Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38712168

ABSTRACT

The hexameric AAA+ disaggregase, Hsp104, collaborates with Hsp70 and Hsp40 via its autoregulatory middle domain (MD) to solubilize aggregated protein conformers. However, how ATP- or ADP-specific MD configurations regulate Hsp104 hexamers remains poorly understood. Here, we define an ATP-specific network of interprotomer contacts between nucleotide-binding domain 1 (NBD1) and MD helix L1, which tunes Hsp70 collaboration. Manipulating this network can: (a) reduce Hsp70 collaboration without enhancing activity; (b) generate Hsp104 hypomorphs that collaborate selectively with class B Hsp40s; (c) produce Hsp70-independent potentiated variants; or (d) create species barriers between Hsp104 and Hsp70. Conversely, ADP-specific intraprotomer contacts between MD helix L2 and NBD1 restrict activity, and their perturbation frequently potentiates Hsp104. Importantly, adjusting the NBD1:MD helix L1 rheostat via rational design enables finely tuned collaboration with Hsp70 to safely potentiate Hsp104, minimize off-target toxicity, and counteract FUS proteinopathy in human cells. Thus, we establish important design principles to tailor Hsp104 therapeutics.

2.
bioRxiv ; 2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37732211

ABSTRACT

RNA-binding proteins with prion-like domains, such as FUS and TDP-43, condense into functional liquids, which can transform into pathological fibrils that underpin fatal neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS)/frontotemporal dementia (FTD). Here, we define short RNAs (24-48 nucleotides) that prevent FUS fibrillization by promoting liquid phases, and distinct short RNAs that prevent and, remarkably, reverse FUS condensation and fibrillization. These activities require interactions with multiple RNA-binding domains of FUS and are encoded by RNA sequence, length, and structure. Importantly, we define a short RNA that dissolves aberrant cytoplasmic FUS condensates, restores nuclear FUS, and mitigates FUS proteotoxicity in optogenetic models and human motor neurons. Another short RNA dissolves aberrant cytoplasmic TDP-43 condensates, restores nuclear TDP-43, and mitigates TDP-43 proteotoxicity. Since short RNAs can be effectively delivered to the human brain, these oligonucleotides could have therapeutic utility for ALS/FTD and related disorders.

3.
Mol Cell ; 83(18): 3314-3332.e9, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37625404

ABSTRACT

Hsp104 is an AAA+ protein disaggregase that solubilizes and reactivates proteins trapped in aggregated states. We have engineered potentiated Hsp104 variants to mitigate toxic misfolding of α-synuclein, TDP-43, and FUS implicated in fatal neurodegenerative disorders. Though potent disaggregases, these enhanced Hsp104 variants lack substrate specificity and can have unfavorable off-target effects. Here, to lessen off-target effects, we engineer substrate-specific Hsp104 variants. By altering Hsp104 pore loops that engage substrate, we disambiguate Hsp104 variants that selectively suppress α-synuclein toxicity but not TDP-43 or FUS toxicity. Remarkably, α-synuclein-specific Hsp104 variants emerge that mitigate α-synuclein toxicity via distinct ATPase-dependent mechanisms involving α-synuclein disaggregation or detoxification of soluble α-synuclein conformers. Importantly, both types of α-synuclein-specific Hsp104 variant reduce dopaminergic neurodegeneration in a C. elegans model of Parkinson's disease more effectively than non-specific variants. We suggest that increasing the substrate specificity of enhanced disaggregases could be applied broadly to tailor therapeutics for neurodegenerative disease.


