Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.327
Filter
1.
Cell ; 181(3): 637-652.e15, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32272059

ABSTRACT

Many cytosolic proteins lacking a signal peptide, called leaderless cargoes, are secreted through unconventional secretion. Vesicle trafficking is a major pathway involved. It is unclear how leaderless cargoes enter into the vesicle. Here, we find a translocation pathway regulating vesicle entry and secretion of leaderless cargoes. We identify TMED10 as a protein channel for the vesicle entry and secretion of many leaderless cargoes. The interaction of TMED10 C-terminal region with a motif in the cargo accounts for the selective release of the cargoes. In an in vitro reconstitution assay, TMED10 directly mediates the membrane translocation of leaderless cargoes into the liposome, which is dependent on protein unfolding and enhanced by HSP90s. In the cell, TMED10 localizes on the endoplasmic reticulum (ER)-Golgi intermediate compartment and directs the entry of cargoes into this compartment. Furthermore, cargo induces the formation of TMED10 homo-oligomers which may act as a protein channel for cargo translocation.


Subject(s)
Protein Translocation Systems/metabolism , Vesicular Transport Proteins/metabolism , Animals , Biological Transport , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Humans , Mice , Mice, Inbred C57BL , Protein Sorting Signals , Protein Translocation Systems/physiology , Protein Transport/physiology , Proteins/metabolism , Secretory Pathway , Vesicular Transport Proteins/physiology
2.
Cell ; 179(1): 205-218.e21, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31522888

ABSTRACT

The molecular chaperone HSP90 facilitates the folding of several client proteins, including innate immune receptors and protein kinases. HSP90 is an essential component of plant and animal immunity, yet pathogenic strategies that directly target the chaperone have not been described. Here, we identify the HopBF1 family of bacterial effectors as eukaryotic-specific HSP90 protein kinases. HopBF1 adopts a minimal protein kinase fold that is recognized by HSP90 as a host client. As a result, HopBF1 phosphorylates HSP90 to completely inhibit the chaperone's ATPase activity. We demonstrate that phosphorylation of HSP90 prevents activation of immune receptors that trigger the hypersensitive response in plants. Consequently, HopBF1-dependent phosphorylation of HSP90 is sufficient to induce severe disease symptoms in plants infected with the bacterial pathogen, Pseudomonas syringae. Collectively, our results uncover a family of bacterial effector kinases with toxin-like properties and reveal a previously unrecognized betrayal mechanism by which bacterial pathogens modulate host immunity.


Subject(s)
Arabidopsis Proteins/metabolism , Bacterial Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Mimicry/immunology , Plant Immunity/physiology , Adenosine Triphosphatases/metabolism , Arabidopsis/immunology , Arabidopsis/metabolism , Arabidopsis/microbiology , Bacterial Proteins/chemistry , HEK293 Cells , HSP90 Heat-Shock Proteins/chemistry , HeLa Cells , Host Microbial Interactions/immunology , Humans , Phosphorylation , Plasmids/genetics , Protein Binding , Protein Folding , Protein Kinases/metabolism , Pseudomonas syringae/metabolism , Saccharomyces cerevisiae/metabolism
3.
Cell ; 173(1): 260-274.e25, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29551266

ABSTRACT

Protein degradation plays important roles in biological processes and is tightly regulated. Further, targeted proteolysis is an emerging research tool and therapeutic strategy. However, proteome-wide technologies to investigate the causes and consequences of protein degradation in biological systems are lacking. We developed "multiplexed proteome dynamics profiling" (mPDP), a mass-spectrometry-based approach combining dynamic-SILAC labeling with isobaric mass tagging for multiplexed analysis of protein degradation and synthesis. In three proof-of-concept studies, we uncover different responses induced by the bromodomain inhibitor JQ1 versus a JQ1 proteolysis targeting chimera; we elucidate distinct modes of action of estrogen receptor modulators; and we comprehensively classify HSP90 clients based on their requirement for HSP90 constitutively or during synthesis, demonstrating that constitutive HSP90 clients have lower thermal stability than non-clients, have higher affinity for the chaperone, vary between cell types, and change upon external stimuli. These findings highlight the potential of mPDP to identify dynamically controlled degradation mechanisms in cellular systems.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Proteome/analysis , Proteomics/methods , Azepines/chemistry , Azepines/metabolism , Azepines/pharmacology , Cell Line , Chromatography, High Pressure Liquid , Cluster Analysis , Estradiol/pharmacology , Humans , Isotope Labeling , Jurkat Cells , MCF-7 Cells , Neoplasm Proteins/metabolism , Proteins/antagonists & inhibitors , Proteins/metabolism , Proteolysis/drug effects , Receptors, Estrogen/metabolism , Tandem Mass Spectrometry , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
4.
Cell ; 168(5): 856-866.e12, 2017 Feb 23.
Article in English | MEDLINE | ID: mdl-28215707

