Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Biol Chem ; 299(2): 102856, 2023 02.
Article in English | MEDLINE | ID: mdl-36596358

ABSTRACT

Heat shock protein 90 (Hsp90) is known to mediate heme insertion and activation of heme-deficient neuronal nitric oxide (NO) synthase (apo-nNOS) in cells by a highly dynamic interaction that has been extremely difficult to study mechanistically with the use of subcellular systems. In that the heme content of many critical hemeproteins is regulated by Hsp90 and the heme chaperone GAPDH, the development of an in vitro system for the study of this chaperone-mediated heme regulation would be extremely useful. Here, we show that use of an antibody-immobilized apo-nNOS led not only to successful assembly of chaperone complexes but the ability to show a clear dependence on Hsp90 and GAPDH for heme-mediated activation of apo-nNOS. The kinetics of binding for Hsp70 and Hsp90, the ATP and K+ dependence, and the absolute requirement for Hsp70 in assembly of Hsp90•apo-nNOS heterocomplexes all point to a similar chaperone machinery to the well-established canonical machine regulating steroid hormone receptors. However, unlike steroid receptors, the use of a purified protein system containing Hsp90, Hsp70, Hsp40, Hop, and p23 is unable to activate apo-nNOS. Thus, heme insertion requires a unique Hsp90-chaperone complex. With this newly developed in vitro system, which recapitulates the cellular process requiring GAPDH as well as Hsp90, further mechanistic studies are now possible to better understand the components of the Hsp90-based chaperone system as well as how this heterocomplex works with GAPDH to regulate nNOS and possibly other hemeproteins.


Subject(s)
Glyceraldehyde-3-Phosphate Dehydrogenases , HSP70 Heat-Shock Proteins , HSP90 Heat-Shock Proteins , Heme , Hemeproteins , Molecular Chaperones , Nitric Oxide Synthase , Heme/chemistry , Hemeproteins/chemistry , Hemeproteins/metabolism , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Protein Binding , Nitric Oxide Synthase/chemistry , Nitric Oxide Synthase/metabolism , Enzymes, Immobilized , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Enzyme Activation
2.
Cell Mol Life Sci ; 77(6): 977-996, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31552448

ABSTRACT

The polyglutamine (polyQ) diseases are a group of nine fatal, adult-onset neurodegenerative disorders characterized by the misfolding and aggregation of mutant proteins containing toxic expansions of CAG/polyQ tracts. The heat shock protein 90 and 70 (Hsp90/Hsp70) chaperone machinery is a key component of cellular protein quality control, playing a role in the regulation of folding, aggregation, and degradation of polyQ proteins. The ability of Hsp70 to facilitate disaggregation and degradation of misfolded proteins makes it an attractive therapeutic target in polyQ diseases. Genetic studies have demonstrated that manipulation of Hsp70 and related co-chaperones can enhance the disaggregation and/or degradation of misfolded proteins in models of polyQ disease. Therefore, the development of small molecules that enhance Hsp70 activity is of great interest. However, it is still unclear if currently available Hsp70 modulators can selectively enhance disaggregation or degradation of misfolded proteins without perturbing other Hsp70 functions essential for cellular homeostasis. This review discusses the multifaceted role of Hsp70 in protein quality control and the opportunities and challenges Hsp70 poses as a potential therapeutic target in polyQ disease.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Peptides/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Humans , Huntington Disease/drug therapy , Huntington Disease/metabolism , Molecular Targeted Therapy , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Folding/drug effects , Proteostasis Deficiencies/drug therapy , Proteostasis Deficiencies/metabolism , Spinocerebellar Ataxias/drug therapy , Spinocerebellar Ataxias/metabolism
3.
Mol Pharmacol ; 94(3): 984-991, 2018 09.
Article in English | MEDLINE | ID: mdl-29941666

ABSTRACT

Several hundred proteins cycle into heterocomplexes with a dimer of the chaperone heat shock protein 90 (Hsp90), regulating their activity and turnover. There are two isoforms of Hsp90, Hsp90α and Hsp90ß, and their relative chaperone activities and composition in these client protein•Hsp90 heterocomplexes has not been determined. Here, we examined the activity of human Hsp90α and Hsp90ß in a purified five-protein chaperone machinery that assembles glucocorticoid receptor (GR)•Hsp90 heterocomplexes to generate high-affinity steroid-binding activity. We found that human Hsp90α and Hsp90ß have equivalent chaperone activities, and when mixed together in this assay, they formed only GR•Hsp90αα and GR•Hsp90ßß homodimers and no GR•Hsp90αß heterodimers. In contrast, GR•Hsp90 heterocomplexes formed in human embryonic kidney (HEK) cells also contain GR•Hsp90αß heterodimers. The formation of GR•Hsp90αß heterodimers in HEK cells probably reflects the longer time permitted for exchange to form Hsp90αß heterodimers in the cell versus in the cell-free assembly conditions. This purified GR-activating chaperone machinery can be used to determine how modifications of Hsp90 affect its chaperone activity. To that effect, we have tested whether the unique phosphorylation of Hsp90α at threonines 5 and 7 that occurs during DNA damage repair affects its chaperone activity. We showed that the phosphomimetic mutant Hsp90α T5/7D has the same intrinsic chaperone activity as wild-type human Hsp90α in activation of GR steroid-binding activity by the five-protein machinery, supporting the conclusion that T5/7 phosphorylation does not affect Hsp90α chaperone activity.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Multimerization/physiology , Receptors, Glucocorticoid/metabolism , Animals , HEK293 Cells , HSP70 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/chemistry , Humans , Mice , Molecular Chaperones/chemistry , Protein Binding/physiology , Receptors, Glucocorticoid/chemistry
4.
Annu Rev Pharmacol Toxicol ; 55: 353-71, 2015.
Article in English | MEDLINE | ID: mdl-25292434