Subject(s)
Neurodegenerative Diseases , Saccharomyces cerevisiae Proteins , Animals , Humans , alpha-Synuclein/genetics , Saccharomyces cerevisiae Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism
4.
Biochem Soc Trans ; 50(2): 895-906, 2022 04 29.
Article in English | MEDLINE | ID: mdl-35356966

ABSTRACT

Numerous ATPases associated with diverse cellular activities (AAA+) proteins form hexameric, ring-shaped complexes that function via ATPase-coupled translocation of substrates across the central channel. Cryo-electron microscopy of AAA+ proteins processing substrate has revealed non-symmetric, staircase-like hexameric structures that indicate a sequential clockwise/2-residue step translocation model for these motors. However, for many of the AAA+ proteins that share similar structural features, their translocation properties have not yet been experimentally determined. In the cases where translocation mechanisms have been determined, a two-residue translocation step-size has not been resolved. In this review, we explore Hsp104, ClpB, ClpA and ClpX as examples to review the experimental methods that have been used to examine, in solution, the translocation mechanisms employed by AAA+ motor proteins. We then ask whether AAA+ motors sharing similar structural features can have different translocation mechanisms. Finally, we discuss whether a single AAA+ motor can adopt multiple translocation mechanisms that are responsive to different challenges imposed by the substrate or the environment. We suggest that AAA+ motors adopt more than one translocation mechanism and are tuned to switch to the most energetically efficient mechanism when constraints are applied.


Subject(s)
AAA Proteins , Escherichia coli Proteins , AAA Proteins/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , Escherichia coli Proteins/metabolism , Models, Molecular
5.
Nature ; 597(7874): 132-137, 2021 09.
Article in English | MEDLINE | ID: mdl-34408321

ABSTRACT

Protein quality control systems are crucial for cellular function and organismal health. At present, most known protein quality control systems are multicomponent machineries that operate via ATP-regulated interactions with non-native proteins to prevent aggregation and promote folding1, and few systems that can broadly enable protein folding by a different mechanism have been identified. Moreover, proteins that contain the extensively charged poly-Asp/Glu (polyD/E) region are common in eukaryotic proteomes2, but their biochemical activities remain undefined. Here we show that DAXX, a polyD/E protein that has been implicated in diverse cellular processes3-10, possesses several protein-folding activities. DAXX prevents aggregation, solubilizes pre-existing aggregates and unfolds misfolded species of model substrates and neurodegeneration-associated proteins. Notably, DAXX effectively prevents and reverses aggregation of its in vivo-validated client proteins, the tumour suppressor p53 and its principal antagonist MDM2. DAXX can also restore native conformation and function to tumour-associated, aggregation-prone p53 mutants, reducing their oncogenic properties. These DAXX activities are ATP-independent and instead rely on the polyD/E region. Other polyD/E proteins, including ANP32A and SET, can also function as stand-alone, ATP-independent molecular chaperones, disaggregases and unfoldases. Thus, polyD/E proteins probably constitute a multifunctional protein quality control system that operates via a distinctive mechanism.


Subject(s)
Co-Repressor Proteins/metabolism , Molecular Chaperones/metabolism , Protein Folding , Animals , Cell Line , Cells/metabolism , Evolution, Molecular , Humans , Models, Molecular , Mutation , Protein Aggregates , Protein Aggregation, Pathological/prevention & control , Protein Conformation , Protein Domains , Protein Unfolding , Proteostasis Deficiencies/prevention & control , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Elife ; 92020 12 15.
Article in English | MEDLINE | ID: mdl-33319748

ABSTRACT

The AAA+ protein disaggregase, Hsp104, increases fitness under stress by reversing stress-induced protein aggregation. Natural Hsp104 variants might exist with enhanced, selective activity against neurodegenerative disease substrates. However, natural Hsp104 variation remains largely unexplored. Here, we screened a cross-kingdom collection of Hsp104 homologs in yeast proteotoxicity models. Prokaryotic ClpG reduced TDP-43, FUS, and α-synuclein toxicity, whereas prokaryotic ClpB and hyperactive variants were ineffective. We uncovered therapeutic genetic variation among eukaryotic Hsp104 homologs that specifically antagonized TDP-43 condensation and toxicity in yeast and TDP-43 aggregation in human cells. We also uncovered distinct eukaryotic Hsp104 homologs that selectively antagonized α-synuclein condensation and toxicity in yeast and dopaminergic neurodegeneration in C. elegans. Surprisingly, this therapeutic variation did not manifest as enhanced disaggregase activity, but rather as increased passive inhibition of aggregation of specific substrates. By exploring natural tuning of this passive Hsp104 activity, we elucidated enhanced, substrate-specific agents that counter proteotoxicity underlying neurodegeneration.