ABSTRACT

HSP90 acts as a protein-folding buffer that shapes the manifestations of genetic variation in model organisms. Whether HSP90 influences the consequences of mutations in humans, potentially modifying the clinical course of genetic diseases, remains unknown. By mining data for >1,500 disease-causing mutants, we found a strong correlation between reduced phenotypic severity and a dominant (HSP90 ≥ HSP70) increase in mutant engagement by HSP90. Examining the cancer predisposition syndrome Fanconi anemia in depth revealed that mutant FANCA proteins engaged predominantly by HSP70 had severely compromised function. In contrast, the function of less severe mutants was preserved by a dominant increase in HSP90 binding. Reducing HSP90's buffering capacity with inhibitors or febrile temperatures destabilized HSP90-buffered mutants, exacerbating FA-related chemosensitivities. Strikingly, a compensatory FANCA somatic mutation from an "experiment of nature" in monozygotic twins both prevented anemia and reduced HSP90 binding. These findings provide one plausible mechanism for the variable expressivity and environmental sensitivity of genetic diseases.


Subject(s)
Fanconi Anemia/genetics , Fanconi Anemia/pathology , HSP90 Heat-Shock Proteins/genetics , Protein Folding , Fanconi Anemia/metabolism , Fanconi Anemia Complementation Group A Protein/chemistry , Fanconi Anemia Complementation Group A Protein/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Mutation, Missense , Protein Interaction Domains and Motifs , Stress, Physiological , Twins, Monozygotic
5.
Cell ; 170(5): 845-859.e19, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28823557

ABSTRACT

The lateral ventricle subventricular zone (SVZ) is a frequent and consequential site of pediatric and adult glioma spread, but the cellular and molecular mechanisms mediating this are poorly understood. We demonstrate that neural precursor cell (NPC):glioma cell communication underpins this propensity of glioma to colonize the SVZ through secretion of chemoattractant signals toward which glioma cells home. Biochemical, proteomic, and functional analyses of SVZ NPC-secreted factors revealed the neurite outgrowth-promoting factor pleiotrophin, along with required binding partners SPARC/SPARCL1 and HSP90B, as key mediators of this chemoattractant effect. Pleiotrophin expression is strongly enriched in the SVZ, and pleiotrophin knock down starkly reduced glioma invasion of the SVZ in the murine brain. Pleiotrophin, in complex with the binding partners, activated glioma Rho/ROCK signaling, and ROCK inhibition decreased invasion toward SVZ NPC-secreted factors. These findings demonstrate a pathogenic role for NPC:glioma interactions and potential therapeutic targets to limit glioma invasion. PAPERCLIP.


Subject(s)
Brain Neoplasms/pathology , Carrier Proteins/metabolism , Cytokines/metabolism , Glioma/pathology , Lateral Ventricles/pathology , Neoplasm Invasiveness/pathology , Aged , Animals , Brain Neoplasms/metabolism , Cell Communication , Child , Drug Delivery Systems , Female , Glioma/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heterografts , Humans , Lateral Ventricles/metabolism , Male , Mice , Neoplasm Transplantation , Signal Transduction , rho GTP-Binding Proteins/metabolism
6.
Mol Cell ; 83(12): 2035-2044.e7, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37295430

ABSTRACT

Molecular chaperones govern proteome health to support cell homeostasis. An essential eukaryotic component of the chaperone system is Hsp90. Using a chemical-biology approach, we characterized the features driving the Hsp90 physical interactome. We found that Hsp90 associated with ∼20% of the yeast proteome using its three domains to preferentially target intrinsically disordered regions (IDRs) of client proteins. Hsp90 selectively utilized an IDR to regulate client activity as well as maintained IDR-protein health by preventing the transition to stress granules or P-bodies at physiological temperatures. We also discovered that Hsp90 controls the fidelity of ribosome initiation that triggers a heat shock response when disrupted. Our study provides insights into how this abundant molecular chaperone supports a dynamic and healthy native protein landscape.


