Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
ACS Chem Biol ; 18(8): 1698-1704, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37450938

ABSTRACT

One of the O-GlcNAc modifications is the protection of cells against a variety of stressors that result in cell death. Previous experiments have focused on the overall ability of O-GlcNAc to prevent protein aggregation under stress as well as its ability to affect stress-response signaling pathways. Less attention has been paid to the potential role for O-GlcNAc in the direct inhibition of a major cell-death pathway, apoptosis. Apoptosis involves the sequential activation of caspase proteases, including the transfer of cell-stress information from initiator caspase-9 to effector caspase-3. Cells have multiple mechanisms to slow the apoptotic cascade, including heat shock protein HSP27, which can directly inhibit the activation of caspase-3 by caspase-9. We have previously shown that O-GlcNAc modification increases the chaperone activity of HSP27 against amyloid aggregation, raising the question as to whether this modification may play important roles in other facets of HSP27 biology. Here, we use protein chemistry to generate different versions of O-GlcNAc modified HSP27 and demonstrate that the modification enhances this antiapoptotic function of the chaperone, at least in an in vitro context. These results provide additional molecular insight into how O-GlcNAc functions as a mediator of cellular stress with important implications for human diseases like cancer and neurodegeneration.


Subject(s)
HSP27 Heat-Shock Proteins , Heat-Shock Proteins , Humans , Caspase 3/metabolism , Caspase 9/metabolism , HSP27 Heat-Shock Proteins/chemistry , Apoptosis/physiology
2.
J Mol Biol ; 435(13): 168139, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37146746

ABSTRACT

Small heat shock proteins (sHSPs) are essential ATP-independent chaperones that protect the cellular proteome. These proteins assemble into polydisperse oligomeric structures, the composition of which dramatically affects their chaperone activity. The biomolecular consequences of variations in sHSP ratios, especially inside living cells, remain elusive. Here, we study the consequences of altering the relative expression levels of HspB2 and HspB3 in HEK293T cells. These chaperones are partners in a hetero-oligomeric complex, and genetic mutations that abolish their mutual interaction are associated with myopathic disorders. HspB2 displays three distinct phenotypes when co-expressed with HspB3 at varying ratios. Expression of HspB2 alone leads to formation of liquid nuclear condensates, while shifting the stoichiometry towards HspB3 resulted in the formation of large solid-like aggregates. Only cells co-expressing HspB2 with a limited amount of HspB3 formed fully soluble complexes that were distributed homogeneously throughout the nucleus. Strikingly, both condensates and aggregates were reversible, as shifting the HspB2:HspB3 balance in situ resulted in dissolution of these structures. To uncover the molecular composition of HspB2 condensates and aggregates, we used APEX-mediated proximity labelling. Most proteins interact transiently with the condensates and were neither enriched nor depleted in these cells. In contrast, we found that HspB2:HspB3 aggregates sequestered several disordered proteins and autophagy factors, suggesting that the cell is actively attempting to clear these aggregates. This study presents a striking example of how changes in the relative expression levels of interacting proteins affects their phase behavior. Our approach could be applied to study the role of protein stoichiometry and the influence of client binding on phase behavior in other biomolecular condensates and aggregates.


Subject(s)
Heat-Shock Proteins, Small , Heat-Shock Proteins , Humans , Heat-Shock Proteins/metabolism , Heat-Shock Proteins, Small/genetics , HEK293 Cells , HSP27 Heat-Shock Proteins/chemistry , Cell Nucleus/metabolism , Protein Aggregates
3.
J Mol Biol ; 434(16): 167711, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35777462

ABSTRACT

Small heat-shock proteins (sHSPs) are ubiquitously expressed molecular chaperones present in all kingdoms of life that inhibit protein misfolding and aggregation. Despite their importance in proteostasis, the structure-function relationships of sHSPs remain elusive. Human sHSPs are characterised by a central, highly conserved α-crystallin domain (ACD) and variable-length N- and C-terminal regions. The ACD forms antiparallel homodimers via an extended ß-strand, creating a shared ß-sheet at the dimer interface. The N- and C-terminal regions mediate formation of higher order oligomers that are thought to act as storage forms for chaperone-active dimers. We investigated the interactions of the ACD of two human sHSPs, αB-crystallin (αB-C) and Hsp27, with apolipoprotein C-II amyloid fibrils using analytical ultracentrifugation and nuclear magnetic resonance spectroscopy. The ACD was found to interact transiently with amyloid fibrils to inhibit fibril elongation and naturally occurring fibril end-to-end joining. This interaction was sensitive to the concentration of fibril ends indicating a 'fibril-capping' interaction. Furthermore, resonances arising from the ACD monomer were attenuated to a greater extent than those of the ACD dimer in the presence of fibrils, suggesting that the monomer may bind fibrils. This hypothesis was supported by mutagenesis studies in which disulfide cross-linked ACD dimers formed by both αB-C and Hsp27 were less effective at inhibiting amyloid fibril elongation and fibril end-to-end joining than ACD constructs lacking disulfide cross-linking. Our results indicate that sHSP monomers inhibit amyloid fibril elongation, highlighting the importance of the dynamic oligomeric nature of sHSPs for client binding.


Subject(s)
Amyloid , HSP27 Heat-Shock Proteins , alpha-Crystallin B Chain , Amyloid/chemistry , Disulfides/chemistry , HSP27 Heat-Shock Proteins/chemistry , Humans , Protein Binding , Protein Domains , Protein Multimerization , alpha-Crystallin B Chain/chemistry
4.
Int J Mol Sci ; 22(15)2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34360542

ABSTRACT

In this study, a reliable and simple method of untagged recombinant human HspB7 preparation was developed. Recombinant HspB7 is presented in two oligomeric forms with an apparent molecular weight of 36 kDa (probably dimers) and oligomers with an apparent molecular weight of more than 600 kDa. By using hydrophobic and size-exclusion chromatography, we succeeded in preparation of HspB7 dimers. Mild oxidation promoted the formation of large oligomers, whereas the modification of Cys 126 by iodoacetamide prevented it. The deletion of the first 13 residues or deletion of the polySer motif (residues 17-29) also prevented the formation of large oligomers of HspB7. Cys-mutants of HspB6 and HspB8 containing a single-Cys residue in the central part of the ß7 strand in a position homologous to that of Cys137 in HspB1 can be crosslinked to the wild-type HspB7 through a disulfide bond. Immobilized on monoclonal antibodies, the wild-type HspB6 interacted with the wild-type HspB7. We suppose that formation of heterodimers of HspB7 with HspB6 and HspB8 may be important for the functional activity of these small heat shock proteins.


Subject(s)
HSP27 Heat-Shock Proteins/chemistry , Protein Multimerization , Recombinant Proteins/chemistry , Humans , Protein Domains , Protein Structure, Quaternary
5.
Int J Mol Sci ; 22(6)2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33809767

ABSTRACT

Small heat shock proteins (HSPs), such as HSP27, are ubiquitously expressed molecular chaperones and are essential for cellular homeostasis. The major functions of HSP27 include chaperoning misfolded or unfolded polypeptides and protecting cells from toxic stress. Dysregulation of stress proteins is associated with many human diseases including neurodegenerative diseases, such as Parkinson's disease (PD). PD is characterized by the presence of aggregates of α-synuclein in the central and peripheral nervous system, which induces the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and in the autonomic nervous system. Autonomic dysfunction is an important non-motor phenotype of PD, which includes cardiovascular dysregulation, among others. Nowadays, the therapies for PD focus on dopamine (DA) replacement. However, certain non-motor symptoms with a great impact on quality of life do not respond to dopaminergic drugs; therefore, the development and testing of new treatments for non-motor symptoms of PD remain a priority. Since small HSP27 was shown to prevent α-synuclein aggregation and cytotoxicity, this protein might constitute a suitable target to prevent or delay the motor and non-motor symptoms of PD. In the first part of our review, we focus on the cardiovascular dysregulation observed in PD patients. In the second part, we present data on the possible role of HSP27 in preventing the accumulation of amyloid fibrils and aggregated forms of α-synuclein. We also include our own studies, highlighting the possible protective cardiac effects induced by L-DOPA treatment through the enhancement of HSP27 levels and activity.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , Parkinson Disease/drug therapy , Protein Aggregates , Animals , HSP27 Heat-Shock Proteins/chemistry , Humans , Parkinson Disease/physiopathology , Protein Binding , alpha-Synuclein/metabolism
6.
Nat Struct Mol Biol ; 27(4): 363-372, 2020 04.
Article in English | MEDLINE | ID: mdl-32231288

ABSTRACT

Protein phase separation drives the assembly of membraneless organelles, but little is known about how these membraneless organelles are maintained in a metastable liquid- or gel-like phase rather than proceeding to solid aggregation. Here, we find that human small heat-shock protein 27 (Hsp27), a canonical chaperone that localizes to stress granules (SGs), prevents FUS from undergoing liquid-liquid phase separation (LLPS) via weak interactions with the FUS low complexity (LC) domain. Remarkably, stress-induced phosphorylation of Hsp27 alters its activity, leading Hsp27 to partition with FUS LC to preserve the liquid phase against amyloid fibril formation. NMR spectroscopy demonstrates that Hsp27 uses distinct structural mechanisms for both functions. Our work reveals a fine-tuned regulation of Hsp27 for chaperoning FUS into either a polydispersed state or a LLPS state and suggests an essential role for Hsp27 in stabilizing the dynamic phase of stress granules.


Subject(s)
HSP27 Heat-Shock Proteins/chemistry , Molecular Chaperones/chemistry , RNA-Binding Protein FUS/chemistry , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/isolation & purification , Humans , Liquid-Liquid Extraction , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Chaperones/genetics , Molecular Chaperones/isolation & purification , Phosphorylation , Protein Binding/genetics , Protein Domains/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/isolation & purification , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Stress, Physiological/genetics
7.
Biochemistry (Mosc) ; 84(11): 1256-1267, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31760916

ABSTRACT

The review discusses the role of small heat shock proteins (sHsps) in human neurodegenerative disorders, such as Charcot-Marie-Tooth disease (CMT), Parkinson's and Alzheimer's diseases, and different forms of tauopathies. The effects of CMT-associated mutations in two small heat shock proteins (HspB1 and HspB8) on the protein stability, oligomeric structure, and chaperone-like activity are described. Mutations in HspB1 shift the equilibrium between different protein oligomeric forms, leading to the alterations in its chaperone-like activity and interaction with protein partners, which can induce damage of the cytoskeleton and neuronal death. Mutations in HspB8 affect its interaction with the adapter protein Bag3, as well as the process of autophagy, also resulting in neuronal death. The impact of sHsps on different forms of amyloidosis is discussed. Experimental studies have shown that sHsps interact with monomers or small oligomers of amyloidogenic proteins, stabilize their structure, prevent their aggregation, and/or promote their specific proteolytic degradation. This effect might be due to the interaction between the ß-strands of sHsps and ß-strands of target proteins, which prevents aggregation of the latter. In cooperation with the other heat shock proteins, sHsps can promote disassembly of oligomers formed by amyloidogenic proteins. Despite significant achievements, further investigations are required for understanding the role of sHsps in protection against various neurodegenerative diseases.


Subject(s)
Heat-Shock Proteins, Small/metabolism , Neurodegenerative Diseases/pathology , Amyloidosis/metabolism , Amyloidosis/pathology , HSP27 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins, Small/chemistry , Heat-Shock Proteins, Small/genetics , Humans , Molecular Chaperones , Neurodegenerative Diseases/metabolism , Protein Conformation, beta-Strand , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Stability
8.
FEBS Open Bio ; 9(10): 1826-1834, 2019 10.
Article in English | MEDLINE | ID: mdl-31441240

ABSTRACT

Small heat shock proteins (sHsps) endow cells with stress tolerance. Of the various sHsps in mammals, HspB1, also known as Hsp27, is the most ubiquitous. To examine the structure and function of HspB1, we expressed, purified, and characterized HspB1 from Chinese hamster (Cricetulus griseus) ovary cells (CgHspB1). CgHspB1 forms a large oligomeric structure. We observed a monodisperse 16-mer with an elongated sphere, but this is affected by changes in various conditions, including temperature. Under dilute conditions, CgHspB1 dissociates into small oligomers at elevated temperatures. The dissociated conformers interacted with the gel filtration column through hydrophobic interactions. In contrast, dissociation of the oligomer was not observed by small-angle X-ray scattering at 55 °C. The result partially coincides with the results of size exclusion chromatography, showing that dissociation did not occur at high protein concentrations. However, a significant structural change in the oligomeric conformations appears to occur between room and higher temperatures. Reflecting their status as homeotherms, mammalian sHsps are regulated by phosphorylation. A phosphorylation mimic mutant of CgHspB1 with the replacement of Ser15 to Asp exhibited relatively lower oligomer stability and greater protective ability against thermal aggregation than the wild-type protein. The result clearly shows a correlation between oligomer dissociation and chaperone activity.


Subject(s)
HSP27 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/metabolism , Amino Acid Sequence , Animals , CHO Cells , Chromatography, Gel , Cloning, Molecular , Cricetulus , HSP27 Heat-Shock Proteins/isolation & purification , Phosphorylation , Protein Aggregates , Sequence Alignment
9.
J Biol Chem ; 294(25): 9985-9994, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31097540

ABSTRACT

Heat shock protein family B (small) member 7 (HSPB7) is a unique, relatively unexplored member within the family of human small heat shock proteins (HSPBs). Unlike most HSPB family members, HSPB7 does not oligomerize and so far has not been shown to associate with any other member of the HSPB family. Intriguingly, it was found to be the most potent member within the HSPB family to prevent aggregation of proteins with expanded polyglutamine (polyQ) stretches. How HSPB7 suppresses polyQ aggregation has remained elusive so far. Here, using several experimental strategies, including in vitro aggregation assay, immunoblotting and fluorescence approaches, we show that the polyQ aggregation-inhibiting activity of HSPB7 is fully dependent on its flexible N-terminal domain (NTD). We observed that the NTD of HSPB7 is both required for association with and inhibition of polyQ aggregation. Remarkably, replacing the NTD of HSPB1, which itself cannot suppress polyQ aggregation, with the NTD of HSPB7 resulted in a hybrid protein that gained anti-polyQ aggregation activity. The hybrid NTDHSPB7-HSPB1 protein displayed a reduction in oligomer size and, unlike WT HSPB1, associated with polyQ. However, experiments with phospho-mimicking HSPB1 mutants revealed that de-oligomerization of HSPB1 alone does not suffice to gain polyQ aggregation-inhibiting activity. Together, our results reveal that the NTD of HSPB7 is both necessary and sufficient to bind to and suppress the aggregation of polyQ-containing proteins.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Peptides/chemistry , Protein Aggregates , HSP27 Heat-Shock Proteins/chemistry , Humans , Peptides/metabolism , Protein Binding , Proteolysis
10.
Biochim Biophys Acta Gen Subj ; 1863(10): 1536-1546, 2019 10.
Article in English | MEDLINE | ID: mdl-31136785

ABSTRACT

Heat Shock Protein 27 (HSP27) is a small molecular chaperone that reduces the development of atherosclerosis by lowering plasma cholesterol levels as well as inflammation. Human studies show an inverse correlation between atherosclerotic burden and HSP27 expression, and are supported by murine models in which augmenting HSP27 levels curbs experimental atherogenesis. Natural HSP27 auto-antibodies (AAb) are found in human plasma, however their role in modulating the athero-protective effects of HSP27 is unknown. The purpose of this study is to characterize the biophysical interaction between human recombinant HSP27 and AAb. A validated polyclonal anti-HSP27 IgG antibody (PAb) was used to mimic natural AAb. Homology modeling and secondary structure prediction tools facilitated the design of HSP27 truncation and phosphorylation mutants. Secondary structural changes were identified using Circular Dichroism (CD) and Dynamic Light Scattering (DLS). Similar to prior structural investigations of HSP27, there was a predominance of α-helical content in the N-terminal truncation and dephosphorylation ("AA") mutants. The α-crystallin domain (ACD) predominantly consists of ß-strands, with the addition of the N-terminal increasing helical content and the C-terminal maintaining ß structure. With increasing ratios of PAb to HSP27 ß structure abundance and particle size increased, with a similar trend observed with the N-terminus, C-terminus and ACD peptides but an opposite trend with the phosphorylation peptides. Taken together, these studies provide insights into the interaction of HSP27 and its AAb that ultimately may aid in optimizing the design of HSP27 peptidomimetics with anti-atherogenic potential.


Subject(s)
Antibodies/immunology , HSP27 Heat-Shock Proteins/immunology , HSP27 Heat-Shock Proteins/metabolism , Animals , Biophysical Phenomena , Circular Dichroism , HSP27 Heat-Shock Proteins/chemistry , Humans , Mice , Phosphorylation , Protein Structure, Secondary
11.
Nat Commun ; 10(1): 1068, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842409

ABSTRACT

The small heat-shock protein HSP27 is a redox-sensitive molecular chaperone that is expressed throughout the human body. Here, we describe redox-induced changes to the structure, dynamics, and function of HSP27 and its conserved α-crystallin domain (ACD). While HSP27 assembles into oligomers, we show that the monomers formed upon reduction are highly active chaperones in vitro, but are susceptible to self-aggregation. By using relaxation dispersion and high-pressure nuclear magnetic resonance (NMR) spectroscopy, we observe that the pair of ß-strands that mediate dimerisation partially unfold in the monomer. We note that numerous HSP27 mutations associated with inherited neuropathies cluster to this dynamic region. High levels of sequence conservation in ACDs from mammalian sHSPs suggest that the exposed, disordered interface present in free monomers or oligomeric subunits may be a general, functional feature of sHSPs.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , Peripheral Nervous System Diseases/genetics , Protein Aggregation, Pathological/genetics , Protein Multimerization/genetics , Protein Unfolding , HSP27 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/genetics , Heat-Shock Proteins , Molecular Chaperones , Mutation , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Protein Conformation, beta-Strand/genetics , Protein Structure, Quaternary/physiology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
FASEB J ; 33(2): 1617-1630, 2019 02.
Article in English | MEDLINE | ID: mdl-30188755

ABSTRACT

Previously, we reported that elevated serum levels of heat shock protein 27 (HSP27) are predictive of a lower risk of having a heart attack, stroke, or death from cardiovascular disease. Moreover, augmenting HSP27 (or the murine ortholog, HSP25) attenuated experimental atherogenesis, reduced inflammation, and lowered cholesterol levels. Recently, we noted that HSP27 activates NF-κB via TLR-4, resulting in attenuation of plaque inflammation; however, the precise anti-atherosclerosis mechanisms mediated by extracellular HSP27 are incompletely understood. Our purpose in this study was to investigate the existence of HSP27 in extracellular vesicles (EVs) and whether HSP27 elicited atheroprotective effects on target cells. Here, we provide evidence that HSP27 localizes to EVs derived from THP-1 cells using transmission electron microscopy (TEM) and immunogold labeling, Western blotting, ELISA, and fluorescence-activated cell sorting. TEM imaging indicated that HSP27 is found at the exosomal membrane. Multiple reactor monitor-mass spectrometric analysis of large vesicles, which included microparticles and exosomes, isolated from human plasma, also led to detection of HSP27 using the unique signature peptide, R.LFDQAFGLPR.L. Studies using THP-1 and human embryonic kidney cells show that HSP27-laden exosomes significantly stimulated NF-κB activation ( P < 0.001) and release of IL-10 ( P < 0.0001), suggesting that HSP27 may be important exosomal cargo with beneficial anti-inflammatory effects.-Shi, C., Ulke-Lemée, A., Deng, J., Batulan, Z., O'Brien, E. R. Characterization of heat shock protein 27 in extracellular vesicles: a potential anti-inflammatory therapy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Exosomes/metabolism , HSP27 Heat-Shock Proteins/metabolism , Amino Acid Sequence , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , HSP27 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/genetics , Heat-Shock Proteins , Humans , Mass Spectrometry , Microscopy, Electron, Transmission , Molecular Chaperones , NF-kappa B/metabolism , THP-1 Cells
13.
Structure ; 26(8): 1116-1126.e4, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29983375

ABSTRACT

Small heat-shock proteins (sHSPs) are molecular chaperones that bind partially and globally unfolded states of their client proteins. Previously, we discovered that the archaeal Hsp16.5, which forms ordered and symmetric 24-subunit oligomers, can be engineered to transition to an ordered and symmetric 48-subunit oligomer by insertion of a peptide from human HspB1 (Hsp27). Here, we uncovered the existence of an array of oligomeric states (30-38 subunits) that can be populated as a consequence of altering the sequence and length of the inserted peptide. Polydisperse Hsp16.5 oligomers displayed higher affinity to a model client protein consistent with a general mechanism for recognition and binding that involves increased access of the hydrophobic N-terminal region. Our findings, which integrate structural and functional analyses from evolutionarily distant sHSPs, support a model wherein the modular architecture of these proteins encodes motifs of oligomer polydispersity, dissociation, and expansion to achieve functional diversity and regulation.


Subject(s)
Archaeal Proteins/chemistry , HSP27 Heat-Shock Proteins/chemistry , Heat-Shock Proteins, Small/chemistry , Peptides/chemistry , Protein Engineering/methods , Amino Acid Motifs , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Binding Sites , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Heat-Shock Proteins, Small/genetics , Heat-Shock Proteins, Small/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Kinetics , Models, Molecular , Molecular Chaperones , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Thermodynamics
14.
Int J Mol Sci ; 19(7)2018 Jul 20.
Article in English | MEDLINE | ID: mdl-30036999

ABSTRACT

Although the N-terminal domain of vertebrate small heat shock proteins (sHsp) is poorly conserved, it contains a core motif preserved in many members of the sHsp family. The role of this RLFDQxFG motif remains elusive. We analyzed the specific role of the first arginine residue of this conserved octet sequence in five human sHsps (HspB1, HspB4, HspB5, HspB6, and HspB8). Substitution of this arginine with an alanine induced changes in thermal stability and/or intrinsic fluorescence of the related HspB1 and HspB8, but yielded only modest changes in the same biophysical properties of HspB4, HspB5, and HspB6 which together belong to another clade of vertebrate sHsps. Removal of the positively charged Arg side chain resulted in destabilization of the large oligomers of HspB1 and formation of smaller size oligomers of HspB5. The mutation induced only minor changes in the structure of HspB4 and HspB6. In contrast, the mutation in HspB8 was accompanied by shifting the equilibrium from dimers towards the formation of larger oligomers. We conclude that the RLFDQxFG motif plays distinct roles in the structure of several sHsp orthologs. This role correlates with the evolutionary relationship of the respective sHsps, but ultimately, it reflects the sequence context of this motif.


Subject(s)
Amino Acid Motifs/physiology , Arginine/chemistry , Crystallins/chemistry , HSP20 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/chemistry , Heat-Shock Proteins, Small/chemistry , Heat-Shock Proteins, Small/metabolism , Heat-Shock Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , alpha-Crystallin B Chain/chemistry , Amino Acid Motifs/genetics , Amino Acid Sequence , Arginine/genetics , Chromatography, Gel , Crystallins/genetics , Crystallins/metabolism , HSP20 Heat-Shock Proteins/genetics , HSP20 Heat-Shock Proteins/metabolism , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Proteins, Small/genetics , Humans , Molecular Chaperones , Molecular Sequence Data , Point Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , alpha-Crystallin B Chain/genetics , alpha-Crystallin B Chain/metabolism
15.
J Mol Biol ; 430(18 Pt B): 3297-3310, 2018 09 14.
Article in English | MEDLINE | ID: mdl-29969581

ABSTRACT

Heterogeneity in small heat shock proteins (sHsps) spans multiple spatiotemporal regimes-from fast fluctuations of part of the protein, to conformational variability of tertiary structure, plasticity of the interfaces, and polydispersity of the inter-converting, and co-assembling oligomers. This heterogeneity and dynamic nature of sHsps has significantly hindered their structural characterization. Atomic coordinates are particularly lacking for vertebrate sHsps, where most available structures are of extensively truncated homomers. sHsps play important roles in maintaining protein levels in the cell and therefore in organismal health and disease. HspB2 and HspB3 are vertebrate sHsps that are found co-assembled in neuromuscular cells, and variants thereof are associated with disease. Here, we present the structure of human HspB2/B3, which crystallized as a hetero-tetramer in a 3:1 ratio. In the HspB2/B3 tetramer, the four α-crystallin domains (ACDs) assemble into a flattened tetrahedron which is pierced by two non-intersecting approximate dyads. Assembly is mediated by flexible "nuts and bolts" involving IXI/V motifs from terminal regions filling ACD pockets. Parts of the N-terminal region bind in an unfolded conformation into the anti-parallel shared ACD dimer grooves. Tracts of the terminal regions are not resolved, most likely due to their disorder in the crystal lattice. This first structure of a full-length human sHsp heteromer reveals the heterogeneous interactions of the terminal regions and suggests a plasticity that is important for the cytoprotective functions of sHsps.


Subject(s)
HSP27 Heat-Shock Proteins/chemistry , Heat-Shock Proteins/chemistry , Models, Molecular , Protein Conformation , Protein Multimerization , Amino Acid Sequence , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Magnetic Resonance Spectroscopy , Protein Binding , Protein Interaction Domains and Motifs
16.
Biochim Biophys Acta Proteins Proteom ; 1866(5-6): 692-701, 2018.
Article in English | MEDLINE | ID: mdl-29635040

ABSTRACT

Small heat shock protein 27 (HSP27) is an essential element of the proteostasis network in human cells. The HSP27 monomer coexists with the dimer, which can bind unfolded client proteins. Here, we evaluated the in-cell dimer-monomer equilibrium and its relevance to the binding of client proteins in a normal human vascular endothelial cell line. When cells were treated with a membrane-permeable crosslinker, the protein existed primarily as a free monomer (27 kDa) with a markedly smaller percentage of dimer (54 kDa), hetero-conjugates, and minor smear-like bands. When the protein was crosslinked in a cell-free lysate, two of the hetero-conjugates that were crosslinked in live cells were also detected, but the dimer and other complexes were absent. However, when cells were pretreated with fatty acid (FA) and/or heat (42.5 °C), dissociation of the dimer was selectively prevented and two types of covalently linked dimers were increased. These changes occurred most prominently in cells treated with docosahexaenoic acid (DHA) and heat, which appeared to intensify the heat resistance of the cell. Both the formation of covalently linked dimers and heat resistance were prevented by N-acetylcysteine. By contrast, nearly all of the free monomers in the lysate converted to disulfide bond-linked dimers by a simple, long incubation at 4 °C. These results strongly suggest that the monomer-dimer equilibrium of HSP27 was inversed between the in-cell and cell-free systems. Temperature- and amphiphile-regulated dimerization was restricted probably due to the low hydration of the in-cell crowding environment.


Subject(s)
Docosahexaenoic Acids/pharmacology , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Response/drug effects , Hot Temperature , Human Umbilical Vein Endothelial Cells/drug effects , Acetylcysteine/pharmacology , Arachidonic Acid/pharmacology , Cells, Cultured , Eicosapentaenoic Acid/pharmacology , HSP27 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/genetics , Heat-Shock Proteins , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Molecular Chaperones , Molecular Weight , Protein Conformation , Protein Multimerization , Structure-Activity Relationship , Time Factors
17.
ACS Chem Biol ; 13(7): 1824-1831, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29397684

ABSTRACT

Small heat shock protein (sHSP)-B7 (HSPB7) is a muscle-specific member of the non-ATP-dependent sHSPs. The precise role of HSPB7 is enigmatic. Here, we disclose that zebrafish Hspb7 is a kinetically privileged sensor that is able to react rapidly with native reactive electrophilic species (RES), when only substoichiometric amounts of RES are available in proximity to Hspb7 expressed in living cells. Among the two Hspb7-cysteines, this RES sensing is fulfilled by a single cysteine (C117). Purification and characterizations in vitro reveal that the rate for RES adduction is among the most efficient reported for protein-cysteines with native carbonyl-based RES. Covalent-ligand binding is accompanied by structural changes (increase in ß-sheet-content), based on circular dichroism analysis. Among the two cysteines, only C117 is conserved across vertebrates; we show that the human ortholog is also capable of RES sensing in cells. Furthermore, a cancer-relevant missense mutation reduces this RES-sensing property. This evolutionarily conserved cysteine-biosensor may play a redox-regulatory role in cardioprotection.


Subject(s)
Aldehydes/metabolism , HSP27 Heat-Shock Proteins/metabolism , Aldehydes/chemistry , Alkylation , Animals , Cell Line, Tumor , Cysteine/chemistry , HEK293 Cells , HSP27 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/genetics , Humans , Kinetics , Mutagenesis , Protein Structure, Secondary , Sequence Homology, Amino Acid , Zebrafish
18.
J Biol Chem ; 293(8): 2687-2700, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29298892

ABSTRACT

The microtubule-associated protein tau forms insoluble, amyloid-type aggregates in various dementias, most notably Alzheimer's disease. Cellular chaperone proteins play important roles in maintaining protein solubility and preventing aggregation in the crowded cellular environment. Although tau is known to interact with numerous chaperones, it remains unclear how these chaperones function mechanistically to prevent tau aggregation and how chaperones from different classes compare in terms of mechanism. Here, we focused on the small heat shock protein HspB1 (also known as Hsp27) and the constitutive chaperone Hsc70 (also known as HspA8) and report how each chaperone interacts with tau to prevent its fibril formation. Using fluorescence and NMR spectroscopy, we show that the two chaperones inhibit tau fibril formation by distinct mechanisms. HspB1 delayed tau fibril formation by weakly interacting with early species in the aggregation process, whereas Hsc70 was highly efficient at preventing tau fibril elongation, possibly by capping the ends of tau fibrils. Both chaperones recognized aggregation-prone motifs within the microtubule-binding repeat region of tau. However, HspB1 binding remained transient in both aggregation-promoting and non-aggregating conditions, whereas Hsc70 binding was significantly tighter under aggregation-promoting conditions. These differences highlight the fact that chaperones from different families play distinct but complementary roles in the prevention of pathological protein aggregation.


Subject(s)
Amyloid/metabolism , Down-Regulation , HSC70 Heat-Shock Proteins/metabolism , HSP27 Heat-Shock Proteins/metabolism , Models, Molecular , Protein Aggregation, Pathological/metabolism , tau Proteins/metabolism , Amino Acid Motifs , Amino Acid Substitution , Amyloid/chemistry , Amyloid/drug effects , Amyloid/ultrastructure , Anticoagulants/pharmacology , Cryoelectron Microscopy , Dimerization , Down-Regulation/drug effects , HSC70 Heat-Shock Proteins/chemistry , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/ultrastructure , HSP27 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/ultrastructure , Heat-Shock Proteins , Heparin/pharmacology , Humans , Kinetics , Molecular Chaperones , Mutation , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/prevention & control , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Solubility , tau Proteins/chemistry , tau Proteins/genetics , tau Proteins/ultrastructure
19.
Sci Rep ; 8(1): 688, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29330367

ABSTRACT

Congenital mutations in human small heat shock protein HSPB1 (HSP27) have been linked to Charcot-Marie-Tooth disease, a commonly occurring peripheral neuropathy. Understanding the molecular mechanism of such mutations is indispensable towards developing future therapies for this currently incurable disorder. Here we describe the physico-chemical properties of the autosomal dominant HSPB1 mutants R127W, S135F and R136W. Despite having a nominal effect on thermal stability, the three mutations induce dramatic changes to quaternary structure. At high concentrations or under crowding conditions, the mutants form assemblies that are approximately two times larger than those formed by the wild-type protein. At low concentrations, the mutants have a higher propensity to dissociate into small oligomers, while the dissociation of R127W and R135F mutants is enhanced by MAPKAP kinase-2 mediated phosphorylation. Specific differences are observed in the ability to form hetero-oligomers with the homologue HSPB6 (HSP20). For wild-type HSPB1 this only occurs at or above physiological temperature, whereas the R127W and S135F mutants form hetero-oligomers with HSPB6 at 4 °C, and the R136W mutant fails to form hetero-oligomers. Combined, the results suggest that the disease-related mutations of HSPB1 modify its self-assembly and interaction with partner proteins thus affecting normal functioning of HSPB1 in the cell.


Subject(s)
Charcot-Marie-Tooth Disease/pathology , HSP27 Heat-Shock Proteins/metabolism , Amino Acid Sequence , Charcot-Marie-Tooth Disease/metabolism , Chromatography, Gel , Dynamic Light Scattering , HSP20 Heat-Shock Proteins/chemistry , HSP20 Heat-Shock Proteins/genetics , HSP20 Heat-Shock Proteins/metabolism , HSP27 Heat-Shock Proteins/chemistry , HSP27 Heat-Shock Proteins/genetics , Heat-Shock Proteins , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Molecular Chaperones , Phosphorylation , Polymorphism, Single Nucleotide , Protein Domains , Protein Multimerization , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Protein Structure, Quaternary , Scattering, Small Angle , Sequence Alignment , Temperature , X-Ray Diffraction , alpha-Crystallins/chemistry
20.
Anal Chem ; 89(24): 13275-13282, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29135234

ABSTRACT

The quaternary structure and dynamics of the human small heat-shock protein Hsp27 are linked to its molecular chaperone function and influenced by post-translational modifications, including phosphorylation. Phosphorylation of Hsp27 promotes oligomer dissociation and can enhance chaperone activity. This study explored the impact of phosphorylation on the quaternary structure and dynamics of Hsp27. Using mutations that mimic phosphorylation, and ion mobility mass spectrometry, we show that successive substitutions result in an increase in the conformational heterogeneity of Hsp27 dimers. In contrast, we did not detect any changes in the structure of an Hsp27 12-mer, representative of larger Hsp27 oligomers. Our data suggest that oligomer dissociation and increased flexibility of the dimer contribute to the enhanced chaperone activity of phosphorylated Hsp27. Thus, post-translational modifications such as phosphorylation play a crucial role in modulating both the tertiary and quaternary structure of Hsp27, which is pivotal to its function as a key component of the proteostasis network in cells. Our data demonstrate the utility of ion mobility mass spectrometry for probing the structure and dynamics of heterogeneous proteins.


Subject(s)
HSP27 Heat-Shock Proteins/chemistry , Ion Mobility Spectrometry , Molecular Dynamics Simulation , Fluorescence Resonance Energy Transfer , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Molecular Chaperones , Phosphorylation , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...