Subject(s)
DNA-Binding Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Protein Aggregation, Pathological/pathology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , alpha-Synuclein/metabolism , Animals , Caenorhabditis elegans , Cell Line , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism , Escherichia coli , Genetic Variation/genetics , HEK293 Cells , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Protein Folding , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/pathology , RNA-Binding Protein FUS/metabolism , Saccharomyces cerevisiae
7.
Nat Struct Mol Biol ; 27(5): 406-416, 2020 05.
Article in English | MEDLINE | ID: mdl-32313240

ABSTRACT

The ClpAP complex is a conserved bacterial protease that unfolds and degrades proteins targeted for destruction. The ClpA double-ring hexamer powers substrate unfolding and translocation into the ClpP proteolytic chamber. Here, we determined high-resolution structures of wild-type Escherichia coli ClpAP undergoing active substrate unfolding and proteolysis. A spiral of pore loop-substrate contacts spans both ClpA AAA+ domains. Protomers at the spiral seam undergo nucleotide-specific rearrangements, supporting substrate translocation. IGL loops extend flexibly to bind the planar, heptameric ClpP surface with the empty, symmetry-mismatched IGL pocket maintained at the seam. Three different structures identify a binding-pocket switch by the IGL loop of the lowest positioned protomer, involving release and re-engagement with the clockwise pocket. This switch is coupled to a ClpA rotation and a network of conformational changes across the seam, suggesting that ClpA can rotate around the ClpP apical surface during processive steps of translocation and proteolysis.


Subject(s)
Endopeptidase Clp/chemistry , Endopeptidase Clp/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , DNA Helicases/metabolism , Endopeptidase Clp/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Models, Molecular , Multiprotein Complexes , Protein Conformation , Protein Unfolding , Trans-Activators/metabolism
8.
Proc Natl Acad Sci U S A ; 117(17): 9384-9392, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32277033

ABSTRACT

Hsp104 provides a valuable model for the many essential proteostatic functions performed by the AAA+ superfamily of protein molecular machines. We developed and used a powerful hydrogen exchange mass spectrometry (HX MS) analysis that can provide positionally resolved information on structure, dynamics, and energetics of the Hsp104 molecular machinery, even during functional cycling. HX MS reveals that the ATPase cycle is rate-limited by ADP release from nucleotide-binding domain 1 (NBD1). The middle domain (MD) serves to regulate Hsp104 activity by slowing ADP release. Mutational potentiation accelerates ADP release, thereby increasing ATPase activity. It reduces time in the open state, thereby decreasing substrate protein loss. During active cycling, Hsp104 transits repeatedly between whole hexamer closed and open states. Under diverse conditions, the shift of open/closed balance can lead to premature substrate loss, normal processing, or the generation of a strong pulling force. HX MS exposes the mechanisms of these functions at near-residue resolution.


Subject(s)
Gene Expression Regulation, Fungal/physiology , Genetic Variation , Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphate , Amino Acid Substitution , Heat-Shock Proteins/genetics , Mutation , Protein Binding , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
9.
Mol Biol Cell ; 31(3): 221-233, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31825716

ABSTRACT

Protein misfolding is a recurring phenomenon that cells must manage; otherwise misfolded proteins can aggregate and become toxic should they persist. To counter this burden, cells have evolved protein quality control (PQC) mechanisms that manage misfolded proteins. Two classes of systems that function in PQC are chaperones that aid in protein folding and ubiquitin-protein ligases that ubiquitinate misfolded proteins for proteasomal degradation. How folding and degradative PQC systems interact and coordinate their respective functions is not yet fully understood. Previous studies of PQC degradation pathways in the endoplasmic reticulum and cytosol have led to the prevailing idea that these pathways require the activity of Hsp70 chaperones. Here, we find that involvement of the budding yeast Hsp70 chaperones Ssa1 and Ssa2 in nuclear PQC degradation varies with the substrate. In particular, nuclear PQC degradation mediated by the yeast ubiquitin-protein ligase San1 often involves Ssa1/Ssa2, but San1 substrate recognition and ubiquitination can proceed without these Hsp70 chaperone functions in vivo and in vitro. Our studies provide new insights into the variability of Hsp70 chaperone involvement with a nuclear PQC degradation pathway.