Subject(s)
Intrinsically Disordered Proteins , Molecular Chaperones , Proteome , Humans , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Proteome/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Intrinsically Disordered Proteins/metabolism
7.
Mol Cell ; 83(11): 1903-1920.e12, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37267907

ABSTRACT

Exercise benefits the human body in many ways. Irisin is secreted by muscle, increased with exercise, and conveys physiological benefits, including improved cognition and resistance to neurodegeneration. Irisin acts via αV integrins; however, a mechanistic understanding of how small polypeptides like irisin can signal through integrins is poorly understood. Using mass spectrometry and cryo-EM, we demonstrate that the extracellular heat shock protein 90α (eHsp90α) is secreted by muscle with exercise and activates integrin αVß5. This allows for high-affinity irisin binding and signaling through an Hsp90α/αV/ß5 complex. By including hydrogen/deuterium exchange data, we generate and experimentally validate a 2.98 Å RMSD irisin/αVß5 complex docking model. Irisin binds very tightly to an alternative interface on αVß5 distinct from that used by known ligands. These data elucidate a non-canonical mechanism by which a small polypeptide hormone like irisin can function through an integrin receptor.


Subject(s)
Cell Communication , Fibronectins , Humans , Fibronectins/metabolism , Signal Transduction
8.
Mol Cell ; 82(3): 555-569.e7, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35063133

ABSTRACT

In the eukaryotic cytosol, the Hsp70 and the Hsp90 chaperone machines work in tandem with the maturation of a diverse array of client proteins. The transfer of nonnative clients between these systems is essential to the chaperoning process, but how it is regulated is still not clear. We discovered that NudC is an essential transfer factor with an unprecedented mode of action: NudC interacts with Hsp40 in Hsp40-Hsp70-client complexes and displaces Hsp70. Then, the interaction of NudC with Hsp90 allows the direct transfer of Hsp40-bound clients to Hsp90 for further processing. Consistent with this mechanism, NudC increases client activation in vitro as well as in cells and is essential for cellular viability. Together, our results show the complexity of the cooperation between the major chaperone machineries in the eukaryotic cytosol.


Subject(s)
Cell Cycle Proteins/metabolism , HSP40 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Nuclear Proteins/metabolism , Binding Sites , Cell Cycle Proteins/genetics , Cell Survival , HEK293 Cells , HSP40 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/genetics , Humans , K562 Cells , Kinetics , Molecular Docking Simulation , Nuclear Proteins/genetics , Protein Binding , Protein Folding , Protein Interaction Domains and Motifs , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Mol Cell ; 82(8): 1543-1556.e6, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35176233

ABSTRACT

Folding of stringent clients requires transfer from Hsp70 to Hsp90. The co-chaperone Hop physically connects the chaperone machineries. Here, we define its role from the remodeling of Hsp70/40-client complexes to the mechanism of client transfer and the conformational switching from stalled to active client-processing states of Hsp90. We show that Hsp70 together with Hsp40 completely unfold a stringent client, the glucocorticoid receptor ligand-binding domain (GR-LBD) in large assemblies. Hop remodels these for efficient transfer onto Hsp90. As p23 enters, Hsp70 leaves the complex via switching between binding sites in Hop. Current concepts assume that to proceed to client folding, Hop dissociates and the co-chaperone p23 stabilizes the Hsp90 closed state. In contrast, we show that p23 functionally interacts with Hop, relieves the stalling Hsp90-Hop interaction, and closes Hsp90. This reaction allows folding of the client and is thus the key regulatory step for the progression of the chaperone cycle.


