domains revealed some residues involved in allosteric control. Recent elucidation of the crystal structure of the Hsp70 homolog DnaK in the ATP bound open conformation as well as numerous NMR and mutagenesis studies bring us closer to an understanding of the communication between NBD and SBD. In this review we will discuss our current view of the allosteric control mechanism of Hsp70 chaperones.

9.
Nat Commun ; 6: 8308, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26383706

ABSTRACT

Central to the protein folding activity of Hsp70 chaperones is their ability to interact with protein substrates in an ATP-controlled manner, which relies on allosteric regulation between their nucleotide-binding (NBD) and substrate-binding domains (SBD). Here we dissect this mechanism by analysing mutant variants of the Escherichia coli Hsp70 DnaK blocked at distinct steps of allosteric communication. We show that the SBD inhibits ATPase activity by interacting with the NBD through a highly conserved hydrogen bond network, and define the signal transduction pathway that allows bound substrates to trigger ATP hydrolysis. We identify variants deficient in only one direction of allosteric control and demonstrate that ATP-induced substrate release is more important for chaperone activity than substrate-stimulated ATP hydrolysis. These findings provide evidence of an unexpected dichotomic allostery mechanism in Hsp70 chaperones and provide the basis for a comprehensive mechanical model of allostery in Hsp70s.


Subject(s)
Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , HSP70 Heat-Shock Proteins/metabolism , Allosteric Regulation , Binding Sites , Circular Dichroism , Escherichia coli Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , Mutagenesis, Site-Directed
10.
Chem Biol ; 22(1): 87-97, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25544045

ABSTRACT

The highly conserved 70 kDa heat shock proteins (Hsp70) play an integral role in proteostasis such that dysregulation has been implicated in numerous diseases. Elucidating the precise role of Hsp70 family members in the cellular context, however, has been hampered by the redundancy and intricate regulation of the chaperone network, and relatively few selective and potent tools. We have characterized a natural product, novolactone, that targets cytosolic and ER-localized isoforms of Hsp70 through a highly conserved covalent interaction at the interface between the substrate-binding and ATPase domains. Biochemical and structural analyses indicate that novolactone disrupts interdomain communication by allosterically inducing a conformational change in the Hsp70 protein to block ATP-induced substrate release and inhibit refolding activities. Thus, novolactone is a valuable tool for exploring the requirements of Hsp70 chaperones in diverse cellular contexts.


Subject(s)
Abietanes/metabolism , Biological Products/metabolism , HSP70 Heat-Shock Proteins/metabolism , Abietanes/chemistry , Adenosine Triphosphatases/metabolism , Allosteric Regulation , Binding Sites , Biological Products/chemistry , Cell Line , Crystallography, X-Ray , Endoplasmic Reticulum/metabolism , Genome, Fungal , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , Humans , Molecular Dynamics Simulation , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Substrate Specificity
11.
Mol Cell ; 48(6): 863-74, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23123194

ABSTRACT

Central to the chaperone function of Hsp70s is the transition between open and closed conformations of their polypeptide substrate binding domain (SBD), which is regulated through an allosteric mechanism via ATP binding and hydrolysis in their nucleotide binding domain (NBD). Although the structure of the closed conformation of Hsp70s is well studied, the open conformation has remained elusive. Here, we report on the 2.4 Å crystal structure of the ATP-bound open conformation of the Escherichia coli Hsp70 homolog DnaK. In the open DnaK structure, the ß sheet and α-helical lid subdomains of the SBD are detached from one another and docked to different faces of the NBD. The contacts between the ß sheet subdomain and the NBD reveal the mechanism of allosteric regulation. In addition, we demonstrate that docking of the ß sheet and α-helical lid subdomains to the NBD is a sequential process influenced by peptide and protein substrates.


Subject(s)
Adenosine Triphosphate/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , HSP70 Heat-Shock Proteins/chemistry , Amino Acid Substitution , Catalytic Domain , Crystallography, X-Ray , Escherichia coli Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , Hydrogen Bonding , Hydrolysis , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Binding , Protein Stability , Protein Structure, Quaternary , Protein Structure, Secondary , Structural Homology, Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...