Subject(s)
Adenosine Triphosphatases/genetics , HSP70 Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Cell Nucleus/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/genetics , Molecular Chaperones/metabolism , Mutation , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Domains , Protein Folding , Proteolysis , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
10.
Cell Rep ; 28(8): 2080-2095.e6, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31433984

ABSTRACT

Hsp104 is an AAA+ protein disaggregase, which can be potentiated via diverse mutations in its autoregulatory middle domain (MD) to mitigate toxic misfolding of TDP-43, FUS, and α-synuclein implicated in fatal neurodegenerative disorders. Problematically, potentiated MD variants can exhibit off-target toxicity. Here, we mine disaggregase sequence space to safely enhance Hsp104 activity via single mutations in nucleotide-binding domain 1 (NBD1) or NBD2. Like MD variants, NBD variants counter TDP-43, FUS, and α-synuclein toxicity and exhibit elevated ATPase and disaggregase activity. Unlike MD variants, non-toxic NBD1 and NBD2 variants emerge that rescue TDP-43, FUS, and α-synuclein toxicity. Potentiating substitutions alter NBD1 residues that contact ATP, ATP-binding residues, or the MD. Mutating the NBD2 protomer interface can also safely ameliorate Hsp104. Thus, we disambiguate allosteric regulation of Hsp104 by several tunable structural contacts, which can be engineered to spawn enhanced therapeutic disaggregases with minimal off-target toxicity.


Subject(s)
DNA-Binding Proteins/toxicity , Heat-Shock Proteins/metabolism , RNA-Binding Protein FUS/toxicity , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , alpha-Synuclein/toxicity , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Azetidinecarboxylic Acid/pharmacology , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense/genetics , Protein Aggregates , Protein Domains , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Temperature
11.
Nat Commun ; 10(1): 2393, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31160557

ABSTRACT

Bacterial ClpB and yeast Hsp104 are homologous Hsp100 protein disaggregases that serve critical functions in proteostasis by solubilizing protein aggregates. Two AAA+ nucleotide binding domains (NBDs) power polypeptide translocation through a central channel comprised of a hexameric spiral of protomers that contact substrate via conserved pore-loop interactions. Here we report cryo-EM structures of a hyperactive ClpB variant bound to the model substrate, casein in the presence of slowly hydrolysable ATPγS, which reveal the translocation mechanism. Distinct substrate-gripping interactions are identified for NBD1 and NBD2 pore loops. A trimer of N-terminal domains define a channel entrance that binds the polypeptide substrate adjacent to the topmost NBD1 contact. NBD conformations at the seam interface reveal how ATP hydrolysis-driven substrate disengagement and re-binding are precisely tuned to drive a directional, stepwise translocation cycle.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Caseins/metabolism , Endopeptidase Clp/ultrastructure , Escherichia coli Proteins/ultrastructure , Escherichia coli/metabolism , Heat-Shock Proteins/ultrastructure , Protein Transport , AAA Domain , Adenosine Triphosphate/metabolism , Cryoelectron Microscopy , Endopeptidase Clp/metabolism , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/metabolism , Hydrolysis , Models, Molecular , Peptides/metabolism , Protein Aggregates , Protein Subunits/metabolism
12.
Biophys J ; 116(10): 1856-1872, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31027887