Subject(s)
Protein Folding , Pyridinolcarbamate , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Binding , Receptors, Glucocorticoid/metabolism
10.
Mol Cell ; 81(6): 1170-1186.e10, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33571422

ABSTRACT

The complex architecture of transmembrane proteins requires quality control (QC) of folding, membrane positioning, and trafficking as prerequisites for cellular homeostasis and intercellular communication. However, it has remained unclear whether transmembrane protein-specific QC hubs exist. Here we identify cereblon (CRBN), the target of immunomodulatory drugs (IMiDs), as a co-chaperone that specifically determines chaperone activity of HSP90 toward transmembrane proteins by means of counteracting AHA1. This function is abrogated by IMiDs, which disrupt the interaction of CRBN with HSP90. Among the multiple transmembrane protein clients of CRBN-AHA1-HSP90 revealed by cell surface proteomics, we identify the amino acid transporter LAT1/CD98hc as a determinant of IMiD activity in multiple myeloma (MM) and present an Anticalin-based CD98hc radiopharmaceutical for MM radio-theranostics. These data establish the CRBN-AHA1-HSP90 axis in the biogenesis of transmembrane proteins, link IMiD activity to tumor metabolism, and nominate CD98hc and LAT1 as attractive diagnostic and therapeutic targets in MM.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , HSP90 Heat-Shock Proteins/metabolism , Immunologic Factors/pharmacology , Large Neutral Amino Acid-Transporter 1/metabolism , Molecular Chaperones/metabolism , Multiple Myeloma/metabolism , Neoplasm Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , HEK293 Cells , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Tumor Cells, Cultured
11.
Mol Cell ; 81(17): 3496-3508.e5, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34380015

ABSTRACT

The Hsp90 chaperone promotes folding and activation of hundreds of client proteins in the cell through an ATP-dependent conformational cycle guided by distinct cochaperone regulators. The FKBP51 immunophilin binds Hsp90 with its tetratricopeptide repeat (TPR) domain and catalyzes peptidyl-prolyl isomerase (PPIase) activity during folding of kinases, nuclear receptors, and tau. Here we determined the cryoelectron microscopy (cryo-EM) structure of the human Hsp90:FKBP51:p23 complex to 3.3 Å, which, together with mutagenesis and crosslinking analyses, reveals the basis for cochaperone binding to Hsp90 during client maturation. A helix extension in the TPR functions as a key recognition element, interacting across the Hsp90 C-terminal dimer interface presented in the closed, ATP conformation. The PPIase domain is positioned along the middle domain, adjacent to Hsp90 client binding sites, whereas a single p23 makes stabilizing interactions with the N-terminal dimer. With this architecture, FKBP51 is positioned to act on specific client residues presented during Hsp90-catalyzed remodeling.


Subject(s)
HSP90 Heat-Shock Proteins/chemistry , Tacrolimus Binding Proteins/chemistry , Amino Acid Sequence , Binding Sites , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/metabolism , Cryoelectron Microscopy/methods , HSP90 Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Molecular Conformation , Protein Binding , Tacrolimus Binding Proteins/metabolism , Tumor Protein, Translationally-Controlled 1
12.
Mol Cell ; 81(14): 2914-2928.e7, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34107307

ABSTRACT

Molecular chaperones assist with protein folding by interacting with nascent polypeptide chains (NCs) during translation. Whether the ribosome can sense chaperone defects and, in response, abort translation of misfolding NCs has not yet been explored. Here we used quantitative proteomics to investigate the ribosome-associated chaperone network in E. coli and the consequences of its dysfunction. Trigger factor and the DnaK (Hsp70) system are the major NC-binding chaperones. HtpG (Hsp90), GroEL, and ClpB contribute increasingly when DnaK is deficient. Surprisingly, misfolding because of defects in co-translational chaperone function or amino acid analog incorporation results in recruitment of the non-canonical release factor RF3. RF3 recognizes aberrant NCs and then moves to the peptidyltransferase site to cooperate with RF2 in mediating chain termination, facilitating clearance by degradation. This function of RF3 reduces the accumulation of misfolded proteins and is critical for proteostasis maintenance and cell survival under conditions of limited chaperone availability.