ABSTRACT

Currently available therapies for adult onset neurodegenerative diseases provide symptomatic relief but do not modify disease progression. Here we explore a new neuroprotective approach based on drugs targeting chaperone-directed protein quality control. Critical target proteins that unfold and aggregate in these diseases, such as the polyglutamine androgen receptor in spinal and bulbar muscular atrophy, huntingtin in Huntington's disease, α-synuclein in Parkinson's disease, and tau in Alzheimer's disease, are client proteins of heat shock protein 90 (Hsp90), and their turnover is regulated by the protein quality control function of the Hsp90/Hsp70-based chaperone machinery. Hsp90 and Hsp70 have opposing effects on client protein stability in protein quality control; Hsp90 stabilizes the clients and inhibits their ubiquitination, whereas Hsp70 promotes ubiquitination dependent on CHIP (C terminus of Hsc70-interacting protein) and proteasomal degradation. We discuss how drugs that modulate proteostasis by inhibiting Hsp90 function or promoting Hsp70 function enhance the degradation of the critical aggregating proteins and ameliorate toxic symptoms in cell and animal disease models.


Subject(s)
HSP70 Heat-Shock Proteins/agonists , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Nervous System/drug effects , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Adult , Age of Onset , Animals , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Nervous System/metabolism , Nervous System/physiopathology , Neurodegenerative Diseases/diagnosis , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Proteasome Endopeptidase Complex/metabolism , Protein Denaturation , Protein Folding , Protein Stability , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
Nat Chem Biol ; 9(2): 112-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23222885

ABSTRACT

We sought new strategies to reduce amounts of the polyglutamine androgen receptor (polyQ AR) and achieve benefits in models of spinobulbar muscular atrophy, a protein aggregation neurodegenerative disorder. Proteostasis of the polyQ AR is controlled by the heat shock protein 90 (Hsp90)- and Hsp70-based chaperone machinery, but mechanisms regulating the protein's turnover are incompletely understood. We demonstrate that overexpression of Hsp70 interacting protein (Hip), a co-chaperone that enhances binding of Hsp70 to its substrates, promotes client protein ubiquitination and polyQ AR clearance. Furthermore, we identify a small molecule that acts similarly to Hip by allosterically promoting Hsp70 binding to unfolded substrates. Like Hip, this synthetic co-chaperone enhances client protein ubiquitination and polyQ AR degradation. Both genetic and pharmacologic approaches targeting Hsp70 alleviate toxicity in a Drosophila model of spinobulbar muscular atrophy. These findings highlight the therapeutic potential of allosteric regulators of Hsp70 and provide new insights into the role of the chaperone machinery in protein quality control.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Peptides/chemistry , Animals , Dose-Response Relationship, Drug , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacology , Drosophila , Female , HEK293 Cells , HeLa Cells , Humans , Inhibitory Concentration 50 , Models, Chemical , Molecular Chaperones/chemistry , Muscular Disorders, Atrophic/metabolism , Neurotoxins/chemistry , PC12 Cells , Protein Structure, Tertiary , Proteins/chemistry , Pyridines/pharmacology , Rats , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Thiazoles/pharmacology , Ubiquitination
6.
J Biol Chem ; 288(37): 26879-86, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23897823

ABSTRACT

An eight-amino acid segment is known to be responsible for the marked difference in the rates of degradation of the EGF receptor (ErbB1) and ErbB2 upon treatment of cells with the Hsp90 inhibitor geldanamycin. We have scrambled the first six amino acids of this segment of the EGF receptor (EGFR), which lies in close association with the ATP binding cleft and the dimerization face. Scrambling these six amino acids markedly reduces EGFR stability, EGF-stimulated receptor dimerization, and autophosphorylation activity. Two peptides were synthesized as follows: one containing the wild-type sequence of the eight-amino acid segment, which we call Disruptin; and one with the scrambled sequence. Disruptin inhibits Hsp90 binding to the EGFR and causes slow degradation of the EGFR in two EGFR-dependent cancer cell lines, whereas the scrambled peptide is inactive. This effect is specific for EGFR versus other Hsp90 client proteins. In the presence of EGF, Disruptin, but not the scrambled peptide, inhibits EGFR dimerization and causes rapid degradation of the EGFR. In contrast to the Hsp90 inhibitor geldanamycin, Disruptin inhibits cancer cell growth by a nonapoptotic mechanism. Disruptin provides proof of concept for the development of a new class of anti-tumor drugs that specifically cause EGFR degradation.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , HSP90 Heat-Shock Proteins/metabolism , Peptide Fragments/pharmacology , Peptides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , CHO Cells , Cell Line, Tumor , Cricetulus , Dimerization , Drug Design , ErbB Receptors/pharmacology , Humans , Lactams, Macrocyclic/pharmacology , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Protein Binding
7.
J Biol Chem ; 287(2): 1556-65, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22128174