ABSTRACT

Heat shock protein (Hsp) 104 is a hexameric ATPases associated with diverse cellular activities motor protein that enables cells to survive extreme stress. Hsp104 couples the energy of ATP binding and hydrolysis to solubilize proteins trapped in aggregated structures. The mechanism by which Hsp104 disaggregates proteins is not completely understood but may require Hsp104 to partially or completely translocate polypeptides across its central channel. Here, we apply transient state, single turnover kinetics to investigate the ATP-dependent translocation of soluble polypeptides by Hsp104 and Hsp104A503S, a potentiated variant developed to resolve misfolded conformers implicated in neurodegenerative disease. We establish that Hsp104 and Hsp104A503S can operate as nonprocessive translocases for soluble substrates, indicating a "partial threading" model of translocation. Remarkably, Hsp104A503S exhibits altered coupling of ATP binding to translocation and decelerated dissociation from polypeptide substrate compared to Hsp104. This altered coupling and prolonged substrate interaction likely increases entropic pulling forces, thereby enabling more effective aggregate dissolution by Hsp104A503S.


Subject(s)
Heat-Shock Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Heat-Shock Proteins/genetics , Hydrolysis , Kinetics , Mutant Proteins/metabolism , Peptides/metabolism , Protein Aggregates , Protein Binding , Protein Conformation , Protein Folding , Saccharomyces cerevisiae Proteins/genetics , Structure-Activity Relationship
13.
Proc Natl Acad Sci U S A ; 116(15): 7333-7342, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30918129

ABSTRACT

Hsp104 is a large AAA+ molecular machine that can rescue proteins trapped in amorphous aggregates and stable amyloids by drawing substrate protein into its central pore. Recent cryo-EM studies image Hsp104 at high resolution. We used hydrogen exchange mass spectrometry analysis (HX MS) to resolve and characterize all of the functionally active and inactive elements of Hsp104, many not accessible to cryo-EM. At a global level, HX MS confirms the one noncanonical interprotomer interface in the Hsp104 hexamer as a marker for the spiraled conformation revealed by cryo-EM and measures its fast conformational cycling under ATP hydrolysis. Other findings enable reinterpretation of the apparent variability of the regulatory middle domain. With respect to detailed mechanism, HX MS determines the response of each Hsp104 structural element to the different bound adenosine nucleotides (ADP, ATP, AMPPNP, and ATPγS). They are distinguished most sensitively by the two Walker A nucleotide-binding segments. Binding of the ATP analog, ATPγS, tightly restructures the Walker A segments and drives the global open-to-closed/extended transition. The global transition carries part of the ATP/ATPγS-binding energy to the somewhat distant central pore. The pore constricts and the tyrosine and other pore-related loops become more tightly structured, which seems to reflect the energy-requiring directional pull that translocates the substrate protein. ATP hydrolysis to ADP allows Hsp104 to relax back to its lowest energy open state ready to restart the cycle.


Subject(s)
Adenine Nucleotides/chemistry , Heat-Shock Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Adenine Nucleotides/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Mass Spectrometry , Protein Domains , Protein Structure, Quaternary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Structure-Activity Relationship
14.
FEMS Yeast Res ; 18(5)2018 08 01.
Article in English | MEDLINE | ID: mdl-29788207

ABSTRACT

Hsp104 is a hexameric AAA + ATPase and protein disaggregase found in yeast, which can be potentiated via mutations in its middle domain (MD) to counter toxic phase separation by TDP-43, FUS and α-synuclein connected to devastating neurodegenerative disorders. Subtle missense mutations in the Hsp104 MD can enhance activity, indicating that post-translational modification of specific MD residues might also potentiate Hsp104. Indeed, several serine and threonine residues throughout Hsp104 can be phosphorylated in vivo. Here, we introduce phosphomimetic aspartate or glutamate residues at these positions and assess Hsp104 activity. Remarkably, phosphomimetic T499D/E and S535D/E mutations in the MD enable Hsp104 to counter TDP-43, FUS and α-synuclein aggregation and toxicity in yeast, whereas T499A/V/I and S535A do not. Moreover, Hsp104T499E and Hsp104S535E exhibit enhanced ATPase activity and Hsp70-independent disaggregase activity in vitro. We suggest that phosphorylation of T499 or S535 may elicit enhanced Hsp104 disaggregase activity in a reversible and regulated manner.


Subject(s)
Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Mutation, Missense , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Aspartic Acid , Glutamic Acid , Models, Molecular , Phosphorylation , Protein Folding , Saccharomyces cerevisiae/metabolism
15.
Science ; 357(6348): 273-279, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28619716

ABSTRACT

Hsp100 polypeptide translocases are conserved members of the AAA+ family (adenosine triphosphatases associated with diverse cellular activities) that maintain proteostasis by unfolding aberrant and toxic proteins for refolding or proteolytic degradation. The Hsp104 disaggregase from Saccharomyces cerevisiae solubilizes stress-induced amorphous aggregates and amyloids. The structural basis for substrate recognition and translocation is unknown. Using a model substrate (casein), we report cryo-electron microscopy structures at near-atomic resolution of Hsp104 in different translocation states. Substrate interactions are mediated by conserved, pore-loop tyrosines that contact an 80-angstrom-long unfolded polypeptide along the axial channel. Two protomers undergo a ratchet-like conformational change that advances pore loop-substrate interactions by two amino acids. These changes are coupled to activation of specific nucleotide hydrolysis sites and, when transmitted around the hexamer, reveal a processive rotary translocation mechanism and substrate-responsive flexibility during Hsp104-catalyzed disaggregation.


Subject(s)
Heat-Shock Proteins/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Caseins/metabolism , Cryoelectron Microscopy , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins/ultrastructure , Hydrolysis , Nucleotides/chemistry , Nucleotides/metabolism , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Promoter Regions, Genetic , Protein Domains , Protein Transport , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/ultrastructure , Substrate Specificity , Tyrosine/genetics , Tyrosine/metabolism
16.
Biochemistry ; 56(15): 2071-2075, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28379007

ABSTRACT

Recent Hsp104 structural studies have reported both planar and helical models of the hexameric structure. The conformation of Hsp104 monomers within the hexamer is affected by nucleotide ligation. After nucleotide-driven hexamer formation, Hsp104-catalyzed disruption of protein aggregates requires binding to the peptide substrate. Here, we examine the oligomeric state of Hsp104 and its peptide binding competency in the absence of nucleotide and in the presence of ADP, ATPγS, AMPPNP, or AMPPCP. Surprisingly, we found that only ATPγS facilitates avid peptide binding by Hsp104. We propose that the modulation between high- and low-peptide affinity states observed with these ATP analogues is an important component of the disaggregation mechanism of Hsp104.


Subject(s)
Heat-Shock Proteins/metabolism , Peptides/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Protein Binding
17.
Biochemistry ; 55(12): 1758-71, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26891079

ABSTRACT

Escherichia coli caseinolytic peptidase B (ClpB) is a molecular chaperone with the unique ability to catalyze protein disaggregation in collaboration with the KJE system of chaperones. Like many AAA+ molecular motors, ClpB assembles into hexameric rings, and this reaction is thermodynamically linked to nucleotide binding. Here we show that ClpB exists in a dynamic equilibrium of monomers, dimers, tetramers, and hexamers in the presence of both limiting and excess ATPγS. We find that ClpB monomer is only able to bind one nucleotide, whereas all 12 sites in the hexameric ring are bound by nucleotide at saturating concentrations. Interestingly, dimers and tetramers exhibit stoichiometries of ∼3 and 7, respectively, which is one fewer than the maximum number of binding sites in the formed oligomer. This observation suggests an open conformation for the intermediates based on the need for an adjacent monomer to fully form the binding pocket. We also report the protein-protein interaction constants for dimers, tetramers, and hexamers and their dependencies on nucleotide. These interaction constants make it possible to predict the concentration of hexamers present and able to bind to cochaperones and polypeptide substrates. Such information is essential for the interpretation of many in vitro studies. Finally, the strategies presented here are broadly applicable to a large number of AAA+ molecular motors that assemble upon nucleotide binding and interact with partner proteins.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Protein Structure, Quaternary , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Binding Sites/physiology , Endopeptidase Clp , Nucleotides/chemistry , Nucleotides/metabolism , Protein Binding/physiology , Protein Structure, Quaternary/physiology
18.
Methods Enzymol ; 562: 161-86, 2015.
Article in English | MEDLINE | ID: mdl-26412651

ABSTRACT

The ATPases associated with diverse cellular activities (AAA+) is a large superfamily of proteins involved in a broad array of biological processes. Many members of this family require nucleotide binding to assemble into their final active hexameric form. We have been studying two example members, Escherichia coli ClpA and ClpB. These two enzymes are active as hexameric rings that both require nucleotide binding for assembly. Our studies have shown that they both reside in a monomer, dimer, tetramer, and hexamer equilibrium, and this equilibrium is thermodynamically linked to nucleotide binding. Moreover, we are finding that the kinetics of the assembly reaction are very different for the two enzymes. Here, we present our strategy for determining the self-association constants in the absence of nucleotide to set the stage for the analysis of nucleotide binding from other experimental approaches including analytical ultracentrifugation.


Subject(s)
Endopeptidase Clp/chemistry , Escherichia coli Proteins/chemistry , Heat-Shock Proteins/chemistry , Endopeptidase Clp/isolation & purification , Escherichia coli Proteins/isolation & purification , Heat-Shock Proteins/isolation & purification , Kinetics , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Thermodynamics , Ultracentrifugation
19.
Biochem J ; 470(1): 39-52, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26251445

ABSTRACT

Escherichia coli caseinolytic protease (Clp)B is a hexameric AAA+ [expanded superfamily of AAA (ATPase associated with various cellular activities)] enzyme that has the unique ability to catalyse protein disaggregation. Such enzymes are essential for proteome maintenance. Based on structural comparisons to homologous enzymes involved in ATP-dependent proteolysis and clever protein engineering strategies, it has been reported that ClpB translocates polypeptide through its axial channel. Using single-turnover fluorescence and anisotropy experiments we show that ClpB is a non-processive polypeptide translocase that catalyses disaggregation by taking one or two translocation steps followed by rapid dissociation. Using single-turnover FRET experiments we show that ClpB containing the IGL loop from ClpA does not translocate substrate through its axial channel and into ClpP for proteolytic degradation. Rather, ClpB containing the IGL loop dysregulates ClpP leading to non-specific proteolysis reminiscent of ADEP (acyldepsipeptide) dysregulation. Our results support a molecular mechanism where ClpB catalyses protein disaggregation by tugging and releasing exposed tails or loops.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Peptides/chemistry , Peptides/metabolism , Amino Acid Sequence , Bacterial Translocation/physiology , Endopeptidase Clp , Escherichia coli Proteins/genetics , Heat-Shock Proteins/genetics , Molecular Sequence Data , Peptides/genetics , Protein Structure, Secondary
20.
Proteins ; 83(11): 2008-24, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26313457

ABSTRACT

Escherichia coli ClpB is a heat shock protein that belongs to the AAA+ protein superfamily. Studies have shown that ClpB and its homologue in yeast, Hsp104, can disrupt protein aggregates in vivo. It is thought that ClpB requires binding of nucleoside triphosphate to assemble into hexameric rings with protein binding activity. In addition, it is widely assumed that ClpB is uniformly hexameric in the presence of nucleotides. Here we report, in the absence of nucleotide, that increasing ClpB concentration leads to ClpB hexamer formation, decreasing NaCl concentration stabilizes ClpB hexamers, and the ClpB assembly reaction is best described by a monomer, dimer, tetramer, hexamer equilibrium under the three salt concentrations examined. Further, we found that ClpB oligomers exhibit relatively fast dissociation on the time scale of sedimentation. We anticipate our studies on ClpB assembly to be a starting point to understand how ClpB assembly is linked to the binding and disaggregation of denatured proteins.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Endopeptidase Clp , Protein Multimerization , Protein Unfolding , Sodium Chloride , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...