Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Mol Biosci ; 9: 875205, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35463950

RESUMO

The G98R mutation in αA-crystallin is associated with presenile cataract development in humans. Previous studies have indicated that mutant proteins altered structure, decreased stability, increased oligomeric size, loss of chaperone-like activity, and susceptibility to proteolysis could be contributing factors to cataract formation. To evaluate the effect of substrate protein interactions with the mutant protein on cataract formation, we have performed chaperone assays with alcohol dehydrogenase (ADH), citrate synthase (CS), and ßB2-crystallin (ßB2), and analyzed the reaction mixtures by multi-angle light scattering (MALS) analysis. It appears that αAG98R protein initially gets stabilized upon interaction with substrate proteins. Analysis of the chaperone-client protein complexes revealed that wild-type αA-crystallin interacts with substrate proteins to form compact complexes leading to a slight increase in oligomeric mass, whereas αAG98R forms less compact and high molecular weight complexes with the substrate, and the resulting complexes continue to increase in size over time. As a result, the soluble complexes formed initially by the mutant protein begin to scatter light and precipitate. We found that the stability and chaperone activity of the αAG98R can be improved by modifying the protein with low concentrations (50 µM) of methylglyoxal (MGO). Incubation of αAG98R protein (1 mg/ml) under aseptic conditions for 30 days at 37°C resulted in precipitation of the mutant protein. In contrast, mutant protein incubations carried out with 50 µM MGO remained soluble and transparent. SDS-PAGE analysis showed gradual autolysis of the mutant protein in the absence of MGO. The average molar mass of the mutant protein oligomers changed from 7,258 ± 12 kDa to 3,950 ± 08 kDa within 60 min of incubation with MGO. There was no further significant change in the molar mass of mutant protein when tested on day 7 of MGO treatment. Our data suggest that the initial stabilization of αAG98R by substrate proteins could delay congenital cataracts' appearance, and the uncontrolled long-term interaction amongst mutant subunits and substrate proteins could be the rationale behind presenile cataracts formation. The results also demonstrate the potential benefit of low concentrations of MGO in stabilizing mutant chaperone protein(s).

2.
Int J Mol Sci ; 23(3)2022 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-35163023

RESUMO

Smaller oligomeric chaperones of α-crystallins (αA- and αB-) have received increasing attention due to their improved therapeutic potential in preventing protein aggregating diseases. Our previous study suggested that deleting 54-61 residues from the N-terminal domain (NTD) of αB-crystallin (αBΔ54-61) decreases the oligomer size and increases the chaperone function. Several studies have also suggested that NTD plays a significant role in protein oligomerization and chaperone function. The current study was undertaken to assess the effect of deleting conserved 21-28 residues from the activated αBΔ54-61 (to get αBΔ21-28, Δ54-61) on the structure-function of recombinant αBΔ21-28, Δ54-61. The αBΔ21-28, Δ54-61 mutant shows an 80% reduction in oligomer size and 3- to 25-fold increases in chaperone activity against model substrates when compared to αB-WT. Additionally, the αB∆21-28, ∆54-61 was found to prevent ß-amyloid (Aß1-42) fibril formation in vitro and suppressed Aß1-42-induced cytotoxicity in ARPE-19 cells in a more effective manner than seen with αB-WT or αB∆54-61. Cytotoxicity and reactive oxygen species (ROS) detection studies with sodium iodate (SI) showed that the double mutant protein has higher anti-apoptotic and anti-oxidative activities than the wild-type or αB∆54-61 in oxidatively stressed cells. Our study shows that the residues 21-28 and 54-61 in αB-crystallin contribute to the oligomerization and modulate chaperone function. The deletion of conserved 21-28 residues further potentiates the activated αBΔ54-61. We propose that increased substrate affinity, altered subunit structure, and assembly leading to smaller oligomers could be the causative factors for the increased chaperone activity of αBΔ21-28, Δ54-61.


Assuntos
Antioxidantes/farmacologia , Chaperonas Moleculares/farmacologia , Mutação , Estresse Oxidativo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Cadeia B de alfa-Cristalina/farmacologia , Sequência de Aminoácidos , Apoptose , Células Cultivadas , Humanos , Mutagênese Sítio-Dirigida , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Cadeia B de alfa-Cristalina/química , Cadeia B de alfa-Cristalina/genética
3.
Int J Mol Sci ; 22(19)2021 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-34639110

RESUMO

Previously, we showed that the removal of the 54-61 residues from αB-crystallin (αBΔ54-61) results in a fifty percent reduction in the oligomeric mass and a ten-fold increase in chaperone-like activity. In this study, we investigated the oligomeric organization changes in the deletion mutant contributing to the increased chaperone activity and evaluated the cytoprotection properties of the mutant protein using ARPE-19 cells. Trypsin digestion studies revealed that additional tryptic cleavage sites become susceptible in the deletion mutant than in the wild-type protein, suggesting a different subunit organization in the oligomer of the mutant protein. Static and dynamic light scattering analyses of chaperone-substrate complexes showed that the deletion mutant has more significant interaction with the substrates than wild-type protein, resulting in increased binding of the unfolding proteins. Cytotoxicity studies carried out with ARPE-19 cells showed an enhancement in anti-apoptotic activity in αBΔ54-61 as compared with the wild-type protein. The improved anti-apoptotic activity of the mutant is also supported by reduced caspase activation and normalization of the apoptotic cascade components level in cells treated with the deletion mutant. Our study suggests that altered oligomeric assembly with increased substrate affinity could be the basis for the enhanced chaperone function of the αBΔ54-61 protein.