Subject(s)
Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Molecular Chaperones/metabolism , Protein Biosynthesis/physiology , Amino Acids/metabolism , Cell Survival/physiology , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Peptide Termination Factors/metabolism , Peptidyl Transferases/metabolism , Protein Binding/physiology , Protein Folding , Proteomics/methods , Proteostasis/physiology , Ribosomes/metabolism
13.
Mol Cell ; 81(13): 2851-2867.e7, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34118193

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19). SARS-CoV-2 relies on cellular RNA-binding proteins (RBPs) to replicate and spread, although which RBPs control its life cycle remains largely unknown. Here, we employ a multi-omic approach to identify systematically and comprehensively the cellular and viral RBPs that are involved in SARS-CoV-2 infection. We reveal that SARS-CoV-2 infection profoundly remodels the cellular RNA-bound proteome, which includes wide-ranging effects on RNA metabolic pathways, non-canonical RBPs, and antiviral factors. Moreover, we apply a new method to identify the proteins that directly interact with viral RNA, uncovering dozens of cellular RBPs and six viral proteins. Among them are several components of the tRNA ligase complex, which we show regulate SARS-CoV-2 infection. Furthermore, we discover that available drugs targeting host RBPs that interact with SARS-CoV-2 RNA inhibit infection. Collectively, our results uncover a new universe of host-virus interactions with potential for new antiviral therapies against COVID-19.


Subject(s)
COVID-19/metabolism , Proteome/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , SARS-CoV-2/physiology , Viral Proteins/metabolism , Virus Replication/physiology , A549 Cells , COVID-19/genetics , Humans , Proteome/genetics , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Viral Proteins/genetics
14.
Trends Biochem Sci ; 49(1): 38-51, 2024 01.
Article in English | MEDLINE | ID: mdl-37980187

ABSTRACT

Molecular chaperones play central roles in sustaining protein homeostasis and preventing protein aggregation. Most studies of these systems have been performed in bulk, providing averaged measurements, though recent single-molecule approaches have provided an in-depth understanding of the molecular mechanisms of their activities and structural rearrangements during substrate recognition. Chaperone activities have been observed to be substrate specific, with some associated with ATP-dependent structural dynamics and others via interactions with co-chaperones. This Review aims to describe the novel mechanisms of molecular chaperones as revealed by single-molecule approaches, and to provide insights into their functioning and its implications for protein homeostasis and human diseases.


Subject(s)
Molecular Chaperones , Protein Folding , Humans , Molecular Chaperones/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism
15.
Trends Biochem Sci ; 49(9): 752-753, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38906726

ABSTRACT

A recent study by Amankwah et al. reports how co-chaperone proteins and ATP hydrolysis fine-tune the function of endoplasmic reticulum (ER)-resident Hsp90 paralog Grp94.


Subject(s)
Molecular Chaperones , Humans , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/chemistry , Endoplasmic Reticulum/metabolism , Animals , Adenosine Triphosphate/metabolism
16.
EMBO J ; 43(12): 2294-2307, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719995

ABSTRACT

Organisms rely on mutations to fuel adaptive evolution. However, many mutations impose a negative effect on fitness. Cells may have therefore evolved mechanisms that affect the phenotypic effects of mutations, thus conferring mutational robustness. Specifically, so-called buffer genes are hypothesized to interact directly or indirectly with genetic variation and reduce its effect on fitness. Environmental or genetic perturbations can change the interaction between buffer genes and genetic variation, thereby unmasking the genetic variation's phenotypic effects and thus providing a source of variation for natural selection to act on. This review provides an overview of our understanding of mutational robustness and buffer genes, with the chaperone gene HSP90 as a key example. It discusses whether buffer genes merely affect standing variation or also interact with de novo mutations, how mutational robustness could influence evolution, and whether mutational robustness might be an evolved trait or rather a mere side-effect of complex genetic interactions.


Subject(s)
Evolution, Molecular , HSP90 Heat-Shock Proteins , Mutation , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Selection, Genetic , Genetic Variation , Humans , Animals , Genetic Fitness
17.
Immunity ; 50(1): 137-151.e6, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30650373

ABSTRACT

Fever is an evolutionarily conserved response that confers survival benefits during infection. However, the underlying mechanism remains obscure. Here, we report that fever promoted T lymphocyte trafficking through heat shock protein 90 (Hsp90)-induced α4 integrin activation and signaling in T cells. By inducing selective binding of Hsp90 to α4 integrins, but not ß2 integrins, fever increased α4-integrin-mediated T cell adhesion and transmigration. Mechanistically, Hsp90 bound to the α4 tail and activated α4 integrins via inside-out signaling. Moreover, the N and C termini of one Hsp90 molecule simultaneously bound to two α4 tails, leading to dimerization and clustering of α4 integrins on the cell membrane and subsequent activation of the FAK-RhoA pathway. Abolishment of Hsp90-α4 interaction inhibited fever-induced T cell trafficking to draining lymph nodes and impaired the clearance of bacterial infection. Our findings identify the Hsp90-α4-integrin axis as a thermal sensory pathway that promotes T lymphocyte trafficking and enhances immune surveillance during infection.