ABSTRACT

Like other nitric-oxide synthase (NOS) enzymes, neuronal NOS (nNOS) turnover and activity are regulated by the Hsp90/Hsp70-based chaperone machinery, which regulates signaling proteins by modulating ligand binding clefts (Pratt, W. B., Morishima, Y., and Osawa, Y. (2008) J. Biol. Chem. 283, 22885-22889). We have previously shown that nNOS turnover is due to Hsp70/CHIP-dependent ubiquitination and proteasomal degradation. In this work, we use an intracellular cross-linking approach to study both chaperone binding and nNOS ubiquitination in intact HEK293 cells. Treatment of cells with N(G)-nitro-L-arginine, a slowly reversible competitive inhibitor that stabilizes nNOS, decreases both nNOS ubiquitination and binding of Hsp90, Hsp70, and CHIP. Treatment with the calcium ionophore A23187, which increases Ca(2+)-calmodulin binding to nNOS, increases nNOS ubiquitination and binding of Hsp90, Hsp70, and CHIP in a manner that is specific for changes in the heme/substrate binding cleft. Both Hsp90 and Hsp70 are bound to the expressed nNOS oxygenase domain, which contains the heme/substrate binding cleft, but not to the reductase domain, and binding is increased to an expressed fragment containing both the oxygenase domain and the calmodulin binding site. Overexpression of Hsp70 promotes nNOS ubiquitination and decreases nNOS protein, and overexpression of Hsp90 inhibits nNOS ubiquitination and increases nNOS protein, showing the opposing effects of the two chaperones as they participate in nNOS quality control in the cell. These observations support the notion that changes in the state of the heme/substrate binding cleft affect chaperone binding and thus nNOS ubiquitination.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Nitric Oxide Synthase Type I/metabolism , Proteolysis , Ubiquitination/physiology , Animals , Calcimycin/pharmacology , Calcium Ionophores/pharmacology , Catalytic Domain/physiology , HEK293 Cells , HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/genetics , Heme/genetics , Heme/metabolism , Humans , Nitric Oxide Synthase Type I/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Rats , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects
8.
Biochemistry ; 50(33): 7146-56, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21755988

ABSTRACT

We have reported that heme-dependent activation of apo-neuronal nitric oxide synthase (apo-nNOS) to the active holo-enzyme dimer is dependent upon factors present in reticulocyte lysate and other cytosols. Here, we find that both Hsp70 and thioredoxin are components of the activation system. The apo-nNOS activating activity of reticulocyte lysate is retained in a pool of fractions containing Hsp70 that elute from DE52 prior to Hsp90. All of the activating activity and 20-30% of the Hsp70 elute in the flow-through fraction upon subsequent ATP-agarose chromatography. Apo-nNOS activation by this flow-through fraction is inhibited by pifithrin-µ, a small molecule inhibitor of Hsp70, suggesting that a non-ATP-binding form of Hsp70 is involved in heme-dependent apo-nNOS activation. Previous work has shown that apo-nNOS can be activated by thiol-disulfide exchange, and we show substantial activation with a small molecule dithiol modeled on the active motifs of thioredoxin and protein disulfide isomerase. Further fractionation of the ATP-agarose flow-through on Sephacryl S-300 separates free thioredoxin from apo-nNOS activating activity, Hsp70, and a small amount of thioredoxin, all of which are eluted throughout the macromolecular peak. Incubation of apo-nNOS with the macromolecular fraction in combination either with the thioredoxin-containing fraction or with purified recombinant human thioredoxin restores full heme-dependent activating activity. This supports a model in which Hsp70 binding to apo-nNOS stabilizes an open state of the heme/substrate binding cleft to facilitate thioredoxin access to the active site cysteine that coordinates with heme iron, permitting heme binding and dimerization to the active enzyme.


Subject(s)
Cytosol/metabolism , Disulfides/metabolism , HSP70 Heat-Shock Proteins/metabolism , Heme/metabolism , Nitric Oxide Synthase/metabolism , Sulfhydryl Compounds/metabolism , Thioredoxins/metabolism , Animals , Apoenzymes , Blood Coagulation Factors/metabolism , Dimerization , Enzyme Activation , Humans , Immunoblotting , Nitric Oxide Synthase Type I , Protein Binding , RNA-Binding Proteins , Rabbits , Rats , Reticulocytes , Ribosomal Proteins , Sepharose/analogs & derivatives
9.
J Biol Chem ; 285(44): 33642-51, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20729196

ABSTRACT

It is established that suicide inactivation of neuronal nitric-oxide synthase (nNOS) by drugs and other xenobiotics leads to ubiquitination and proteasomal degradation of the enzyme. The exact mechanism is not known, although it is widely thought that the covalent alteration of the active site during inactivation triggers the degradation. A mechanism that involves recognition of the altered nNOS by Hsp70 and its cochaperone CHIP, an E3-ubiquitin ligase, has been proposed. To further address how alterations of the active site trigger ubiquitination of nNOS, we examined a C331A nNOS mutant, which was reported to have impaired ability to bind L-arginine and tetrahydrobiopterin. We show here that C331A nNOS is highly susceptible to ubiquitination by a purified system containing ubiquitinating enzymes and chaperones, by the endogenous ubiquitinating system in reticulocyte lysate fraction II, and by intact HEK293 cells. The involvement of the altered heme cleft in regulating ubiquitination is confirmed by the finding that the slowly reversible inhibitor of nNOS, N(G)-nitro-L-arginine, but not its inactive D-isomer, protects the C331A nNOS from ubiquitination in all these experimental systems. We also show that both Hsp70 and CHIP play a major role in the ubiquitination of C331A nNOS, although Hsp90 protects from ubiquitination. Thus, these studies further strengthen the link between the mobility of the substrate-binding cleft and chaperone-dependent ubiquitination of nNOS. These results support a general model of chaperone-mediated protein quality control and lead to a novel mechanism for substrate stabilization based on nNOS interaction with the chaperone machinery.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , HSP70 Heat-Shock Proteins/metabolism , Mutation , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin/chemistry , Catalytic Domain , Cell Line , Chromatin Immunoprecipitation , Heat-Shock Proteins/chemistry , Humans , Ligands , Molecular Chaperones/chemistry , Protein Structure, Tertiary , Subcellular Fractions , Xenobiotics/chemistry
10.
J Biol Chem ; 285(21): 15714-23, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20348093

ABSTRACT

The Hsp90/Hsp70-based chaperone machinery regulates the activity and degradation of many signaling proteins. Cycling with Hsp90 stabilizes client proteins, whereas Hsp70 interacts with chaperone-dependent E3 ubiquitin ligases to promote protein degradation. To probe these actions, small molecule inhibitors of Hsp70 would be extremely useful; however, few have been identified. Here we test the effects of methylene blue, a recently described inhibitor of Hsp70 ATPase activity, in three well established systems of increasing complexity. First, we demonstrate that methylene blue inhibits the ability of the purified Hsp90/Hsp70-based chaperone machinery to enable ligand binding by the glucocorticoid receptor and show that this effect is due to specific inhibition of Hsp70. Next, we establish that ubiquitination of neuronal nitric-oxide synthase by the native ubiquitinating system of reticulocyte lysate is dependent upon both Hsp70 and the E3 ubiquitin ligase CHIP and is blocked by methylene blue. Finally, we demonstrate that methylene blue impairs degradation of the polyglutamine expanded androgen receptor, an Hsp90 client mutated in spinal and bulbar muscular atrophy. In contrast, degradation of an amino-terminal fragment of the receptor, which lacks the ligand binding domain and, therefore, is not a client of the Hsp90/Hsp70-based chaperone machinery, is enhanced through homeostatic induction of autophagy that occurs when Hsp70-dependent proteasomal degradation is inhibited by methylene blue. Our data demonstrate the utility of methylene blue in defining Hsp70-dependent functions and reveal divergent effects on polyglutamine protein degradation depending on whether the substrate is an Hsp90 client.


Subject(s)
Enzyme Inhibitors/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Methylene Blue/pharmacology , Peptides/metabolism , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/drug effects , Animals , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Mice , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I/metabolism , Proteasome Endopeptidase Complex/metabolism , Rats
11.
Hum Mol Genet ; 17(24): 3942-52, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18784277

ABSTRACT

CHIP (carboxy terminus of Hsc70-interacting protein) an E3 ubiquitin ligase that binds to Hsp70 and Hsp90, promotes degradation of several Hsp90-regulated signaling proteins and disease-causing proteins containing expanded glutamine tracts. In polyglutamine disease models, CHIP has been considered a primary protection factor by promoting degradation of these misfolded proteins. Here, we show that two CHIP substrates, the glucocorticoid receptor (GR), a classic Hsp90-regulated signaling protein, and the expanded glutamine androgen receptor (AR112Q), are degraded at the same rate in CHIP(-/-) and CHIP(+/+) mouse embryonic fibroblasts after treatment with the Hsp90 inhibitor geldanamycin. CHIP(-/-) cytosol has the same ability as CHIP(+/+) cytosol to ubiquitinate purified neuronal nitric oxide synthase (nNOS), another established CHIP substrate. To determine whether other E3 ubiquitin ligases that bind to Hsp70 (Parkin) or Hsp90 (Mdm2) act on CHIP substrates, each E3 ligase was co-expressed with the GR, nNOS, AR112Q or Q78 ataxin-3. CHIP lowered the levels of all four proteins, Parkin acted on nNOS and Q78 ataxin-3 but not on the steroid receptors, and Mdm2 did not affect any of the co-expressed proteins. Moreover, both CHIP and Parkin co-localized to aggregates of the expanded glutamine AR formed in cell culture and in a knock-in mouse model of spinal and bulbar muscular atrophy. These observations establish that CHIP does not play an exclusive role in regulating the turnover of Hsp90 client signaling proteins or expanded glutamine tract proteins, and show that the Hsp70-dependent E3 ligase Parkin acts redundantly to CHIP on some substrates.


Subject(s)
Gene Deletion , Glutamine/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Cells, Cultured , Glutamine/metabolism , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Mice , Mice, Knockout , Signal Transduction/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
12.
Clin Cancer Res ; 26(19): 5246-5257, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32718999

ABSTRACT

PURPOSE: Treatment approaches using Hsp90 inhibitors at their maximum tolerated doses (MTDs) have not produced selective tumor toxicity. Inhibition of Hsp90 activity causes degradation of client proteins including those involved in recognizing and repairing DNA lesions. We hypothesized that if DNA repair proteins were degraded by concentrations of an Hsp90 inhibitor below those required to cause nonspecific cytotoxicity, significant tumor-selective radiosensitization might be achieved. EXPERIMENTAL DESIGN: Tandem mass tagged-mass spectrometry was performed to determine the effect of a subcytotoxic concentration of the Hsp90 inhibitor, AT13387 (onalespib), on global protein abundance. The effect of AT13387 on in vitro radiosensitization was assessed using a clonogenic assay. Pharmacokinetics profiling was performed in mice bearing xenografts. Finally, the effect of low-dose AT13387 on the radiosensitization of three tumor models was assessed. RESULTS: A subcytotoxic concentration of AT13387 reduced levels of DNA repair proteins, without affecting the majority of Hsp90 clients. The pharmacokinetics study using one-third of the MTD showed 40-fold higher levels of AT13387 in tumors compared with plasma. This low dose enhanced Hsp70 expression in peripheral blood mononuclear cells (PBMCs), which is a biomarker of Hsp90 inhibition. Low dose monotherapy was ineffective, but when combined with radiotherapy, produced significant tumor growth inhibition. CONCLUSIONS: This study shows that a significant therapeutic ratio can be achieved by a low dose of Hsp90 inhibitor in combination with radiotherapy. Hsp90 inhibition, even at a low dose, can be monitored by measuring Hsp70 expression in PBMCs in human studies.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/administration & dosage , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Animals , Benzamides/pharmacology , DNA Repair/drug effects , DNA Repair/radiation effects , Dose-Response Relationship, Drug , HSP90 Heat-Shock Proteins/genetics , Heterografts , Humans , Isoindoles/pharmacology , Leukocytes, Mononuclear/drug effects , Mice , Proteolysis/drug effects , Proteolysis/radiation effects , Radiation Tolerance/genetics , Radiation-Sensitizing Agents/adverse effects , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology
13.
Biochemistry ; 48(35): 8483-90, 2009 Sep 08.
Article in English | MEDLINE | ID: mdl-19642705

ABSTRACT

NO production by neuronal nitric oxide synthase (nNOS) requires calmodulin and is enhanced by the chaperone Hsp90, which cycles dynamically with the enzyme. The proteasomal degradation of nNOS is enhanced by suicide inactivation and by treatment with Hsp90 inhibitors, the latter suggesting that dynamic cycling with Hsp90 stabilizes nNOS. Here, we use a purified ubiquitinating system containing CHIP (carboxyl terminus of Hsp70-interacting protein) as the E3 ligase to show that Hsp90 inhibits CHIP-dependent nNOS ubiquitination. Like the established Hsp90 enhancement of NO synthesis, Hsp90 inhibition of nNOS ubiquitination is Ca2+/calmodulin-dependent, suggesting that the same interaction of Hsp90 with the enzyme is responsible for both enhancement of nNOS activity and inhibition of ubiquitination. It is established that CHIP binds to Hsp90 as well as to Hsp70, but we show here the two chaperones have opposing actions on nNOS ubiquitination, with Hsp70 stimulating and Hsp90 inhibiting. We have used two mechanism-based inactivators, guanabenz and NG-amino-L-arginine, to alter the heme/substrate binding cleft and promote nNOS ubiquitination that can be inhibited by Hsp90. We envision that, as nNOS undergoes toxic damage, the heme/substrate binding cleft opens exposing hydrophobic residues as the initial step in unfolding. As long as Hsp90 can form even transient complexes with the opening cleft, ubiquitination by Hsp70-dependent ubiquitin E3 ligases, like CHIP, is inhibited. When unfolding of the cleft progresses to a state that cannot cycle with Hsp90, Hsp70-dependent ubiquitination is unopposed. In this way, the Hsp70/Hsp90 machinery makes the quality control decision for stabilization versus degradation of nNOS.


Subject(s)
Calmodulin/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Nitric Oxide Synthase Type I/metabolism , Ubiquitin/metabolism , Ubiquitination/drug effects , Animals , Cell Line , Cyclization , Dimerization , Humans , Rabbits , Reticulocytes/enzymology , Reticulocytes/metabolism , Signal Transduction , Spodoptera
14.
Oncotarget ; 7(50): 82450-82457, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27738310

ABSTRACT

Heat shock protein 90 is a chaperone that plays an essential role in the stabilization of a large number of signal transduction molecules, many of which are associated with oncogenesis. An Hsp90 isoform (Hsp90α) has been shown to be selectively phosphorylated on two N-terminal threonine residues (threonine 5 and 7) and is involved in the DNA damage response and apoptosis. However, the kinase that phosphorylates Hsp90α after ionizing radiation (IR) and its role in post-radiation DNA repair remains unclear. Inasmuch as several proteins of the DNA damage response machinery are Hsp90 clients, the functional consequences of Hsp90α phosphorylation following IR have implications for the design of novel radiosensitizing agents that specifically target the Hsp90α isoform. Here we show that ATM phosphorylates Hsp90α at the T5/7 residues immediately after IR. The kinetics of Hsp90α T5/7 phosphorylation correlate with the kinetics of H2AX S139 phosphorylation (γH2AX). Although Hsp90α is located in both the cytoplasm and nucleus, only nuclear Hsp90α is phosphorylated by ATM after IR. The siRNA mediated knockdown of Hsp90α sensitizes head and neck squamous cell carcinoma cells, lung cancer cells and lung fibroblasts to IR. Furthermore, MEF cells that are Hsp90α null have reduced levels of γH2AX indicating that Hsp90α is important for the formation of γH2AX. Thus, this study provides evidence that Hsp90α is a component of the signal transduction events mediated by ATM following IR, and that Hsp90α loss decreases γH2AX levels. This work supports additional investigation into Hsp90α T5/7 phosphorylation with the goal of developing targeted radiosensitizing therapies.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Carcinoma, Squamous Cell/radiotherapy , Fibroblasts/radiation effects , HSP90 Heat-Shock Proteins/metabolism , Head and Neck Neoplasms/radiotherapy , Lung Neoplasms/radiotherapy , Animals , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Nucleus/pathology , Cell Nucleus/radiation effects , DNA Damage , Dose-Response Relationship, Radiation , Fibroblasts/enzymology , Fibroblasts/pathology , HSP90 Heat-Shock Proteins/genetics , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/pathology , Histones/metabolism , Humans , Kinetics , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Phosphorylation , RNA Interference , Squamous Cell Carcinoma of Head and Neck , Time Factors , Transfection
15.
J Neurosci ; 24(20): 4758-66, 2004 May 19.
Article in English | MEDLINE | ID: mdl-15152036

ABSTRACT

The delivery of neurotransmitter receptors into synapses is essential for synaptic function and plasticity. In particular, AMPA-type glutamate receptors (AMPA receptors) reach excitatory synapses according to two distinct routes: a regulated pathway, which operates transiently during synaptic plasticity, and a constitutive pathway, which maintains synaptic function under conditions of basal transmission. However, the specific mechanisms that distinguish these two trafficking pathways are essentially unknown. Here, we evaluate the role of the molecular chaperone hsp90 (heat shock protein 90) in excitatory synaptic transmission in the hippocampus. On one hand, we found that hsp90 is necessary for the efficient neurotransmitter release at the presynaptic terminal. In addition, we identified hsp90 as a critical component of the cellular machinery that delivers AMPA receptors into the postsynaptic membrane. Using the hsp90-specific inhibitors radicicol and geldanamycin, we show that hsp90 is required for the constitutive trafficking of AMPA receptors into synapses during their continuous cycling between synaptic and nonsynaptic sites. In contrast, hsp90 function is not required for either the surface delivery of AMPA receptors into the nonsynaptic plasma membrane or for the acute, regulated delivery of AMPA receptors into synapses during plasticity induction (long-term potentiation). The synaptic cycling of AMPA receptors was also blocked by an hsp90-binding tetratricopeptide repeat (TPR) domain, suggesting that the role of hsp90 in AMPA receptor trafficking is mediated by a TPR domain-containing protein. These results demonstrate new roles for hsp90 in synaptic function by controlling neurotransmitter release and, independently, by mediating the continuous cycling of synaptic AMPA receptors.


Subject(s)
HSP90 Heat-Shock Proteins/physiology , Neurotransmitter Agents/metabolism , Pyramidal Cells/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , Vesicular Transport Proteins , Adenosine Triphosphatases/physiology , Animals , Carrier Proteins/physiology , Cell Membrane/metabolism , Enzyme Inhibitors/pharmacology , Genetic Vectors/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Hippocampus/cytology , Hippocampus/physiology , In Vitro Techniques , Long-Term Potentiation/physiology , N-Ethylmaleimide-Sensitive Proteins , Patch-Clamp Techniques , Protein Structure, Tertiary/physiology , Protein Transport/drug effects , Protein Transport/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repetitive Sequences, Amino Acid/physiology , Signal Transduction/physiology , Sindbis Virus/genetics , Synaptic Transmission/physiology
16.
Essays Biochem ; 40: 41-58, 2004.
Article in English | MEDLINE | ID: mdl-15242338

ABSTRACT

Unliganded steroid receptors are assembled into heterocomplexes with heat-shock protein (hsp) 90 by a multiprotein chaperone machinery. In addition to binding the receptors at the chaperone site, hsp90 binds cofactors at other sites that are part of the assembly machinery, as well as immunophilins that connect the assembled receptor-hsp90 heterocomplexes to a protein trafficking pathway. The hsp90-/hsp70-based chaperone machinery interacts with the unliganded glucocorticoid receptor to open the steroid-binding cleft to access by a steroid, and the machinery interacts in very dynamic fashion with the liganded, transformed receptor to facilitate its translocation along microtubular highways to the nucleus. In the nucleus, the chaperone machinery interacts with the receptor in transcriptional regulatory complexes after hormone dissociation to release the receptor and terminate transcriptional activation. By forming heterocomplexes with hsp90, the chaperone machinery stabilizes the receptor to degradation by the ubiquitin-proteasome pathway of proteolysis.


Subject(s)
Molecular Chaperones/physiology , Receptors, Steroid/metabolism , Animals , HSP90 Heat-Shock Proteins/physiology , Humans , Protein Binding , Protein Transport , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Receptors, Steroid/chemistry
17.
Cell Signal ; 16(8): 857-72, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15157665

ABSTRACT

The ubiquitous protein chaperone hsp90 has been shown to regulate more than 100 proteins involved in cellular signalling. These proteins are called 'client proteins' for hsp90, and a multiprotein hsp90/hsp70-based chaperone machinery forms client protein.hsp90 heterocomplexes in the cytoplasm and the nucleus. In the case of signalling proteins that act as transcription factors, the client protein.hsp90 complexes also contain one of several TPR domain immunophilins or immunophilin homologs that bind to a TPR domain binding site on hsp90. Using several intracellular receptors and the tumor suppressor p53 as examples, we review evidence that dynamic assembly of heterocomplexes with hsp90 is required for rapid movement through the cytoplasm to the nucleus along microtubular tracks. The role of the immunophilin in this system is to connect the client protein.hsp90 complex to cytoplasmic dynein, the motor protein for retrograde movement toward the nucleus. Upon arrival at the nuclear pores, the receptor.hsp90.immunophilin complexes are transferred to the nuclear interior by importin-dependent facilitated diffusion. The unliganded receptors then distribute within the nucleus to diffuse patches from which they proceed in a ligand-dependent manner to discrete nuclear foci where chromatin binding occurs. We review evidence that dynamic assembly of heterocomplexes with hsp90 is required for movement to these foci and for the dynamic exchange of transcription factors between chromatin and the nucleoplasm.


Subject(s)
Cell Nucleus/metabolism , HSP90 Heat-Shock Proteins/metabolism , Immunophilins/metabolism , Molecular Chaperones/metabolism , Signal Transduction/physiology , Animals , Cell Cycle Proteins/metabolism , Humans , Tumor Suppressor Protein p53/metabolism
18.
Brain Res Mol Brain Res ; 123(1-2): 27-36, 2004 Apr 07.
Article in English | MEDLINE | ID: mdl-15046863

ABSTRACT

Here, we have used a chimera of green fluorescent protein (GFP) and the glucocorticoid receptor (GR) to study retrograde movement of a model soluble (i.e., non-vesicle-associated) protein in axons and dendrites of cultured NT2-N neurons. It is known that in non-neuronal cells, the GFP-GR moves from cytoplasm to the nucleus in a steroid-dependent manner by a rapid, hsp90-dependent mechanism. When rapid movement is inhibited by geldanamycin (GA), a specific inhibitor of the protein chaperone hsp90, the GFP-GR translocates slowly to the nucleus by diffusion. Here we show that GFP-GR expressed in hormone-free neurons is localized in both cytoplasm and neurites, and upon treatment with dexamethasone (DEX), it moves to the nucleus. In neurites, movement by diffusion is not possible, and we show that movement of the GFP-GR from neurites is blocked by geldanamycin, suggesting that the hsp90-dependent movement machinery is required for retrograde movement. In cells treated with both dexamethasone and geldanamycin, the GFP-GR becomes concentrated in fluorescent globules located periodically along the neurites. Carboxyl terminus of Hsc70-interacting protein (CHIP), the E3 ubiquitin ligase for the GR, also concentrates in the same loci in a steroid-dependent and geldanamycin-dependent manner. If geldanamycin is removed, the GFP-GR exits the globules and continues its retrograde movement. However, in the continued presence of geldanamycin, the GFP-GR in the globules undergoes proteasomal degradation, suggesting that the globules function as degradasomes. This is the first evidence for a linkage between receptor trafficking along neurites and receptor degradation by the proteasome.