Assuntos
Apoptose , Chaperonas Moleculares/metabolismo , Fragmentos de Peptídeos/química , Epitélio Pigmentado da Retina/patologia , Cadeia B de alfa-Cristalina/química , Células Cultivadas , Humanos , Estrutura Secundária de Proteína , Epitélio Pigmentado da Retina/metabolismo , Deleção de Sequência , Cadeia B de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/metabolismo
4.
Biochemistry ; 58(40): 4148-4158, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31523965

RESUMO

The G98R mutation in αA-crystallin is associated with the onset of presenile cataract and is characterized biochemically by an increased oligomeric mass, altered chaperone function, and loss of structural stability over time. Thus, far, it is not known whether the inherent instability caused by gain-of-charge mutation could be rescued by a compensatory loss of charge mutation elsewhere on the protein. To answer this question, we investigated whether αA-G98R-mediated instability could be rescued through suppressor mutations by introducing site-specific "compensatory" mutations in αA-G98R-crystallin, αA-R21Q/G98R, αA-G98R/R116C, and αA-R157Q/G98R. The recombinant proteins were expressed, purified, characterized, and evaluated by circular dichroism (CD), intrinsic fluorescence, and bis-ANS-binding studies. Chaperone-like activities of recombinant proteins were assessed using alcohol dehydrogenase (ADH) and insulin as unfolding substrates. Far-UV CD studies revealed an increased α-helical content in αA-G98R in comparison to αA-WT, αA-R21Q, R157Q, and the double mutants, αA-R21Q/G98R, and αA-R157Q/G98R. Compared to αA-WT, αA-R21Q, and αA-G98R, the double mutants showed an increased intrinsic tryptophan fluorescence, whereas the highest hydrophobicity (bis-ANS-binding) was shown by αA-G98R. Introduction of a second mutation in αA-G98R reduced its bis-ANS-binding activity. Both αA-R21Q/G98R and αA-R157Q/G98R showed greater chaperone-like activity against ADH aggregation than αA-G98R. However, among the three G98R mutants, only αA-R21Q/G98R protected ARPE-19 cells from H2O2-induced cytotoxicity. These results suggest that the lost chaperone-like activity of αA-G98R-crystallin can be rescued by another targeted mutation and that substitution of αA-R21Q-crystallin at the N-terminal region can rescue a deleterious mutation in the conserved α-crystallin domain of the protein.


Assuntos
Proteínas Recombinantes/metabolismo , Cadeia A de alfa-Cristalina/metabolismo , Álcool Desidrogenase/metabolismo , Sequência de Bases , Linhagem Celular , Sobrevivência Celular/genética , Humanos , Peróxido de Hidrogênio/farmacologia , Insulina/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Multimerização Proteica/genética , Estabilidade Proteica , Desdobramento de Proteína , Proteínas Recombinantes/genética , Supressão Genética , Cadeia A de alfa-Cristalina/genética
5.
Sci Rep ; 9(1): 8459, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31186457

RESUMO

The paradigm that cataracts are irreversible and that vision from cataracts can only be restored through surgery has recently been challenged by reports that oxysterols such as lanosterol and 25-hydroxycholesterol can restore vision by binding to αB-crystallin chaperone protein to dissolve or disaggregate lenticular opacities. To confirm this premise, in vitro rat lens studies along with human lens protein solubilization studies were conducted. Cataracts were induced in viable rat lenses cultured for 48 hours in TC-199 bicarbonate media through physical trauma, 10 mM ouabain as Na+/K+ ATPase ion transport inhibitor, or 1 mM of an experimental compound that induces water influx into the lens. Subsequent 48-hour incubation with 15 mM of lanosterol liposomes failed to either reverse these lens opacities or prevent the further progression of cataracts to the nuclear stage. Similarly, 3-day incubation of 47-year old human lenses in media containing 0.20 mM lanosterol or 60-year-old human lenses in 0.25 and 0.50 mM 25-hydroxycholesterol failed to increase the levels of soluble lens proteins or decrease the levels of insoluble lens proteins. These binding studies were followed up with in silico binding studies of lanosterol, 25-hydroxycholesterol, and ATP as a control to two wild type (2WJ7 and 2KLR) and one R120G mutant (2Y1Z) αB-crystallins using standard MOETM (Molecular Operating Environment) and Schrödinger's Maestro software. Results confirmed that compared to ATP, both oxysterols failed to reach the acceptable threshold binding scores for good predictive binding to the αB-crystallins. In summary, all three studies failed to provide evidence that lanosterol or 25-hydroxycholesterol have either anti-cataractogenic activity or bind aggregated lens protein to dissolve cataracts.