Subject(s)
Fever/immunology , HSP90 Heat-Shock Proteins/metabolism , Integrin alpha4/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/immunology , T-Lymphocytes/immunology , Animals , Bacterial Load , Cell Adhesion , Cell Movement , Dimerization , Focal Adhesion Kinase 1/metabolism , Immunologic Surveillance , Integrin alpha4/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Signal Transduction , rhoA GTP-Binding Protein/metabolism
18.
Trends Biochem Sci ; 48(8): 662-664, 2023 08.
Article in English | MEDLINE | ID: mdl-37328388

ABSTRACT

The interactions of molecular chaperones with clients can be regulated by chaperone post-translational modification (PTMs) collectively known as the 'chaperone code'. What is less understood is how PTMs on client proteins may impact chaperone-client interactions. In this forum, we discuss the possibility of a 'client code'.


Subject(s)
HSP90 Heat-Shock Proteins , Molecular Chaperones , Humans , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Processing, Post-Translational , HSP70 Heat-Shock Proteins/metabolism , Protein Binding
19.
Mol Cell ; 74(1): 73-87.e8, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30876805

ABSTRACT

The Hsp90 chaperone machinery in eukaryotes comprises a number of distinct accessory factors. Cns1 is one of the few essential co-chaperones in yeast, but its structure and function remained unknown. Here, we report the X-ray structure of the Cns1 fold and NMR studies on the partly disordered, essential segment of the protein. We demonstrate that Cns1 is important for maintaining translation elongation, specifically chaperoning the elongation factor eEF2. In this context, Cns1 interacts with the novel co-factor Hgh1 and forms a quaternary complex together with eEF2 and Hsp90. The in vivo folding and solubility of eEF2 depend on the presence of these proteins. Chaperoning of eEF2 by Cns1 is essential for yeast viability and requires a defined subset of the Hsp90 machinery as well as the identified eEF2 recruiting factor Hgh1.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Chaperones/metabolism , Peptide Chain Elongation, Translational , Peptide Elongation Factor 2/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Crystallography, X-Ray , Peptidyl-Prolyl Isomerase F , Cyclophilins/genetics , Cyclophilins/metabolism , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/genetics , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Nuclear Magnetic Resonance, Biomolecular , Peptide Elongation Factor 2/chemistry , Peptide Elongation Factor 2/genetics , Protein Binding , Protein Folding , Protein Interaction Domains and Motifs , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Structure-Activity Relationship
20.
Mol Cell ; 74(4): 816-830.e7, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31027879

ABSTRACT

p53, the guardian of the genome, requires chaperoning by Hsp70 and Hsp90. However, how the two chaperone machineries affect p53 conformation and regulate its function remains elusive. We found that Hsp70, together with Hsp40, unfolds p53 in an ATP-dependent reaction. This unfolded state of p53 is susceptible to aggregation after release induced by the nucleotide exchange factor Bag-1. However, when Hsp90 and the adaptor protein Hop are present, p53 is transferred from Hsp70 to Hsp90, allowing restoration of the native state upon ATP hydrolysis. Our results suggest that the p53 conformation is constantly remodeled by the two major chaperone machineries. This connects p53 activity to stress, and the levels of free molecular chaperones are important factors regulating p53 activity. Together, our findings reveal an intricate interplay and cooperation of Hsp70 and Hsp90 in regulating the conformation of a client.


Subject(s)
HSP70 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/chemistry , Protein Conformation , Tumor Suppressor Protein p53/chemistry , Adenosine Triphosphate/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/genetics , Humans , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Protein Aggregates/genetics , Protein Binding/genetics , Protein Folding , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL