Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Biochimie ; 219: 146-154, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38016530

ABSTRACT

Small heat shock proteins are the well-known regulators of the cytoskeleton integrity, yet their complexes with actin-binding proteins are underexplored. Filamin C, a dimeric 560 kDa protein, abundant in cardiac and skeletal muscles, crosslinks actin filaments and contributes to Z-disc formation and membrane-cytoskeleton attachment. Here, we analyzed the interaction of a human filamin C fragment containing immunoglobulin-like domains 22-24 (FLNC22-24) with five small heat shock proteins (HspB1, HspB5, HspB6, HspB7, HspB8) and their α-crystallin domains. On size-exclusion chromatography, only HspB7 or its α-crystallin domain formed complexes with FLNC22-24. Despite similar isoelectric points of the small heat shock proteins analyzed, only HspB7 and its α-crystallin domain interacted with FLNC22-24 on native gel electrophoresis. Crosslinking with glutaraldehyde confirmed the formation of complexes between HspB7 (or its α-crystallin domain) and the filamin С fragment, inhibiting intersubunit FLNC crosslinking. These data are consistent with the structure modeling using Alphafold. Thus, the C-terminal fragment (immunoglobulin-like domains 22-24) of filamin C contains the site for HspB7 (or its α-crystallin domain) interaction, which competes with FLNC22-24 dimerization and its probable interaction with different target proteins.


Subject(s)
Heat-Shock Proteins, Small , alpha-Crystallins , Humans , alpha-Crystallins/metabolism , Filamins/metabolism , Heat-Shock Proteins, Small/chemistry , Heat-Shock Proteins, Small/metabolism , HSP27 Heat-Shock Proteins/metabolism , Immunoglobulin Domains
2.
Int J Mol Sci ; 24(2)2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36674601

ABSTRACT

The α-crystallin domain (ACD) is the hallmark of a diverse family of small heat shock proteins (sHsps). We investigated some of the ACD properties of five human sHsps as well as their interactions with different full-length sHsps. According to size-exclusion chromatography, at high concentrations, the ACDs of HspB1 (B1ACD), HspB5 (B5ACD) and HspB6 (B6ACD) formed dimers of different stabilities, which, upon dilution, dissociated to monomers to different degrees. Upon dilution, the B1ACD dimers possessed the highest stabilities, and those of B6ACD had the lowest. In striking contrast, the ACDs of HspB7 (B7ACD) and HspB8 (B8ACD) formed monomers in the same concentration range, which indicated the compromised stabilities of their dimer interfaces. B1ACD, B5ACD and B6ACD transiently interacted with full-length HspB1 and HspB5, which are known to form large oligomers, and modulated their oligomerization behavior. The small oligomers formed by the 3D mutant of HspB1 (mimicking phosphorylation at Ser15, Ser78 and Ser82) effectively interacted with B1ACD, B5ACD and B6ACD, incorporating these α-crystallin domains into their structures. The inherently dimeric full-length HspB6 readily formed heterooligomeric complexes with B1ACD and B5ACD. In sharp contrast to the abovementioned ACDs, B7ACD and B8ACD were unable to interact with full-length HspB1, the 3D mutant of HspB1, HspB5 or HspB6. Thus, their high sequence homology notwithstanding, B7ACD and B8ACD differ from the other three ACDs in their inability to form dimers and interact with the full-length small heat shock proteins. Having conservative primary structures and being apparently similar, the ACDs of the different sHsps differ in terms of their dimer stabilities, which can influence the heterooligomerization preferences of sHsps.


Subject(s)
Heat-Shock Proteins, Small , alpha-Crystallins , Humans , Heat-Shock Proteins, Small/metabolism , Phosphorylation , HSP27 Heat-Shock Proteins/metabolism
3.
Biochemistry (Mosc) ; 87(8): 800-811, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36171660

ABSTRACT

Small heat shock proteins (sHsps) play an important role in the maintenance of proteome stability and, particularly, in stabilization of the cytoskeleton and cell contractile apparatus. Cell exposure to different types of stress is accompanied by the translocation of sHsps onto actin filaments; therefore, it is commonly believed that the sHsps are true actin-binding proteins. Investigations of last years have shown that this assumption is incorrect. Stress-induced translocation of sHsp to actin filaments is not the result of direct interaction of these proteins with intact actin, but results from the chaperone-like activity of sHsps and their interaction with various actin-binding proteins. HspB1 and HspB5 interact with giant elastic proteins titin and filamin thus providing an integrity of the contractile apparatus and its proper localization in the cell. HspB6 binds to the universal adapter protein 14-3-3 and only indirectly affects the structure of actin filament. HspB7 interacts with filamin C and controls actin filament assembly. HspB8 forms tight complex with the universal regulatory and adapter protein Bag3 and participates in the chaperone-assisted selective autophagy (CASA) of actin-binding proteins (e.g., filamin), as well as in the actin-depending processes taking place in mitoses. Hence, the mechanisms of sHsp participation in the maintenance of the contractile apparatus and cytoskeleton are much more complicated and diverse than it has been postulated earlier and are not limited to direct interactions of sHsps with actin. The old hypothesis on the direct binding of sHsps to intact actin should be revised and further detailed investigation on the sHsp interaction with minor proteins participating in the formation and remodeling of actin filaments is required.


Subject(s)
Heat-Shock Proteins, Small , 14-3-3 Proteins/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Connectin , Filamins/metabolism , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins, Small/chemistry , Proteome/metabolism
4.
Biochimie ; 202: 103-109, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35977674

ABSTRACT

It is postulated that the small heat shock proteins directly interact with actin, affect formation and stabilize actin filaments. To verify this suggestion, we have analyzed interaction of recombinant human small heat shock protein HspB7 with skeletal muscle actin. In blot overlay HspB7 binds both G- and F-actin. The sites of interaction are located in the C-terminal large core domain of actin. In the course of ultracentrifugation F-actin and F-actin/tropomyosin complexes were pelleted and trapped HspB7. However, HspB7 pelleting was nonspecific and saturation was not achieved even at very high HspB7 concentration. HspB7 was unable to retard or prevent heat-induced F-actin aggregation. Native gel electrophoresis and chemical crosslinking failed to detect interaction of G-actin with HspB7, although both these methods clearly demonstrated formation of complexes formed by G-actin with DNAse I and cofilin-2. It is concluded that HspB7 is not a genuine actin-binding protein and its effect on actin filaments seems to be determined by interaction of HspB7 with minor regulatory proteins of actin filaments.


Subject(s)
Actins , Heat-Shock Proteins, Small , Humans , HSP27 Heat-Shock Proteins , Actin Cytoskeleton , Tropomyosin
5.
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
6.
PLoS One ; 16(6): e0253432, 2021.
Article in English | MEDLINE | ID: mdl-34143841

ABSTRACT

The small heat shock protein (sHsp) called HspB8 (formerly, Hsp22) is one of the least typical sHsp members, whose oligomerization status remains debatable. Here we analyze the effect of mutations in a highly conservative sequence located in the N-terminal domain of human HspB8 on its physico-chemical properties and chaperone-like activity. According to size-exclusion chromatography coupled to multi-angle light scattering, the wild type (WT) HspB8 is present as dominating monomeric species (~24 kDa) and a small fraction of oligomers (~60 kDa). The R29A amino acid substitution leads to the predominant formation of 60-kDa oligomers, leaving only a small fraction of monomers. Deletion of the 28-32 pentapeptide (Δ mutant) results in the formation of minor quantities of dimers (~49 kDa) and large quantities of the 24-kDa monomers. Both the WT protein and its Δ mutant efficiently bind a hydrophobic probe bis-ANS and are relatively rapidly hydrolyzed by chymotrypsin, whereas the R29A mutant weakly binds bis-ANS and resists chymotrypsinolysis. In contrast to HspB8 WT and its Δ mutant, which are well phosphorylated by cAMP-dependent and ERK1 protein kinases, the R29A mutant is poorly phosphorylated. R29A mutation affects the chaperone-like activity of HspB8 measured in vitro. It is concluded that the irreplaceable Arg residue located in the only highly conservative motif in the N-terminal domain of all sHsp proteins affects the oligomeric structure and key properties of HspB8.


Subject(s)
Amino Acid Substitution , Heat-Shock Proteins, Small/metabolism , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Amino Acid Sequence , Humans , Mutation
7.
Biochemistry (Mosc) ; 86(Suppl 1): S1-S11, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33827396

ABSTRACT

HspB7 is one of ten human small heat shock proteins. This protein is expressed only in insulin-dependent tissues (heart, skeletal muscle, and fat tissue), and expression of HspB7 is regulated by many different factors. Single nucleotide polymorphism is characteristic for the HspB7 gene and this polymorphism correlates with cardio-vascular diseases and obesity. HspB7 has an unusual N-terminal sequence, a conservative α-crystallin domain, and very short C-terminal domain lacking conservative IPV tripeptide involved in a small heat shock proteins oligomer formation. Nevertheless, in the isolated state HspB7 forms both small oligomers (probably dimers) and very large oligomers (aggregates). HspB7 is ineffective in suppression of amorphous aggregation of model proteins induced by heating or reduction of disulfide bonds, however it is very effective in prevention of aggregation of huntingtin fragments enriched with Gln residues. HspB7 can be an effective sensor of electrophilic agents. This protein interacts with the contractile and cytoskeleton proteins (filamin C, titin, and actin) and participates in protection of the contractile apparatus and cytoskeleton from different adverse conditions. HspB7 possesses tumor suppressive activity. Further investigations are required to understand molecular mechanisms of HspB7 participation in numerous biological processes.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , Animals , Gene Expression Regulation , HSP27 Heat-Shock Proteins/genetics , Humans , Muscle, Skeletal/metabolism , Myocardium/metabolism
8.
Int J Mol Sci ; 21(12)2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32549212

ABSTRACT

Ubiquitously expressed human small heat shock proteins (sHsps) HspB1, HspB5, HspB6 and HspB8 contain a conserved motif (S/G)RLFD in their N-terminal domain. For each of them, we prepared mutants with a replacement of the conserved R by A (R/A mutants) and a complete deletion of the pentapeptide (Δ mutants) and analyzed their heterooligomerization with other wild-type (WT) human sHsps. We found that WT HspB1 and HspB5 formed heterooligomers with HspB6 only upon heating. In contrast, both HspB1 mutants interacted with WT HspB6 even at low temperature. HspB1/HspB6 heterooligomers revealed a broad size distribution with equimolar ratio suggestive of heterodimers as building blocks, while HspB5/HspB6 heterooligomers had an approximate 2:1 ratio. In contrast, R/A or Δ mutants of HspB6, when mixed with either HspB1 or HspB5, resulted in heterooligomers with a highly variable molar ratio and a decreased HspB6 incorporation. No heterooligomerization of HspB8 or its mutants with either HspB1 or HspB5 could be detected. Finally, R/A or Δ mutations had no effect on heterooligomerization of HspB1 and HspB5 as analyzed by ion exchange chromatography. We conclude that the conserved N-terminal motif plays an important role in heterooligomer formation, as especially pronounced in HspB6 lacking the C-terminal IXI motif.


Subject(s)
Heat-Shock Proteins, Small/chemistry , Heat-Shock Proteins, Small/metabolism , Amino Acid Motifs , Chromatography, Gel , Heat-Shock Proteins, Small/genetics , Humans , Mutation , Protein Domains , Protein Multimerization
9.
Exp Eye Res ; 197: 108091, 2020 08.
Article in English | MEDLINE | ID: mdl-32533979

ABSTRACT

Physico-chemical properties of three cataract-associated missense mutants of αB-crystallin (HspB5) (R11H, P20S, R56W) were analyzed. The oligomers formed by the R11H mutant were smaller, whereas the oligomers of the P20S and R56W mutants were larger than those of the wild-type protein. The P20S mutant possessed lower thermal stability than the wild-type HspB5 or two other HspB5 mutants. All HspB5 mutants were able to form heterooligomeric complexes with αA-crystallin (HspB4), a genuine component of eye lens. However, the P20S and R56W mutants were less effective in the formation of these complexes and properties of heterooligomeric complexes formed by these mutants and HspB4 and analyzed by ion-exchange chromatography were different from those formed by the wild-type HspB5 and HspB4. All HspB5 variants also heterooligomerized with another partner protein, HspB6. Specifically for the P20S mutant forming two distinct sizes of homooligomers, only the smaller homooligomer population was able to interact with HspB6. P20S and R56W mutants possessed lower chaperone-like activity than the wild-type HspB5 when UV-irradiated ßL-crystallin was used as a model substrate. Importantly, all three mutations are localized in three earlier postulated short α-helical regions present in the N-terminal domain of αB-crystallin. These observations suggest an important structural and functional role of these regions. Correspondingly, therein localized mutations ultimately result in clinically relevant cataracts.


Subject(s)
Cataract/genetics , DNA/genetics , Mutation , alpha-Crystallin B Chain/genetics , Cataract/metabolism , DNA Mutational Analysis , Humans , alpha-Crystallin B Chain/metabolism
10.
Biochimie ; 174: 126-135, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32353387

ABSTRACT

Physico-chemical properties of HspB6 S10F and P20L mutants with abrogated cardioprotective activity and associated with different forms of cardiomyopathy were analyzed. Under normal conditions both the wild-type HspB6 and its mutants formed small size oligomers (dimers) with apparent molecular weight of 50-60 kDa. Under crowding conditions (0.5 M trimethylamine N-oxide, TMAO) the wild-type HspB6 remained predominantly dimeric or formed small molecular weight complexes, whereas both mutants tended to form high molecular weight complexes. Catalytic subunit of cAMP-dependent protein kinase phosphorylated the wild-type HspB6 and its S10F mutant with comparable rate. The rate of P20L mutant phosphorylation was higher than that of the wild-type HspB6. S10F and P20L mutations did not affect interaction of phosphorylated HspB6 with universal adapter proteins 14-3-3. The wild-type HspB6 was resistant to heat-induced denaturation and aggregation, whereas both its mutants were denatured and started to aggregate at temperature much lower than its wild-type counterpart. Titration with fluorescent probe bis-ANS was accompanied by larger increase of fluorescence in the case of both mutants than in the case of the wild-type HspB6. Both mutants possessed higher chaperone-like activity than the wild-type protein. It is concluded that both S10F and P20L mutations are accompanied by increase of hydrophobicity of the very N-terminal region of HspB6 leading to increased aggregation at elevated temperature, formation of large complexes under crowding conditions and increased chaperone-like activity measured in vitro. Increased hydrophobicity and self-association can affect substrate specificity and interaction with certain target proteins thus leading to decrease or complete abrogation of cardioprotective activity.


Subject(s)
HSP20 Heat-Shock Proteins/chemistry , HSP20 Heat-Shock Proteins/genetics , Humans , Mutation , Phosphorylation , Protein Structure, Quaternary
11.
Cell Stress Chaperones ; 25(4): 655-665, 2020 07.
Article in English | MEDLINE | ID: mdl-32301006

ABSTRACT

Charcot-Marie-Tooth (CMT) disease is major hereditary neuropathy. CMT has been linked to mutations in a range of proteins, including the small heat shock protein HspB1. Here we review the properties of several HspB1 mutants associated with CMT. In vitro, mutations in the N-terminal domain lead to a formation of larger HspB1 oligomers when compared with the wild-type (WT) protein. These mutants are resistant to phosphorylation-induced dissociation and reveal lower chaperone-like activity than the WT on a range of model substrates. Mutations in the α-crystallin domain lead to the formation of yet larger HspB1 oligomers tending to dissociate at low protein concentration and having variable chaperone-like activity. Mutations in the conservative IPV motif within the C-terminal domain induce the formation of very large oligomers with low chaperone-like activity. Most mutants interact with a partner small heat shock protein, HspB6, in a manner different from that of the WT protein. The link between the altered physico-chemical properties and the pathological CMT phenotype is a subject of discussion. Certain HspB1 mutations appear to have an effect on cytoskeletal elements such as intermediate filaments and/or microtubules, and by this means damage the axonal transport. In addition, mutations of HspB1 can affect the metabolism in astroglia and indirectly modulate the viability of motor neurons. While the mechanisms of pathological mutations in HspB1 are likely to vary greatly across different mutations, further in vitro and in vivo studies are required for a better understanding of the CMT disease at molecular level.


Subject(s)
Astrocytes/metabolism , Charcot-Marie-Tooth Disease/metabolism , Heat-Shock Proteins , Molecular Chaperones , Motor Neurons/metabolism , Astrocytes/pathology , HSP20 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/physiology , Humans , Molecular Chaperones/genetics , Molecular Chaperones/physiology , Motor Neurons/pathology , Mutation , Protein Domains/genetics , Proteostasis
12.
Cell Stress Chaperones ; 24(2): 419-426, 2019 03.
Article in English | MEDLINE | ID: mdl-30756294

ABSTRACT

This study analyzed the interaction of commercial monoclonal anti-methylglyoxal antibodies that predominantly recognize argpyrimidine with unmodified and modified model proteins and small heat shock proteins. These antibodies specifically recognize methylglyoxal (MG)-modified bovine serum albumin and lysozyme, but they react equally well with both unmodified and MG-modified HspB1. Mutation R188W decreased the interaction of these antibodies with unmodified HspB1, thus indicating that this residue participates in the formation of antigenic determinant. However, these antibodies did not recognize either short (ESRAQ) or long (IPVTFESRAQLGGP) peptides with primary structure identical to that at Arg188 of HspB1. Neither of the peptides obtained after the cleavage of HspB1 at Met or Cys residues were recognized by anti-argpyrimidine antibodies. This means that unmodified HspB1 contains a discontinuous epitope that includes the sequence around Arg188 and that this epitope is recognized by anti-argpyrimidine antibodies in unmodified HspB1. Incubation of HspB1 with MG is accompanied by the accumulation of hydroimidazolones, but not argpyrimidines. Therefore, conclusions based on utilization of anti-argpyrimidine antibodies and indicating that HspB1 is the predominant and preferential target of MG modification in the cell require revision.


Subject(s)
HSP27 Heat-Shock Proteins/metabolism , Protein Processing, Post-Translational , Pyruvaldehyde/metabolism , Antibodies, Monoclonal , Binding Sites , HSP27 Heat-Shock Proteins/genetics , HeLa Cells , Heat-Shock Proteins , Humans , Molecular Chaperones , Mutation , Ornithine/analogs & derivatives , Ornithine/immunology , Peptides/metabolism , Protein Binding , Pyrimidines/immunology , Pyruvaldehyde/immunology
13.
Cell Stress Chaperones ; 24(2): 295-308, 2019 03.
Article in English | MEDLINE | ID: mdl-30758704

ABSTRACT

Small Heat Shock Proteins (sHSPs) evolved early in the history of life; they are present in archaea, bacteria, and eukaryota. sHSPs belong to the superfamily of molecular chaperones: they are components of the cellular protein quality control machinery and are thought to act as the first line of defense against conditions that endanger the cellular proteome. In plants, sHSPs protect cells against abiotic stresses, providing innovative targets for sustainable agricultural production. In humans, sHSPs (also known as HSPBs) are associated with the development of several neurological diseases. Thus, manipulation of sHSP expression may represent an attractive therapeutic strategy for disease treatment. Experimental evidence demonstrates that enhancing the chaperone function of sHSPs protects against age-related protein conformation diseases, which are characterized by protein aggregation. Moreover, sHSPs can promote longevity and healthy aging in vivo. In addition, sHSPs have been implicated in the prognosis of several types of cancer. Here, sHSP upregulation, by enhancing cellular health, could promote cancer development; on the other hand, their downregulation, by sensitizing cells to external stressors and chemotherapeutics, may have beneficial outcomes. The complexity and diversity of sHSP function and properties and the need to identify their specific clients, as well as their implication in human disease, have been discussed by many of the world's experts in the sHSP field during a dedicated workshop in Québec City, Canada, on 26-29 August 2018.


Subject(s)
Heat-Shock Proteins, Small , Aging/metabolism , Evolution, Molecular , Heat-Shock Proteins, Small/chemistry , Heat-Shock Proteins, Small/metabolism , Heat-Shock Proteins, Small/physiology , Humans , Neoplasms/metabolism , Nervous System Diseases/metabolism , Plants/metabolism , Protein Conformation
14.
Biochem Biophys Res Commun ; 508(4): 1101-1105, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30551876

ABSTRACT

Small heat shock proteins (sHsps) are molecular chaperones preventing protein aggregation. Dynamics of quaternary structure plays an important role in the chaperone-like activity of sHsps. However, an interrelation between the oligomeric state and chaperone-like activity of sHsps remains insufficiently characterized. Most of the accumulated data were obtained in dilute protein solutions, leaving the question of the oligomeric state of sHsps in crowded intracellular media largely unanswered. Here, we analyzed the effect of crowding on the oligomeric state of αB-crystallin (αB-Cr) using analytical ultracentrifugation. Marked increase in the sedimentation coefficient of αB-Cr was observed in the presence of polyethylene glycol (PEG), polyvinylpyrrolidone (PVP) and trimethylamine N-oxide (TMAO) at 48 °C. An especially pronounced effect was detected for the PEG and TMAO mixture, where the sedimentation coefficient (s20,w) of αB-Cr increased from 10.7 S in dilute solution up to 40.7 S in the presence of crowding agents. In the PEG + TMAO mixture, addition of model protein substrate (muscle glycogen phosphorylase b) induced dissociation of large αB-Cr oligomers and formation of complexes with smaller sedimentation coefficients, supporting the idea that, under crowding conditions, protein substrates can promote dissociation of large αB-Cr oligomers.


Subject(s)
Protein Multimerization , alpha-Crystallin B Chain/chemistry , Area Under Curve , Dynamic Light Scattering , Glycogen Phosphorylase/metabolism , Humans , Protein Structure, Quaternary , Temperature
15.
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
16.
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
18.
Biochimie ; 142: 168-178, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28919577

ABSTRACT

Physico-chemical properties of G154S, R157H and A171T mutants of αB-crystallin (HspB5) associated with congenital human diseases including certain myopathies and cataract were investigated. Oligomers formed by G154S and A171T mutants have the size and apparent molecular weight indistinguishable from those of the wild-type HspB5, whereas the size of oligomers formed by R157H mutant is slightly smaller. All mutants are less thermostable and start to aggregate at a lower temperature than the wild-type protein. All mutants effectively interact with a triple phosphomimicking mutant of HspB1 and form large heterooligomeric complexes of similar composition. All mutants interact with HspB6 forming heterooligomeric complexes with size and composition dependent on the molar ratio of two proteins. The wild-type HspB5 and its G154S and A171T mutants form only high molecular weight (300-450 kDa) heterooligomeric complexes with HspB6, whereas the R157H mutant forms both high and low (∼120 kDa) molecular weight complexes. The wild-type HspB5 and its G154S and A171T mutants form two types of heterooligomers with HspB4, whereas R157H mutant effectively forms only one type of heterooligomers with HspB4. G154S and A171T mutants have lower chaperone-like activity than the wild-type protein when subfragment S1 of myosin or ßL-crystallin are used as a model substrates. With these substrates, the R157H mutant shows equal or higher chaperone activity than the wild-type HspB5. We hypothesize that the mutations in the C-terminal region modulate the binding of the IP(I/V) motif to the core α-crystallin domain. The R157H mutation is located in the immediate proximity of this motif. Such modulation could cause altered interaction of HspB5 with partners and substrates and eventually lead to pathological processes.


Subject(s)
Amino Acid Substitution , Mutation , alpha-Crystallin B Chain/chemistry , alpha-Crystallin B Chain/genetics , Crystallins/metabolism , HSP20 Heat-Shock Proteins/metabolism , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins , Humans , Molecular Chaperones , Molecular Weight , Protein Domains , Proteolysis , Temperature , Trypsin/metabolism , alpha-Crystallin B Chain/metabolism
19.
Biochem Biophys Res Commun ; 489(2): 200-205, 2017 07 22.
Article in English | MEDLINE | ID: mdl-28551403

ABSTRACT

Polyelectrolytes are a prospective tool for protection of proteins against aggregation. We compared synthetic polyanion, poly(styrene sulfonate), and natural chaperones of different types, namely, GroEL-like chaperonin from Pseudomonas aeruginosa phage EL and human small heat shock protein HspB5 (αB-crystallin), in their ability to prevent aggregation of client proteins. At 45 °C, all three agents efficiently suppressed thermal aggregation of phage endolysin. At higher temperatures, HspB5 and poly(styrene sulfonate) also inhibited endolysin aggregation, though polyanion became less efficient than HspB5 at 55 °C and 60 °C. However, the polyanion completely protected another protein, glyceraldehyde-3-phosphate dehydrogenase, even at 60 °C, in contrast to both natural chaperones whose effect disappeared at 50-55 °C. These results provide a platform for the development of artificial chaperones based on synthetic polyelectrolytes.


Subject(s)
Hot Temperature , Molecular Chaperones/metabolism , Polystyrenes/metabolism , Pseudomonas aeruginosa/chemistry , alpha-Crystallin B Chain/metabolism , Endopeptidases/metabolism , Humans , Molecular Chaperones/chemistry , Polystyrenes/chemistry , Pseudomonas aeruginosa/metabolism , alpha-Crystallin B Chain/chemistry
20.
Cell Stress Chaperones ; 22(4): 601-611, 2017 07.
Article in English | MEDLINE | ID: mdl-28364346

ABSTRACT

Small heat shock proteins (sHSPs) are present in all kingdoms of life and play fundamental roles in cell biology. sHSPs are key components of the cellular protein quality control system, acting as the first line of defense against conditions that affect protein homeostasis and proteome stability, from bacteria to plants to humans. sHSPs have the ability to bind to a large subset of substrates and to maintain them in a state competent for refolding or clearance with the assistance of the HSP70 machinery. sHSPs participate in a number of biological processes, from the cell cycle, to cell differentiation, from adaptation to stressful conditions, to apoptosis, and, even, to the transformation of a cell into a malignant state. As a consequence, sHSP malfunction has been implicated in abnormal placental development and preterm deliveries, in the prognosis of several types of cancer, and in the development of neurological diseases. Moreover, mutations in the genes encoding several mammalian sHSPs result in neurological, muscular, or cardiac age-related diseases in humans. Loss of protein homeostasis due to protein aggregation is typical of many age-related neurodegenerative and neuromuscular diseases. In light of the role of sHSPs in the clearance of un/misfolded aggregation-prone substrates, pharmacological modulation of sHSP expression or function and rescue of defective sHSPs represent possible routes to alleviate or cure protein conformation diseases. Here, we report the latest news and views on sHSPs discussed by many of the world's experts in the sHSP field during a dedicated workshop organized in Italy (Bertinoro, CEUB, October 12-15, 2016).


Subject(s)
Heat-Shock Proteins, Small/chemistry , Heat-Shock Proteins, Small/metabolism , Animals , Heart Diseases/metabolism , Humans , Muscular Diseases/metabolism , Neurodegenerative Diseases/metabolism , Protein Aggregates , Protein Conformation , Protein Interaction Maps
SELECTION OF CITATIONS
SEARCH DETAIL
...