Assuntos
Catarata/tratamento farmacológico , Lanosterol/farmacologia , Cristalino/efeitos dos fármacos , Cadeia B de alfa-Cristalina/genética , Animais , Catarata/metabolismo , Catarata/patologia , Cristalinas/genética , Modelos Animais de Doenças , Humanos , Hidroxicolesteróis/metabolismo , Lanosterol/efeitos adversos , Cristalino/metabolismo , Chaperonas Moleculares/metabolismo , Chaperonas Moleculares/farmacologia , Oxisteróis/efeitos adversos , Oxisteróis/farmacologia , Ratos
6.
Adv Biosyst ; 2(1)2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30271873

RESUMO

Many of the newly discovered therapeutic peptides and molecules are limited by their inability to cross the cell membrane. In the present study we employed a cell penetrating peptide (CPP), VPTLK, derived from Ku70 protein, to facilitate the entry of a mini-chaperone across the cell membrane. Our previous studies suggest that the mini-chaperone peptide representing the chaperone site in αA-crystallin, which can inhibit protein aggregation associated with proteopathies, has therapeutic potential. We have prepared a synthetic mini-chaperone by fusing the VPTLK sequence to N-terminus of mini-chaperone (FVIFLDVKHFSPEDLTVKGRD) to get VPTLKFVIFLDVKHFSPEDLTVKGRD peptide, which we call "CPPGRD." The amino acids, GRD, were added to increase the solubility of the peptide. The chaperone-like function of CPPGRD was measured using unfolding conditions for alcohol dehydrogenase and α-lactalbumin. The anti-apoptotic action of the peptide chaperone was evaluated using H2O2-induced Cos-7 and ARPE-19 cell apoptosis assays. The results show that the CPPGRD has both chaperone function and anti-apoptotic activity. Additionally, the CPPGRD was found to prevent ß-amyloid fibril formation and suppress ß-amyloid toxicity. The present study demonstrates that the CPPGRD protects unfolding proteins from aggregation and prevents cellular apoptosis. Therefore, the CPPGRD is a mini-chaperone with potential to become a therapeutic agent for protein aggregation diseases.

7.
Mol Vis ; 24: 297-304, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29706763

RESUMO

Purpose: The G98R mutant of αA-crystallin is associated with the development of presenile cataracts. In vitro, the recombinant mutant protein exhibits altered structural and functional characteristics, along with the propensity to aggregate by itself and precipitate. Previously, we have reported that the N-terminal aspartate substituted form of the antiaggregation peptide, D71FVIFLDVKHFSPEDLTVK88 (αA-minichaperone or mini-αA) prevented aggregation of αAG98R. However, the mechanism of stabilization of αAG98R from aggregation is not fully understood. The purpose of this study was to determine whether the surface charge (zeta (ζ) potential) of αAG98R in the presence of the peptide chaperone contributed to the stabilization of mutant protein, and to identify the sites of interaction between αAG98R and the peptide chaperone. Methods: Wild-type αA-crystallin (αAWT) and recombinant mutant αAG98R were purified from Escherichia coli BL21(DE3)pLysS cells. The ζ potential values of αA-crystallins in the presence or absence of αA-minichaperone and purified protein-peptide complexes were estimated in a ζ potential analyzer. Potential regions within αAG98R that bind the αA-minichaperone were investigated by incubating the protein with a photoactivable minichaperone variant, followed by mass spectrometric analysis. Results: Binding of the αA-minichaperone to aggregation-prone αAG98R was accompanied by an increase in the ζ potential from -15.19±0.870 mV corresponding to αAG98R alone to -28.64±1.640 mV for the purified complex. Mass spectrometric analysis identified 1MDVTIQHPWFK11, 13TLGPFYPSR21, 55TVLDSGISEVR65, and 113EFHRR117 as the αA-minichaperone-binding regions in αAG98R. The results suggest the involvement of the N-terminal region and the α-crystallin domain in the peptide-mediated stabilization of αAG98R. Conclusions: The αA-crystallin-derived minichaperone stabilizes αAG98R by compensating its lost surface charge. Methods for increasing the ζ potential of aggregating proteins can be a potential approach for therapy to protein aggregation diseases.


Assuntos
Chaperonas Moleculares/química , Mutação , Peptídeos/química , Agregados Proteicos , Cadeia A de alfa-Cristalina/química , Sequência de Aminoácidos , Sítios de Ligação , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutagênese Sítio-Dirigida , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Eletricidade Estática , Cadeia A de alfa-Cristalina/genética , Cadeia A de alfa-Cristalina/metabolismo
8.
Exp Eye Res ; 174: 185-195, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29782825

RESUMO

Several mutations associated with congenital cataracts in human beings target conserved arginine residues in αA-crystallin. The N-terminal region of αA-crystallin is a "mutational hotspot," with multiple cataract-related mutations reported in this region. Two mutations at arginine 21 in the N-terminal domain of αA-crystallin - αA-R21L and αA-R21W have been associated with congenital cataract. A third mutant of R21, αA-R21Q, was recently identified to be associated with congenital cataract in a South Australian family. The point mutation was reported to compromise the quaternary structure of αA-crystallin by preventing its assembly into higher ordered oligomers. To assess the effect of the αA-R21Q mutation on αA-crystallin function, recombinant αA-R21Q was expressed, purified and characterized in vitro. Compared to wild-type αA-crystallin, the recombinant αA-R21Q exhibits enhanced chaperone-like activity, increased surface hydrophobicity, lesser stability in urea and increased susceptibility to digestion by trypsin. αA-R21Q demonstrated increased binding affinity towards unfolding ADH and bovine lens fiber cell membranes. αA-R21Q homo-oligomers and hetero-oligomers also prevented H2O2-induced apoptosis in ARPE-19 cells. Taken together, αA-R21Q exhibited a gain of function despite subtle structural differences as compared to wild-type αA-crystallin. This study further validates the involvement of arginine 21 in regulating αA-crystallin structure and function.


Assuntos
Catarata , Cristalino/química , Cadeia A de alfa-Cristalina , Animais , Apoptose/fisiologia , Arginina/genética , Arginina/fisiologia , Catarata/genética , Catarata/metabolismo , Bovinos , Humanos , Chaperonas Moleculares/fisiologia , Mutação Puntual , Ligação Proteica/fisiologia , Dobramento de Proteína , Ureia/metabolismo , Cadeia A de alfa-Cristalina/química , Cadeia A de alfa-Cristalina/genética , Cadeia A de alfa-Cristalina/fisiologia
9.
Aging Dis ; 8(1): 57-70, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28203481

RESUMO

In previous studies, we reported the presence of a large number of low-molecular-weight (LMW) peptides in aged and cataract human lens tissues. Among the LMW peptides, a peptide derived from αA-crystallin, αA66-80, was found in higher concentration in aged and cataract lenses. Additional characterization of the αA66-80 peptide showed beta sheet signature, and it formed well-defined unbranched fibrils. Further experimental data showed that αA66-80 peptide binds α-crystallin, impairs its chaperone function, and attracts additional crystallin proteins to the peptide α-crystallin complex, leading to the formation of larger light scattering aggregates. It is well established that Aß peptide exhibits cell toxicity by the generation of hydrogen peroxide. The αA66-80 peptide shares the principal properties of Aß peptide. Therefore, the present study was undertaken to determine whether the fibril-forming peptide αA66-80 has the ability to generate hydrogen peroxide. The results show that the αA66-80 peptide generates hydrogen peroxide, in the amount of 1.2 nM H2O2 per µg of αA66-80 peptide by incubation at 37°C for 4h. We also observed cytotoxicity and apoptotic cell death in αA66-80 peptide-transduced Cos7 cells. As evident, we found more TUNEL-positive cells in αA66-80 peptide transduced Cos7 cells than in control cells, suggesting peptide-mediated cell apoptosis. Additional immunohistochemistry analysis showed the active form of caspase-3, suggesting activation of the caspase-dependent pathway during peptide-induced cell apoptosis. These results confirm that the αA66-80 peptide generates hydrogen peroxide and promotes hydrogen peroxide-mediated cell apoptosis.

10.
Biochim Biophys Acta ; 1864(11): 1529-38, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27524665

RESUMO

The chaperone-like activity of αA-crystallin has an important role in maintaining lens transparency. Previously we identified residues 70-88 as a chaperone site in αA-crystallin. In this study, we deleted the chaperone site residues to generate αAΔ70-76 and αAΔ70-88 mutants and investigated if there are additional substrate-binding sites in αA-crystallin. Both mutant proteins when expressed in E. coli formed inclusion bodies, and on solubilizing and refolding, they exhibited similar structural properties, with a 2- to 3-fold increase in molar mass compared to the molar mass of wild-type protein. The deletion mutants were less stable than the wild-type αA-crystallin. Functionally αAΔ70-88 was completely inactive as a chaperone, while αAΔ70-76 demonstrated a 40-50% reduction in anti-aggregation activity against alcohol dehydrogenase (ADH). Deletion of residues 70-88 abolished the ADH binding sites in αA-crystallin at physiological temperature. At 45°C, cryptic ADH binding site(s) became exposed, which contributed subtly to the chaperone-like activity of αAΔ70-88. Both of the deletion mutants were completely inactive in suppressing aggregation of ßL-crystallin at 53°C. The mutants completely lost the anti-apoptotic property that αA-crystallin exhibits while they protected ARPE-19 (a human retinal pigment epithelial cell line) and primary human primary lens epithelial (HLE) cells from oxidative stress. Our studies demonstrate that residues 70-88 in αA-crystallin act as a primary substrate binding site and account for the bulk of the total chaperone activity. The ß3 and ß4 strands in αA-crystallin comprising 70-88 residues play an important role in maintenance of the structure and in preventing aggregation of denaturing proteins.


Assuntos
Cristalinas/química , Células Epiteliais/metabolismo , Chaperonas Moleculares/química , Epitélio Pigmentado da Retina/metabolismo , Álcool Desidrogenase/química , Álcool Desidrogenase/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Clonagem Molecular , Cristalinas/genética , Cristalinas/metabolismo , Células Epiteliais/citologia , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Modelos Moleculares , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Ligação Proteica , Desnaturação Proteica , Domínios Proteicos , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Epitélio Pigmentado da Retina/citologia , Deleção de Sequência , Relação Estrutura-Atividade
11.
Biochim Biophys Acta ; 1860(1 Pt B): 246-51, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26141743

RESUMO

BACKGROUND: The demonstration of chaperone-like activity in peptides (mini-chaperones) derived from α-crystallin's chaperone region has generated significant interest in exploring the therapeutic potential of peptide chaperones in diseases of protein aggregation. Recent studies in experimental animals show that mini-chaperones could reach intended targets and alter the disease phenotype. Although mini-chaperones show potential benefits against protein aggregation diseases, they do tend to form aggregates on storage. There is thus a need to fine-tune peptide chaperones to increase their solubility, pharmacokinetics, and biological efficacy. SCOPE OF REVIEW: This review summarizes the properties and the potential therapeutic roles of mini-chaperones in protein aggregation diseases and highlights some of the refinements needed to increase the stability and biological efficacy of mini-chaperones while maintaining or enhancing their chaperone-like activity against precipitation of unfolding proteins. MAJOR CONCLUSIONS: Mini-chaperones suppress the aggregation of proteins, block amyloid fibril formation, stabilize mutant proteins, sequester metal ions, and exhibit antiapoptotic properties. Much work must be done to fine-tune mini-chaperones and increase their stability and biological efficacy. Peptide chaperones could have a great therapeutic value in diseases associated with protein aggregation and apoptosis. GENERAL SIGNIFICANCE: Accumulation of misfolded proteins is a primary cause for many age-related diseases, including cataract, macular degeneration, and various neurological diseases. Stabilization of native proteins is a logical therapeutic approach for such diseases. Mini-chaperones, with their inherent antiaggregation and antiapoptotic properties, may represent an effective therapeutic molecule to prevent the cascade of protein conformational disorders. Future studies will further uncover the therapeutic potential of mini-chaperones. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.


Assuntos
Glaucoma/tratamento farmacológico , Peptídeos/uso terapêutico , Agregação Patológica de Proteínas/tratamento farmacológico , Doenças Retinianas/tratamento farmacológico , alfa-Cristalinas/química , Animais , Antioxidantes/uso terapêutico , Glaucoma/patologia , Chaperonas Moleculares/uso terapêutico , Peptídeos/química , Doenças Retinianas/patologia
12.
Invest Ophthalmol Vis Sci ; 55(8): 4731-40, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24994865

RESUMO

PURPOSE: Posterior capsule opacification (PCO) after cataract surgery is due in part to proliferation of the adhering lens epithelial cells and transdifferentiation into mesenchymal cells. The histone deacetylase (HDAC) inhibitors, trichostatin A (TSA) and vorinostat (suberoylanilidehydroxamic acid [SAHA]) are known to modulate cell proliferation and epithelial-mesenchymal transition (EMT). Studies have shown that TGFß2 can induce EMT similar to that seen during PCO. This study evaluated the effects of TSA and SAHA on TGFß2-induced EMT in lens epithelial explants. METHODS: Epithelial cells adherent to lens capsules were isolated from fresh pig lenses and human donor lenses and cultured for 12 hours. Explants were pretreated with TSA or SAHA for 1 hour and then treated with TGFß2 for up to 3 days. Scratch wound healing assay was used to determine epithelial cell proliferation and migration in the samples. The effects of TSA and SAHA on histone acetylation and HDAC 1 to 6 levels were analyzed by Western blotting. RESULTS: Western blotting and immunocytochemistry demonstrated high expression of α-SMA in lens epithelial cells treated with TGFß2. The HDAC inhibitors exerted dose-dependent inhibition of α-SMA expression, with complete inhibition occurring with 0.5 µM of TSA and 2.5 µM of SAHA. Transforming growth factor ß2-induced EMT was suppressed by TSA and SAHA. Histone deacetylase inhibition in pig lens epithelia led to increased acetylation of histone 3 and 4 at multiple sites. CONCLUSIONS: Histone deacetylase inhibitors, TSA, and SAHA prevent EMT in lens epithelial explants. The results also suggest that the epigenetic modifiers are the potential targets to control PCO after cataract surgery.


Assuntos
Actinas/biossíntese , Opacificação da Cápsula/prevenção & controle , Células Epiteliais/metabolismo , Ácidos Hidroxâmicos/farmacologia , Cápsula do Cristalino/metabolismo , Fator de Crescimento Transformador beta2/efeitos adversos , Actinas/efeitos dos fármacos , Animais , Western Blotting , Opacificação da Cápsula/etiologia , Opacificação da Cápsula/metabolismo , Extração de Catarata/efeitos adversos , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Radioisótopos de Flúor , Inibidores de Histona Desacetilases/farmacologia , Humanos , Imuno-Histoquímica , Cápsula do Cristalino/efeitos dos fármacos , Cápsula do Cristalino/patologia , Microscopia de Fluorescência , Pessoa de Meia-Idade , Suínos , Fator de Crescimento Transformador beta2/metabolismo , Vorinostat
13.
Biochemistry ; 53(16): 2615-23, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24697516

RESUMO

It has been shown that αA-mini-chaperone, a peptide representing the chaperone binding site in αA-crystallin, prevents destabilized protein aggregation. αA-Mini-chaperone has been shown to form amyloid fibrils. This study was undertaken to improve the stability of αA-mini-chaperone while preserving its anti-aggregation activity by fusing the flexible and solvent-exposed C-terminal 164-173 region of αA-crystallin to the mini-chaperone sequence DFVIFLDVKHFSPEDLT. The resulting chimeric chaperone peptide, DFVIFLDVKHFSPEDLTEEKPTSAPSS (designated CP1), was characterized. Circular dichroism studies showed that unlike αA-mini-chaperone with its ß-sheet structure, the CP1 peptide exhibited a random structure. Transmission electron microscopy (TEM) examination of the CP1 peptide incubated in a shaker at 37 °C for 72 h did not reveal amyloid fibrils, whereas αA-mini-chaperone showed distinct fibrils. Consistent with TEM observation, the thioflavin T binding assay showed an increased level of dye binding in the mini-chaperone incubated at 37 °C and subjected to shaking but not of the CP1 peptide incubated under similar conditions. The chaperone activity of the CP1 peptide was comparable to that of αA-mini-chaperone against denaturing alcohol dehydrogenase, citrate synthase, and α-lactalbumin. Transduction of both peptide chaperones to COS-7 cells showed no cytotoxic effects. The antioxidation assay involving the H2O2 treatment of COS-7 cells revealed that αA-mini-chaperone and the CP1 peptide have comparable cytoprotective properties against H2O2-induced oxidative damage in COS-7 cells. This study therefore shows that the addition of C-terminal sequence 164-173 of αA-crystallin to αA-mini-chaperone influences the conformation of αA-mini-chaperone without affecting its chaperone function or cytoprotective activity.


Assuntos
Chaperonas Moleculares/metabolismo , Cadeia A de alfa-Cristalina/metabolismo , Sequência de Aminoácidos , Animais , Benzotiazóis , Células COS/efeitos dos fármacos , Células COS/metabolismo , Chlorocebus aethiops , Dicroísmo Circular , Peróxido de Hidrogênio/farmacologia , Microscopia Eletrônica de Transmissão , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Conformação Proteica , Engenharia de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tiazóis/metabolismo , Cadeia A de alfa-Cristalina/química , Cadeia A de alfa-Cristalina/genética
14.
J Biol Chem ; 289(13): 9039-52, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24554718

RESUMO

The accumulation of crystallin fragments in vivo and their subsequent interaction with crystallins are responsible, in part, for protein aggregation in cataracts. Transgenic mice overexpressing acylpeptide hydrolase (APH) specifically in the lens were prepared to test the role of protease in the generation and accumulation of peptides. Cataract development was seen at various postnatal days in the majority of mice expressing active APH (wt-APH). Cataract onset and severity of the cataracts correlated with the APH protein levels. Lens opacity occurred when APH protein levels were >2.6% of the total lens protein and the specific activity, assayed using Ac-Ala-p-nitroanilide substrate, was >1 unit. Transgenic mice carrying inactive APH (mt-APH) did not develop cataract. Cataract development also correlated with N-terminal cleavage of the APH to generate a 57-kDa protein, along with an increased accumulation of low molecular weight (LMW) peptides, similar to those found in aging human and cataract lenses. Nontransgenic mouse lens proteins incubated with purified wt-APH in vitro resulted in a >20% increase in LMW peptides. Crystallin modifications and cleavage were quite dramatic in transgenic mouse lenses with mature cataract. Affected lenses showed capsule rupture at the posterior pole, with expulsion of the lens nucleus and degenerating fiber cells. Our study suggests that the cleaved APH fragment might exert catalytic activity against crystallins, resulting in the accumulation of distinct LMW peptides that promote protein aggregation in lenses expressing wt-APH. The APH transgenic model we developed will enable in vivo testing of the roles of crystallin fragments in protein aggregation.


Assuntos
Catarata/metabolismo , Cristalinas/metabolismo , Cristalino/metabolismo , Peptídeo Hidrolases/genética , Sequência de Aminoácidos , Animais , Catarata/genética , Catarata/patologia , Cristalinas/química , Expressão Gênica , Humanos , Hidrólise , Cristalino/patologia , Camundongos , Camundongos Transgênicos , Mutação , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Peptídeo Hidrolases/metabolismo , Regiões Promotoras Genéticas/genética
15.
Biochemistry ; 52(45): 8126-38, 2013 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-24128140

RESUMO

αB-Crystallin is a chaperone and an anti-apoptotic protein that is strongly expressed in many tissues, including the lens, retina, heart, and kidney. In the human lens, several lysine residues in αB-crystallin are acetylated. We have previously shown that such acetylation is predominant at lysine 92 (K92) and lysine 166 (K166). We have investigated the effect of lysine acetylation on the structure and functions of αB-crystallin by the specific introduction of an N(ε)-acetyllysine (AcK) mimic at K92. The introduction of AcK slightly altered the secondary and tertiary structures of the protein. The introduction of AcK also resulted in an increase in the molar mass and hydrodynamic radius of the protein, and the protein became structurally more open and more stable than the native protein. The acetyl protein acquired higher surface hydrophobicity and exhibited 25-55% higher chaperone activity than the native protein. The acetyl protein had more client protein binding per subunit of the protein and higher binding affinity relative to that of the native protein. The acetyl protein was at least 20% more effective in inhibiting chemically induced apoptosis than the native protein. Molecular modeling suggests that acetylation of K92 makes the "α-crystallin domain" more hydrophobic. Together, our results reveal that the acetylation of a single lysine residue in αB-crystallin makes the protein structurally more stable and improves its chaperone and anti-apoptotic activities. Our findings suggest that lysine acetylation of αB-crystallin is an important chemical modification for enhancing αB-crystallin's protective functions in the eye.


Assuntos
Cristalinas/química , Cristalinas/metabolismo , Lisina/química , Lisina/metabolismo , Acetilação , Western Blotting , Humanos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Chaperonas Moleculares , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
16.
PLoS One ; 8(6): e65610, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23755258

RESUMO

Cataract is characterized by progressive protein aggregation and loss of vision. α-Crystallins are the major proteins in the lens responsible for maintaining transparency. They exist in the lens as highly polydisperse oligomers with variable numbers of subunits, and mutations in α-crystallin are associated with some forms of cataract in humans. Because the stability of proteins is dependent on optimal subunit interactions, the structural transformations and aggregation of mutant proteins that underlie cataract formation can be understood best by identifying the residue-specific inter- and intra-subunit interactions. Chemical crosslinking combined with mass spectrometry is increasingly used to provide structural insights into intra- and inter-protein interactions. We used isotope-labeled cross-linker in combination with LC-MS/MS to determine the subunit-subunit interaction sites in cataract-causing mutant αA-G98R crystallin. Peptides cross-linked by isotope-labeled (heavy and light forms) cross-linkers appear as doublets in mass spectra, thus facilitating the identification of cross-linker-containing peptides. In this study, we cross-linked wild-type (αA-WT) and mutant (αA-G98R) crystallins using the homobifunctional amine-reactive, isotope-labeled (d0 and d4) cross-linker-BS²G (bis[sulfosuccinimidyl]glutarate). Tryptic in-solution digest of cross-linked complexes generates a wide array of peptide mixtures. Cross-linked peptides were enriched using strong cation exchange (SCX) chromatography followed by both MS and MS/MS to identify the cross-linked sites. We identified a distinct intermolecular interaction site between K88-K99 in the ß5 strand of the mutant αA-G98R crystallin that is not found in wild-type αA-crystallin. This interaction could explain the conformational instability and aggregation nature of the mutant protein that results from incorrect folding and assembly.


Assuntos
Cadeia A de alfa-Cristalina/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Cromatografia por Troca Iônica , Reagentes de Ligações Cruzadas/química , Glutaratos/química , Humanos , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/isolamento & purificação , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/isolamento & purificação , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Estrutura Secundária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Succinimidas/química , Espectrometria de Massas em Tandem , Cadeia A de alfa-Cristalina/genética , Cadeia A de alfa-Cristalina/isolamento & purificação
17.
Biochemistry ; 52(21): 3638-50, 2013 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-23631441

RESUMO

Formation of protein aggregates in the aging eye lens has been shown to correlate with progressive accumulation of specific low-molecular weight (LMW) peptides derived from crystallins. Prominent among the LMW fragments is αA66-80, a peptide derived from αA-crystallin and present at higher concentrations in the water-insoluble nuclear fractions of the aging lens. The αA66-80 peptide has amyloid-like properties and preferentially insolubilizes α-crystallin from soluble lens fractions. However, the specific interactions and mechanisms by which the peptide induces α-crystallin aggregation have not been delineated. To gain insight into the mechanisms of peptide-induced aggregation, we investigated the interactions of the peptide with α-crystallin by various biochemical approaches. The peptide weakens α-crystallin chaperone ability and drastically promotes α-crystallin aggregation via the formation of insoluble peptide-protein complexes through transient intermediates. 4,4'-Dianilino-1,1'-binaphthyl-5,5'-disulfonic acid studies suggest that the peptide induces changes in the hydrophobicity of α-crystallin that could trigger the formation and growth of aggregates. The peptide-α-crystallin aggregates were found to be resistant to dissociation by high ionic strengths, whereas guanidinium hydrochloride and urea were effective dissociating agents. We conclude that the αA66-80 peptide forms a hydrophobically driven, stable complex with α-crystallin and reduces its solubility. Using isotope-labeled chemical cross-linking and mass spectrometry, we show that the peptide binds to multiple sites, including the chaperone site, the C-terminal extension, and subunit interaction sites in αB-crystallin, which may explain the antichaperone property of the peptide and the consequential age-related accumulation of aggregated proteins. Thus, the α-crystallin-derived peptide could play a role in the pathogenesis of cataract formation in the aging lens.


Assuntos
Envelhecimento/metabolismo , Catarata/metabolismo , Fragmentos de Peptídeos/metabolismo , alfa-Cristalinas/metabolismo , Sequência de Aminoácidos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Ligação Proteica , alfa-Cristalinas/química
18.
J Biol Chem ; 288(18): 13022-35, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23508955

RESUMO

α-Crystallin is a member of the small heat-shock protein (sHSP) family and consists of two subunits, αA and αB. Both αA- and αB-crystallin act as chaperones and anti-apoptotic proteins. Previous studies have identified the peptide (70)KFVIFLDVKHFSPEDLTVK(88) in αA-crystallin and the peptide (73)DRFSVNLDVKHFSPEELKVK(92) in αB-crystallin as mini-chaperones. In the human lens, lysine 70 (Lys(70)) of αA and Lys(92) of αB (in the mini-chaperone sequences) are acetylated. In this study, we investigated the cellular effects of the unmodified and acetyl mini-chaperones. The αA- and αB-crystallin peptides inhibited stress-induced aggregation of four client proteins, and the αA-acetyl peptide was more effective than the native peptide against three of the client proteins. Both the acetyl and native crystallin peptides inhibited stress-induced apoptosis in two mammalian cell types, and this property was directly related to the inhibition of cytochrome c release from mitochondria and the activity of caspase-3 and -9. In organ-cultured rat lenses, the peptides inhibited calcimycin-induced epithelial cell apoptosis. Intraperitoneal injection of the peptides inhibited cataract development in selenite-treated rats, which was accompanied by inhibition of oxidative stress, protein insolubilization, and caspase activity in the lens. These inhibitory effects were more pronounced for acetyl peptides than native peptides. A scrambled αA-crystallin peptide produced no such effects. The results suggest that the α-crystallin chaperone peptides could be used as therapeutic agents to treat cataracts and diseases in which protein aggregation and apoptosis are contributing factors.


Assuntos
Apoptose , Catarata/metabolismo , Células Epiteliais/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Chaperonas Moleculares/metabolismo , Cadeia A de alfa-Cristalina/metabolismo , Cadeia B de alfa-Cristalina/metabolismo , Adulto , Animais , Células CHO , Caspase 3/genética , Caspase 3/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Catarata/genética , Catarata/patologia , Células Cultivadas , Cricetinae , Cricetulus , Citocromos c/genética , Citocromos c/metabolismo , Modelos Animais de Doenças , Células Epiteliais/patologia , Humanos , Proteínas Inibidoras de Apoptose/química , Proteínas Inibidoras de Apoptose/genética , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Ratos , Ratos Sprague-Dawley , Cadeia A de alfa-Cristalina/química , Cadeia A de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/química , Cadeia B de alfa-Cristalina/genética
19.
Exp Eye Res ; 109: 51-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23410823

RESUMO

Proteins of lens fiber cells are prone to accumulate extensive post-translational modifications because of very little protein turnover. Lens proteins are degraded via the lens proteolytic systems into peptides, which are subsequently hydrolyzed by downstream aminopeptidases. Inefficient degradation can lead to accumulation of protein fragments and subsequent aggregation. Previously we showed that αA-66-80 peptide and its truncated products accumulate in aging and cataract human lenses. These peptides interact with crystallins, causing crystallin aggregation and precipitation. N- and C-terminal-blocked peptides that have the cleavage sites to generate the αA-66-80 fragment were used to test lens extracts for sequence-specific proteases in lens extracts. An internally quenched fluorogenic peptide substrate containing the sequence-specific site for a lens protease to generate αA-66-80 peptide was designed, synthesized and used to characterize protease(s) that are capable of generating this peptide in bovine and human lenses. We show that proteases with the potential to generate αA-66-80 peptide are present in bovine and human lenses. We also show that the αA-66-80 peptides are resistant to hydrolysis by aminopeptidases present in the lenses and they can suppress the degradation of other peptides. Failure of complete hydrolysis of these peptides in vivo can lead to their accumulation in the lens and subsequent lens protein aggregation, which may ultimately lead to the formation of cataract.


Assuntos
Catarata/metabolismo , Cristalino/enzimologia , Peptídeo Hidrolases/metabolismo , Cadeia A de alfa-Cristalina/metabolismo , Adulto , Envelhecimento/metabolismo , Envelhecimento/patologia , Animais , Catarata/patologia , Bovinos , Fracionamento Celular , Bancos de Olhos , Humanos , Hidrólise , Cristalino/patologia , Pessoa de Meia-Idade , Fragmentos de Peptídeos/metabolismo , Especificidade por Substrato , Extratos de Tecidos/metabolismo , Adulto Jovem
20.
PLoS One ; 7(9): e44077, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22970163

RESUMO

BACKGROUND: A substitution mutation in human αA-crystallin (αAG98R) is associated with autosomal dominant cataract. The recombinant mutant αAG98R protein exhibits altered structure, substrate-dependent chaperone activity, impaired oligomer stability and aggregation on prolonged incubation at 37 °C. Our previous studies have shown that αA-crystallin-derived mini-chaperone (DFVIFLDVKHFSPEDLTVK) functions like a molecular chaperone by suppressing the aggregation of denaturing proteins. The present study was undertaken to determine the effect of αA-crystallin-derived mini-chaperone on the stability and chaperone activity of αAG98R-crystallin. METHODOLOGY/PRINCIPAL FINDINGS: Recombinant αAG98R was incubated in presence and absence of mini-chaperone and analyzed by chromatographic and spectrometric methods. Transmission electron microscope was used to examine the effect of mini-chaperone on the aggregation propensity of mutant protein. Mini-chaperone containing photoactive benzoylphenylalanine was used to confirm the interaction of mini-chaperone with αAG98R. The rescuing of chaperone activity in mutantα-crystallin (αAG98R) by mini-chaperone was confirmed by chaperone assays. We found that the addition of the mini-chaperone during incubation of αAG98R protected the mutant crystallin from forming larger aggregates that precipitate with time. The mini-chaperone-stabilized αAG98R displayed chaperone activity comparable to that of wild-type αA-crystallin. The complexes formed between mini-αA-αAG98R complex and ADH were more stable than the complexes formed between αAG98R and ADH. Western-blotting and mass spectrometry confirmed the binding of mini-chaperone to mutant crystallin. CONCLUSION/SIGNIFICANCE: These results demonstrate that mini-chaperone stabilizes the mutant αA-crystallin and modulates the chaperone activity of αAG98R. These findings aid in our understanding of how to design peptide chaperones that can be used to stabilize mutant αA-crystallins and preserve the chaperone function.


Assuntos
Catarata/genética , Proteínas Mutantes/metabolismo , Cadeia A de alfa-Cristalina/genética , Cadeia A de alfa-Cristalina/metabolismo , Sequência de Aminoácidos , Dicroísmo Circular , Eletroforese em Gel de Poliacrilamida , Humanos , Dados de Sequência Molecular , Peso Molecular , Proteínas Mutantes/química , Proteínas Mutantes/ultraestrutura , Ligação Proteica , Desnaturação Proteica , Estabilidade Proteica , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Reprodutibilidade dos Testes , Solubilidade , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Temperatura , Cadeia A de alfa-Cristalina/química , Cadeia A de alfa-Cristalina/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...