Subject(s)
Cysteine Endopeptidases/metabolism , HSP90 Heat-Shock Proteins/metabolism , Multienzyme Complexes/metabolism , Neurites/metabolism , Receptors, Glucocorticoid/metabolism , Ubiquitin-Protein Ligases/metabolism , Axons/drug effects , Axons/metabolism , Benzoquinones , Cell Compartmentation/drug effects , Cell Compartmentation/physiology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoplasm/drug effects , Cytoplasm/metabolism , Dendrites/drug effects , Dendrites/metabolism , Dexamethasone/pharmacology , Humans , Lactams, Macrocyclic , Macromolecular Substances , Neurites/drug effects , Neurites/ultrastructure , Proteasome Endopeptidase Complex , Protein Transport/drug effects , Protein Transport/physiology , Quinones/pharmacology , Recombinant Fusion Proteins/metabolism
19.
Exp Biol Med (Maywood) ; 228(2): 111-33, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12563018

ABSTRACT

Nearly 100 proteins are known to be regulated by hsp90. Most of these substrates or "client proteins" are involved in signal transduction, and they are brought into complex with hsp90 by a multiprotein hsp90/hsp70-based chaperone machinery. In addition to binding substrate proteins at the chaperone site(s), hsp90 binds cofactors at other sites that are part of the heterocomplex assembly machinery as well as immunophilins that connect assembled substrate*hsp90 complexes to protein-trafficking systems. In the 5 years since we last reviewed this subject, much has been learned about hsp90 structure, nucleotide-binding, and cochaperone interactions; the most important concept is that ATP hydrolysis by an intrinsic ATPase activity results in a conformational change in hsp90 that is required to induce conformational change in a substrate protein. The conformational change induced in steroid receptors is an opening of the steroid-binding cleft so that it can be accessed by steroid. We have now developed a minimal system of five purified proteins-hsp90, hsp70, Hop, hsp40, and p23- that assembles stable receptor*hsp90 heterocomplexes. An hsp90*Hop*hsp70*hsp40 complex opens the cleft in an ATP-dependent process to produce a receptor*hsp90 heterocomplex with hsp90 in its ATP-bound conformation, and p23 then interacts with the hsp90 to stabilize the complex. Stepwise assembly experiments have shown that hsp70 and hsp40 first interact with the receptor in an ATP-dependent reaction to produce a receptor*hsp70*hsp40 complex that is "primed" to be activated to the steroid-binding state in a second ATP-dependent step with hsp90, Hop, and p23. Successful use of the five-protein system with other substrates indicates that it can assemble signal protein*hsp90 heterocomplexes whether the substrate is a receptor, a protein kinase, or a transcription factor. This purified system should facilitate understanding of how eukaryotic hsp70 and hsp90 work together as essential components of a process that alters the conformations of substrate proteins to states that respond in signal transduction.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Transport/physiology , Signal Transduction/physiology , Animals , Drosophila Proteins , Humans , Immunophilins/metabolism , Janus Kinases , Macromolecular Substances , Models, Biological , Multiprotein Complexes , Nucleotides/metabolism , Protein Binding , Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Structure-Activity Relationship , Transcription Factors/metabolism
20.
Exp Biol Med (Maywood) ; 239(11): 1405-13, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24990484

ABSTRACT

In an EBM Minireview published in 2010, we proposed that the heat shock protein (Hsp)90/Hsp70-based chaperone machinery played a major role in determining the selection of proteins that have undergone oxidative or other toxic damage for ubiquitination and proteasomal degradation. The proposal was based on a model in which the Hsp90 chaperone machinery regulates signaling by modulating ligand-binding clefts. The model provides a framework for thinking about the development of neuroprotective therapies for protein-folding diseases like Alzheimer's disease (AD), Parkinson's disease (PD), and the polyglutamine expansion disorders, such as Huntington's disease (HD) and spinal and bulbar muscular atrophy (SBMA). Major aberrant proteins that misfold and accumulate in these diseases are "client" proteins of the abundant and ubiquitous stress chaperone Hsp90. These Hsp90 client proteins include tau (AD), α-synuclein (PD), huntingtin (HD), and the expanded glutamine androgen receptor (polyQ AR) (SBMA). In this Minireview, we update our model in which Hsp90 acts on protein-folding clefts and show how it forms a rational basis for developing drugs that promote the targeted elimination of these aberrant proteins.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Neuroprotective Agents/metabolism , Proteostasis Deficiencies/therapy , Humans , Huntingtin Protein , Nerve Tissue Proteins/metabolism , Protein Folding , Proteolysis , Receptors, Neurotransmitter/metabolism , Ubiquitination , alpha-Synuclein/metabolism , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL