Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 252
Filtrar
1.
J Mol Biol ; 436(8): 168499, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38401625

RESUMO

Small heat shock proteins (sHSPs) are ATP-independent chaperones vital to cellular proteostasis, preventing protein aggregation events linked to various human diseases including cataract. The α-crystallins, αA-crystallin (αAc) and αB-crystallin (αBc), represent archetypal sHSPs that exhibit complex polydispersed oligomeric assemblies and rapid subunit exchange dynamics. Yet, our understanding of how this plasticity contributes to chaperone function remains poorly understood. Using biochemical and biophysical analyses combined with single-particle electron microscopy (EM), we examined structural changes in αAc, αBc and native heteromeric lens α-crystallins (αLc) in their apo-states and at varying degree of chaperone saturation leading to co-aggregation, using lysozyme and insulin as model clients. Quantitative single-particle analysis unveiled a continuous spectrum of oligomeric states formed during the co-aggregation process, marked by significant client-triggered expansion and quasi-ordered elongation of the sHSP oligomeric scaffold, whereby the native cage-like sHSP assembly displays a directional growth to accommodate saturating conditions of client sequestration. These structural modifications culminated in an apparent amorphous collapse of chaperone-client complexes, resulting in the creation of co-aggregates capable of scattering visible light. Intriguingly, these co-aggregates maintain internal morphological features of highly elongated sHSP oligomers with striking resemblance to polymeric α-crystallin species isolated from aged lens tissue. This mechanism appears consistent across αAc, αBc and αLc, albeit with varying degrees of susceptibility to client-induced co-aggregation. Importantly, our findings suggest that client-induced co-aggregation follows a distinctive mechanistic and quasi-ordered trajectory, distinct from a purely amorphous process. These insights reshape our understanding of the physiological and pathophysiological co-aggregation processes of α-crystallins, carrying potential implications for a pathway toward cataract formation.


Assuntos
Catarata , Cristalinas , Proteínas de Choque Térmico Pequenas , alfa-Cristalinas , Humanos , Idoso , alfa-Cristalinas/metabolismo , Chaperonas Moleculares/metabolismo , Cristalinas/metabolismo , Catarata/metabolismo
2.
J Chem Theory Comput ; 20(4): 1740-1752, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38078935

RESUMO

Cataracts, a major cause of global blindness, contribute significantly to the overall prevalence of blindness. The opacification of the lens, resulting in cataract formation, primarily occurs due to the aggregation of crystallin proteins within the eye lens. Despite the high concentration of these crystallins, they remarkably maintain the lens transparency and refractive index. α-Crystallins (α-crys), acting as chaperones, play a crucial role in preventing crystallin aggregation, although the exact molecular mechanism remains uncertain. In this study, we employed a combination of molecular docking, all-atom molecular dynamics simulations, and advanced free energy calculations to investigate the interaction between γD-crystallin (γD-crys), a major structural protein of the eye lens, and α-crystallin proteins. Our findings demonstrate that α-crys exhibits an enhanced affinity for the NTD2 and CTD4 regions of γD-crys. The NTD2 and CTD4 regions form the interface between the N-terminal domain (NTD) and the C-terminal domain (CTD) of the γD-crys protein. By binding to the interface region between the NTD and CTD of the protein, α-crys effectively inhibits the formation of domain-swapped aggregates and mitigates protein aggregation. Analysis of the Markov state models using molecular dynamics trajectories confirms that minimum free energy conformations correspond to the binding of the α-crystallin domain (ACD) of α-crys to NTD2 and CTD4 that form the interdomain interface.


Assuntos
Catarata , alfa-Cristalinas , gama-Cristalinas , Humanos , alfa-Cristalinas/metabolismo , gama-Cristalinas/química , Simulação de Acoplamento Molecular , Catarata/metabolismo , Cegueira
3.
Plant Cell ; 36(4): 1098-1118, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38092516

RESUMO

DNA methylation is an important epigenetic mark implicated in selective rRNA gene expression, but the DNA methylation readers and effectors remain largely unknown. Here, we report a protein complex that reads DNA methylation to regulate variant-specific 45S ribosomal RNA (rRNA) gene expression in Arabidopsis (Arabidopsis thaliana). The complex, consisting of METHYL-CpG-BINDING DOMAIN PROTEIN5 (MBD5), MBD6, ALPHA-CRYSTALLIN DOMAIN PROTEIN15.5 (ACD15.5), and ACD21.4, directly binds to 45S rDNA. While MBD5 and MBD6 function redundantly, ACD15.5 and ACD21.4 are indispensable for variant-specific rRNA gene expression. These 4 proteins undergo phase separation in vitro and in vivo and are interdependent for their phase separation. The α-crystallin domain of ACD15.5 and ACD21.4, which is essential for their function, enables phase separation of the complex, likely by mediating multivalent protein interactions. The effector MICRORCHIDIA6 directly interacts with ACD15.5 and ACD21.4, but not with MBD5 and MBD6, and is recruited to 45S rDNA by the MBD-ACD complex to regulate variant-specific 45S rRNA expression. Our study reveals a pathway in Arabidopsis through which certain 45S rRNA gene variants are silenced, while others are activated.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , alfa-Cristalinas , Arabidopsis/genética , Arabidopsis/metabolismo , Genes de RNAr , Metilação de DNA/genética , RNA Ribossômico/genética , RNA Ribossômico/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , DNA Ribossômico/genética , DNA Ribossômico/metabolismo , alfa-Cristalinas/genética , alfa-Cristalinas/metabolismo
4.
Naunyn Schmiedebergs Arch Pharmacol ; 397(1): 161-171, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37395794

RESUMO

Aldose reductase (ALR2) is a rate-limiting component of the polyol pathway, which is essential for the NADPH-mediated conversion from glucose to sorbitol. ALR2 dysregulation has been linked to α-crystallin aggregation, increased oxidative stress, and calcium inflow, all of which contribute to a diabetic cataract. Given its crucial role in occular pathologies, ALR2 has emerged as a promising target to treat oxidative stress and hyperglycaemic condition which form the underlying cause of diabetic cataracts. However, several of them had issues with sensitivity and specificity to ALR2, despite being screened as effective ALR2 inhibitors from a wide range of structurally varied molecules. The current study investigates the inhibitory potential of Nifedipine, an analog of the dihydro nicotinamide class of compounds against ALR2 activity. The enzyme inhibition studies were supported by in vitro biomolecular interactions, molecular modeling approaches, and in vivo validation in diabetic rat models. Nifedipine demonstrated appreciable inhibitory potential with the purified recombinant hAR (human aldose reductase; with an IC50 value of 2.5 µM), which was further supported by Nifedipine-hAR binding affinity (Kd = 2.91 ± 1.87 × 10-4 M) by ITC and fluorescence quenching assays. In the in vivo models of STZ-induced diabetic rats, Nifedipine delayed the onset progression of cataracts by preserving the antioxidant enzyme activity (SOD, CAT, and GPX GSH, TBARS, and protein carbonyls) and was shown to retain the α-crystallin chaperone activity by reducing the calcium levels in the diabetic rat lens. In conclusion, our results demonstrate effective inhibition of ALR2 by Nifedipine, resulting in amelioration of diabetic cataract conditions by lowering oxidative and osmotic stress while retaining the chaperone activity of α-crystallins. The present study could be envisaged to improve the eye condition in older adults upon Nifedipine treatment.


Assuntos
Catarata , Diabetes Mellitus Experimental , alfa-Cristalinas , Ratos , Humanos , Animais , Idoso , Nifedipino/farmacologia , Nifedipino/uso terapêutico , Diabetes Mellitus Experimental/tratamento farmacológico , Aldeído Redutase , Cálcio , Catarata/tratamento farmacológico , Catarata/prevenção & controle , Antioxidantes/uso terapêutico , Inibidores Enzimáticos/farmacologia , alfa-Cristalinas/metabolismo
5.
Biochimie ; 219: 146-154, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38016530

RESUMO

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.


Assuntos
Proteínas de Choque Térmico Pequenas , alfa-Cristalinas , Humanos , alfa-Cristalinas/metabolismo , Filaminas/metabolismo , Proteínas de Choque Térmico Pequenas/química , Proteínas de Choque Térmico Pequenas/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Domínios de Imunoglobulina
6.
Protein J ; 43(1): 39-47, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38017314

RESUMO

Therapeutic proteins are potent, fast-acting drugs that are highly effective in treating various conditions. Medicinal protein usage has increased in the past 10 years, and it will evolve further as we better understand disease molecular pathways. However, it is associated with high processing costs, limited stability, difficulty in being administered as an oral medication, and the inability of large proteins to penetrate tissue and reach their target locations. Many methods have been developed to overcome the problems with the stability and chaperone activity of therapeutic proteins, viz., the addition of external agents (changing the properties of the surrounding solvent by using stabilizing excipients, e.g., amino acids, sugars, polyols) and internal agents (chemical modifications that influence its structural properties, e.g., mutations, glycosylation). However, these methods must completely clear protein instability and chaperone issues. There is still much work to be done on finetuning chaperone proteins to increase their biological efficacy and stability. Methylglyoxal (MGO), a potent dicarbonyl compound, reacts with proteins and forms covalent cross-links. Much research on MGO scavengers has been conducted since they are known to alter protein structure, which may result in alterations in biological activity and stability. MGO is naturally produced within our body, however, its impact on chaperones and protein stability needs to be better understood and seems to vary based on concentration. This review highlights the efforts of several research groups on the effect of MGO on various proteins. It also addresses the impact of MGO on a client protein, α-crystallin, to understand the potential solutions to the protein's chaperone and stability problems.


Assuntos
Aldeído Pirúvico , alfa-Cristalinas , Humanos , Aldeído Pirúvico/química , Aldeído Pirúvico/farmacologia , Óxido de Magnésio , alfa-Cristalinas/química , alfa-Cristalinas/metabolismo , Chaperonas Moleculares/química , Dobramento de Proteína
7.
J Biol Chem ; 299(9): 105108, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37517700

RESUMO

Bacterial small heat shock proteins, such as inclusion body-associated protein A (IbpA) and IbpB, coaggregate with denatured proteins and recruit other chaperones for the processing of aggregates thereby assisting in protein refolding. In addition, as a recently revealed uncommon feature, Escherichia coli IbpA self-represses its own translation through interaction with the 5'-untranslated region of the ibpA mRNA, enabling IbpA to act as a mediator of negative feedback regulation. Although IbpA also suppresses the expression of IbpB, IbpB does not have this self-repression activity despite the two Ibps being highly homologous. In this study, we demonstrate that the self-repression function of IbpA is conserved in other γ-proteobacterial IbpAs. Moreover, we show a cationic residue-rich region in the α-crystallin domain of IbpA, which is not conserved in IbpB, is critical for the self-suppression activity. Notably, we found arginine 93 (R93) located within the α-crystallin domain is an essential residue that cannot be replaced by any of the other 19 amino acids including lysine. We observed that IbpA-R93 mutants completely lost the interaction with the 5' untranslated region of the ibpA mRNA, but retained almost all chaperone activity and were able to sequester denatured proteins. Taken together, we propose the conserved Arg93-mediated translational control of IbpA through RNA binding would be beneficial for a rapid and massive supply of the chaperone on demand.


Assuntos
Arginina , Gammaproteobacteria , Proteínas de Choque Térmico Pequenas , RNA Mensageiro , Regiões 5' não Traduzidas/genética , alfa-Cristalinas/metabolismo , Arginina/metabolismo , Sequência Conservada , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Gammaproteobacteria/metabolismo , Proteínas de Choque Térmico Pequenas/química , Proteínas de Choque Térmico Pequenas/genética , Proteínas de Choque Térmico Pequenas/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutação , Biossíntese de Proteínas , Desnaturação Proteica , Domínios Proteicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
8.
Int J Biol Macromol ; 242(Pt 1): 124590, 2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-37116845

RESUMO

α-crystallin is a structurally essential small heat shock protein (sHSP) with a chaperone-like activity which maintains transparency of the lenticular tissues during a period of time that is as long as human life. α-crystallin is a multimeric protein consisting of αA and αB subunits, with 57 % homology. The CRYAB gene on chromosome 11 encodes human αB-crystallin (αB-Cry), which contains 175 amino acid residues. In the current study, the cataractogenic mutations R12C, P20R, R69C, and double mutations R12C/P20R and R12C/P20R were embedded into the human CRYAB gene. Following successful expression in the prokaryotic system and purification, a number of spectroscopic techniques, gel electrophoresis, dynamic light scattering (DLS), and transmission electron microscopy (TEM) were applied to assess the role of these mutations on the structure, amyloidogenicity, and biological function of human αB-Cry. The created mutations caused significant changes in the structure, and oligomeric state of human αB-Cry. These mutations, particularly R12C, R12C/P20R, and R12C/R69C, dramatically enhanced the tendency of this protein for the amyloid fibril formation and reduced its chaperone-like activity. Since double mutations R12C/P20R and R12C/P20R were able to intensely change the protein's structure and chaperone function, it can be suggested that they may play a destructive role in a cumulative manner. Our findings indicated that the simultaneous presence of two pathogenic mutations may have a cumulative destructive impacts on the structure and function of human αB-Cry and this observation is likely related to the disease severity of the mutated proteins.


Assuntos
Catarata , alfa-Cristalinas , Humanos , Catarata/genética , Cadeia B de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/química , Mutação , Dobramento de Proteína , alfa-Cristalinas/metabolismo
9.
ACS Chem Neurosci ; 14(9): 1659-1671, 2023 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-37057966

RESUMO

Altered protein folding leading to the formation of structured aggregates such as amyloid fibrils has gained significant attention due to its association with neurodegenerative diseases. α-Synuclein, a small intrinsically disordered protein, gets transformed into amyloid fibrils under unfavorable conditions and contributes to the progression and pathology of Parkinson's disease (PD). Under normal physiological conditions, amyloid formation is controlled by many chaperones and chaperone-like proteins. However, with aging, the protein homeostasis machinery becomes less efficient, causing the loss of proper functioning of chaperones and leading to aberrant protein folding and amyloid formation. Here, we provide in-depth information on the modulation of α-synuclein amyloid assembly by a heterogeneous complex of bovine eye lens protein, α-crystallin, which is known to possess chaperone-like activity. We have used a multiparametric approach to discern the critical events through which α-crystallin abolishes α-synuclein amyloid formation. Our biochemical and biophysical data analysis revealed that α-crystallin, at substoichiometric ratios, alleviates α-synuclein amyloid assembly and drives it into soluble dead-end intermediates. We also demonstrated that α-crystallin was equally efficient in arresting amyloid assembly by some of the PD-related mutants suggesting the significance of chaperone-like activity of α-crystallin under pathological conditions. Finally, we validated our results using human crystallin derived from cataract patients. Based on our findings, we propose that the interaction of α-crystallin directs α-synuclein into a soluble amyloid-incompetent form. Our results suggest that the generic antiamyloid property of chaperone-like proteins, such as α-crystallin, can be harnessed to design protein and peptide-based novel therapeutics for prevention and treatment of deadly neurodegenerative diseases.


Assuntos
Doença de Parkinson , alfa-Cristalinas , Humanos , Animais , Bovinos , alfa-Cristalinas/metabolismo , alfa-Sinucleína/metabolismo , Amiloide/metabolismo , Chaperonas Moleculares/metabolismo , Dobramento de Proteína , Proteínas Amiloidogênicas , Doença de Parkinson/metabolismo
10.
Sci Rep ; 13(1): 4832, 2023 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-36964267

RESUMO

Cataract, the leading cause of blindness worldwide, is caused by crystallin protein aggregation within the protected lens environment. Phase separation has been implicated as an important mechanism of protein aggregation diseases, such as neurodegeneration. Similarly, cataract has been proposed to be a protein condensation disease in the last century. However, whether crystallin proteins aggregate via a phase separation mechanism and which crystallin protein initiates the aggregation remain unclear. Here, we showed that all types of crystallin-GFP proteins remain soluble under physiological conditions, including protein concentrations, ion strength, and crowding environments. However, in age or disease-induced aberrant conditions, α-crystallin-GFP, including αA- and αB-crystallin-GFP, but not other crystallin-GFP proteins, undergo phase separation in vivo and in vitro. We found that aging-related changes, including higher crystallin concentrations, increased Na+, and decreased K+ concentrations, induced the aggregation of α-crystallin-GFP. Furthermore, H2O2, glucose, and sorbitol, the well-known risk factors for cataract, significantly enhanced the aggregation of αB-crystallin-GFP. Taken together, our results revealed that α-crystallin-GFP forms aggregates via a phase transition process, which may play roles in cataract disease. Opposite to the previously reported function of enhancing the solubility of other crystallin, α-crystallin may be the major aggregated crystallin in the lens of cataract patients.


Assuntos
Catarata , Cristalinas , Cristalino , Cadeia A de alfa-Cristalina , alfa-Cristalinas , Humanos , alfa-Cristalinas/metabolismo , Cristalinas/genética , Cristalinas/metabolismo , Agregados Proteicos , Peróxido de Hidrogênio/metabolismo , Catarata/metabolismo , Cristalino/metabolismo
11.
J Fluoresc ; 33(4): 1347-1358, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36648626

RESUMO

The aggregation of lens proteins induced by glycation is one of the key drivers of diabetic retinopathy and development of diabetic cataracts. Moreover, glycation also causes numerous alterations not only to the tertiary structure of lens proteins but also to serum proteins. There are also evidences of covalent crosslinking among lens crystallins resulting in development of cataract. In this article, the inhibitory potential of butein was tested against the glucose induced glycation and the aggregation α-crystallin (α-cry). The results showed that there was inhibition of advanced glycation products (78.28%) and early glycation products (86.30%) following the treatment of butein. Additionally, the presence of butein caused a significant improvement in the tested biochemical markers of glycation. The treatment with butein reduced the free lysine modification to 23.67%. The secondary and tertiary structural distortions of α-cry were also protected. The mechanism of inhibition further investigated at the molecular level using biophysical and computational techniques. The interaction data showed the butein exhibited strong affinity towards the α-cry. The binding event was entropically driven and energetically favourable. The Gibb's free energy of the interaction was found to be -5.99 to -7.17 kcal mol-1. The binding site of butein in α-cry was deciphered by molecular docking and the dynamics was studied using molecular dynamics (MD) simulations. The simulation data showed that butein formed stable complex with α-cry under physiological conditions. Most of the tested parameters from molecular simulations, such as secondary structure, was found to be stable. The data clearly show the potential of butein in inhibiting the glycation induced aggregation of α-cry and hence can be developed as useful inhibitor in the management of diabetic cataract and retinopathy.


Assuntos
Catarata , Cristalinas , Diabetes Mellitus , Doenças Retinianas , alfa-Cristalinas , Humanos , alfa-Cristalinas/química , alfa-Cristalinas/metabolismo , Reação de Maillard , Simulação de Acoplamento Molecular , Glicosilação , Cristalinas/química , Cristalinas/metabolismo , Catarata/etiologia , Catarata/metabolismo , Catarata/prevenção & controle , Doenças Retinianas/complicações , Produtos Finais de Glicação Avançada/metabolismo
12.
Proc Natl Acad Sci U S A ; 120(6): e2213765120, 2023 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-36719917

RESUMO

Small heat-shock proteins (sHSPs) are a widely expressed family of ATP-independent molecular chaperones that are among the first responders to cellular stress. Mechanisms by which sHSPs delay aggregation of client proteins remain undefined. sHSPs have high intrinsic disorder content of up to ~60% and assemble into large, polydisperse homo- and hetero-oligomers, making them challenging structural and biochemical targets. Two sHSPs, HSPB4 and HSPB5, are present at millimolar concentrations in eye lens, where they are responsible for maintaining lens transparency over the lifetime of an organism. Together, HSPB4 and HSPB5 compose the hetero-oligomeric chaperone known as α-crystallin. To identify the determinants of sHSP function, we compared the effectiveness of HSPB4 and HSPB5 homo-oligomers and HSPB4/HSPB5 hetero-oligomers in delaying the aggregation of the lens protein γD-crystallin. In chimeric versions of HSPB4 and HSPB5, chaperone activity tracked with the identity of the 60-residue disordered N-terminal regions (NTR). A short 10-residue stretch in the middle of the NTR ("Critical sequence") contains three residues that are responsible for high HSPB5 chaperone activity toward γD-crystallin. These residues affect structure and dynamics throughout the NTR. Abundant interactions involving the NTR Critical sequence reveal it to be a hub for a network of interactions within oligomers. We propose a model whereby the NTR critical sequence influences local structure and NTR dynamics that modulate accessibility of the NTR, which in turn modulates chaperone activity.


Assuntos
Proteínas de Choque Térmico Pequenas , Cristalino , alfa-Cristalinas , Humanos , alfa-Cristalinas/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Choque Térmico Pequenas/metabolismo , Cadeia B de alfa-Cristalina/metabolismo , Cristalino/metabolismo
13.
Exp Eye Res ; 227: 109358, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36572168

RESUMO

The α-crystallin small heat shock proteins contribute to the transparency and refractive properties of the vertebrate eye lens and prevent the protein aggregation that would otherwise produce lens cataracts, the leading cause of human blindness. There are conflicting data in the literature as to what role the α-crystallins may play in early lens development. In this study, we used CRISPR gene editing to produce zebrafish lines with mutations in each of the three α-crystallin genes (cryaa, cryaba and cryabb) to prevent protein production. The absence of each α-crystallin protein was analyzed by mass spectrometry, and lens phenotypes were assessed with differential interference contrast microscopy and histology. Loss of αA-crystallin produced a variety of lens defects with varying severity in larvae at 3 and 4 dpf but little substantial change in normal fiber cell denucleation. Loss of αBa-crystallin produced no substantial lens defects. Our cryabb mutant produced a truncated αBb-crystallin protein and showed no substantial change in lens development. Mutation of each α-crystallin gene did not alter the mRNA levels of the remaining two, suggesting a lack of genetic compensation. These data suggest that αA-crystallin plays some role in lens development, but the range of phenotype severity in null mutants indicates its loss simply increases the chance for defects and that the protein is not essential. Our finding that cryaba and cryabb mutants lack noticeable lens defects is congruent with insubstantial transcript levels for these genes in lens epithelial and fiber cells through five days of development. Future experiments can explore the molecular mechanisms leading to lens defects in cryaa null mutants and the impact of αA-crystallin loss during zebrafish lens aging.


Assuntos
Catarata , Cristalinas , Cristalino , Cadeia A de alfa-Cristalina , alfa-Cristalinas , Animais , Humanos , Peixe-Zebra , alfa-Cristalinas/genética , alfa-Cristalinas/metabolismo , Cristalinas/genética , Cristalinas/metabolismo , Cadeia A de alfa-Cristalina/metabolismo , Cristalino/metabolismo , Proteínas/metabolismo , Catarata/metabolismo
14.
PLoS Negl Trop Dis ; 16(9): e0010766, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36112664

RESUMO

Fasciola hepatica is a trematode parasite that infects animals and humans causing fasciolosis, a worldwide-distributed disease responsible for important economic losses and health problems. This disease is of growing public health concern since parasite isolates resistant to the current treatment (triclabendazole) have increasingly been described. F. hepatica infects its vertebrate host after ingestion of the encysted parasite (metacercariae), which are found in the water or attached to plants. Upon ingestion, newly excysted juveniles of F. hepatica (FhNEJ) emerge in the intestinal lumen and cross the intestinal barrier, reach the peritoneum and migrate to the biliary ducts, where adult worms fully develop. Despite the efforts made to develop new therapeutic and preventive tools, to date, protection against F. hepatica obtained in different animal models is far from optimal. Early events of host-FhNEJ interactions are of paramount importance for the infection progress in fasciolosis, especially those occurring at the host-parasite interface. Nevertheless, studies of FhNEJ responses to the changing host environment encountered during migration across host tissues are still scarce. Here, we set-up an ex vivo model coupled with quantitative SWATH-MS proteomics to study early host-parasite interaction events in fasciolosis. After comparing tegument and somatic fractions from control parasites and FhNEJ that managed to cross a mouse intestinal section ex vivo, a set of parasite proteins whose expression was statistically different were found. These included upregulation of cathepsins L3 and L4, proteolytic inhibitor Fh serpin 2, and a number of molecules linked with nutrient uptake and metabolism, including histone H4, H2A and H2B, low density lipoprotein receptor, tetraspanin, fatty acid binding protein a and glutathione-S-transferase. Downregulated proteins in FhNEJ after gut passage were more numerous than the upregulated ones, and included the heath shock proteins HSP90 and alpha crystallin, amongst others. This study brings new insights into early host-parasite interactions in fasciolosis and sheds light on the proteomic changes in FhNEJ triggered upon excystment and intestinal wall crossing, which could serve to define new targets for the prevention and treatment of this widespread parasitic disease.


Assuntos
Fasciola hepatica , Fasciolíase , alfa-Cristalinas , Animais , Catepsinas , Fasciola hepatica/metabolismo , Fasciolíase/parasitologia , Proteínas de Ligação a Ácido Graxo , Glutationa/metabolismo , Proteínas de Helminto/metabolismo , Histonas/metabolismo , Humanos , Camundongos , Proteômica , Receptores de LDL/metabolismo , Transferases/metabolismo , Triclabendazol , alfa-Cristalinas/metabolismo
15.
Allergol. immunopatol ; 50(5): 23-29, sept. 2022. graf
Artigo em Inglês | IBECS | ID: ibc-208623

RESUMO

Background: Chronic obstructive pulmonary disease (COPD) is a familiar airway disease characterized by chronic immune response in the lungs. More and more evidences have assured that cigarette smoking is the primary reason for the progression of COPD, but its related regulatory mechanism requires further clarification. The α-B-crystallin (CRYAB) has been identified to exhibit vital functions in different diseases, and is down-regulated in the alveoli of mice mediated by cigarette smoke extract (CSE).Methods: The messenger RNA expression of CRYAB was assessed by reverse transcription--quantitative polymerase chain reaction. The proteins’ expressions were tested using Western blot method. The cytotoxicity was measured by lactate dehydrogenase assay. The levels of malondialdehyde, superoxide dismutase, catalase, myeloperoxidase, tumor necrosis factor-α, interleukin (IL)-1β, and IL-6 were assessed through enzyme-linked-immunosorbent serologic assay (ELISA).Results: In this study, it was discovered that the expression of CRYAB was markedly decreased with the increased time of cigarette smoking. Moreover, CRYAB overexpression increased cell viability and decreased cell apoptosis induced by cigarette smoke. In addition, the strengthened oxidative stress and inflammation mediated by CSE treatment was relieved after overexpression of CRYAB. Eventually, results OF Western blot method confirmed that CRYAB retarded the activation of phosphatidylinositol 3-kinase–Ak strain transforming (PI3K–Akt) and nuclear factor kappa B (NF-κB) signaling pathways.Conclusion: Our results manifested that CRYAB reduced cigarette smoke-induced inflammation, apoptosis, and oxidative stress in normal and diseased bronchial epithelial (NHBE) and human bronchial epithelial (BEAS-2B) cells by suppressing PI3K/Akt and NF-κB signaling pathways, which highlighted the functioning of CRYAB in preventing or treating COPD (AU)


Assuntos
Humanos , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Doença Pulmonar Obstrutiva Crônica , Fumar Cigarros/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Proteína Oncogênica v-akt/metabolismo , Transdução de Sinais , alfa-Cristalinas , alfa-Cristalinas/metabolismo , Apoptose , Estresse Oxidativo , Inflamação , Células Cultivadas
16.
J Biol Chem ; 298(10): 102417, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36037967

RESUMO

Γ-Crystallins play a major role in age-related lens transparency. Their destabilization by mutations and physical chemical insults are associated with cataract formation. Therefore, drugs that increase their stability should have anticataract properties. To this end, we screened 2560 Federal Drug Agency-approved drugs and natural compounds for their ability to suppress or worsen H2O2 and/or heat-mediated aggregation of bovine γ-crystallins. The top two drugs, closantel (C), an antihelminthic drug, and gambogic acid (G), a xanthonoid, attenuated thermal-induced protein unfolding and aggregation as shown by turbidimetry fluorescence spectroscopy dynamic light scattering and electron microscopy of human or mouse recombinant crystallins. Furthermore, binding studies using fluorescence inhibition and hydrophobic pocket-binding molecule bis-8-anilino-1-naphthalene sulfonic acid revealed static binding of C and G to hydrophobic sites with medium-to-low affinity. Molecular docking to HγD and other γ-crystallins revealed two binding sites, one in the "NC pocket" (residues 50-150) of HγD and one spanning the "NC tail" (residues 56-61 to 168-174 in the C-terminal domain). Multiple binding sites overlap with those of the protective mini αA-crystallin chaperone MAC peptide. Mechanistic studies using bis-8-anilino-1-naphthalene sulfonic acid as a proxy drug showed that it bound to MAC sites, improved Tm of both H2O2 oxidized and native human gamma D, and suppressed turbidity of oxidized HγD, most likely by trapping exposed hydrophobic sites. The extent to which these drugs act as α-crystallin mimetics and reduce cataract progression remains to be demonstrated. This study provides initial insights into binding properties of C and G to γ-crystallins.


Assuntos
Materiais Biomiméticos , Catarata , Cristalino , Chaperonas Moleculares , Agregação Patológica de Proteínas , Salicilanilidas , Xantonas , alfa-Cristalinas , gama-Cristalinas , Animais , Bovinos , Humanos , Camundongos , alfa-Cristalinas/metabolismo , Catarata/tratamento farmacológico , Catarata/prevenção & controle , Catarata/genética , gama-Cristalinas/metabolismo , Peróxido de Hidrogênio/metabolismo , Cristalino/metabolismo , Chaperonas Moleculares/metabolismo , Simulação de Acoplamento Molecular , Naftalenos/metabolismo , Ácidos Sulfônicos/metabolismo , Salicilanilidas/química , Salicilanilidas/farmacologia , Salicilanilidas/uso terapêutico , Xantonas/química , Xantonas/farmacologia , Xantonas/uso terapêutico , Agregação Patológica de Proteínas/tratamento farmacológico , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Materiais Biomiméticos/uso terapêutico
17.
Biochemistry (Mosc) ; 87(2): 106-120, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35508906

RESUMO

The absence of cellular organelles in fiber cells and very high cytoplasmic protein concentration (up to 900 mg/ml) minimize light scattering in the lens and ensure its transparency. Low oxygen concentration, powerful defense systems (antioxidants, antioxidant enzymes, chaperone-like protein alpha-crystallin, etc.) maintain lens transparency. On the other hand, the ability of crystallins to accumulate age-associated post-translational modifications, which reduce the resistance of lens proteins to oxidative stress, is an important factor contributing to the cataract formation. Here, we suggest a mechanism of cataractogenesis common for the action of different cataractogenic factors, such as age, radiation, ultraviolet light, diabetes, etc. Exposure to these factors leads to the damage and death of lens epithelium, which allows oxygen to penetrate into the lens through the gaps in the epithelial layer and cause oxidative damage to crystallins, resulting in protein denaturation, aggregation, and formation of multilamellar bodies (the main cause of lens opacification). The review discusses various approaches to the inhibition of lens opacification (cataract development), in particular, a combined use of antioxidants and compounds enhancing the chaperone-like properties of alpha-crystallin. We also discuss the paradox of high efficiency of anti-cataract drugs in laboratory settings with the lack of their clinical effect, which might be due to the late use of the drugs at the stage, when the opacification has already formed. A probable solution to this situation will be development of new diagnostic methods that will allow to predict the emergence of cataract long before the manifestation of its clinical signs and to start early preventive treatment.


Assuntos
Catarata , Cristalinas , Cristalino , alfa-Cristalinas , Antioxidantes/metabolismo , Catarata/etiologia , Cristalinas/análise , Cristalinas/metabolismo , Humanos , Cristalino/metabolismo , Chaperonas Moleculares/metabolismo , Oxigênio/metabolismo , alfa-Cristalinas/análise , alfa-Cristalinas/química , alfa-Cristalinas/metabolismo
18.
Biochemistry (Mosc) ; 87(2): 131-140, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35508910

RESUMO

Loss of eye lens transparency due to cataract is the leading cause of blindness all over the world. While aggregation of lens crystallins is the most common endpoint in various types of cataracts, chaperone-like activity (CLA) of α-crystallin preventing protein aggregation is considered to be important for maintaining the eye lens transparency. Osmotic stress due to increased accumulation of sorbitol under hyperglycemic conditions is believed to be one of the mechanisms for diabetic cataract. In addition, compromised CLA of α-crystallin in diabetic cataract has been reported. However, the effect of sorbitol on the structure and function of α-crystallin has not been elucidated yet. Hence, in the present exploratory study, we described the effect of varying concentrations of sorbitol on the structure and function of α-crystallin. Alpha-crystallin purified from the rat lens was incubated with varying concentrations of sorbitol in the dark under sterile conditions for up to 5 days. At the end of incubation, structural properties and CLA were evaluated by spectroscopic methods. Interestingly, different concentrations of sorbitol showed contrasting results: at lower concentrations (5 and 50 mM) there was a decrease in CLA and subtle alterations in secondary and tertiary structure but not at higher concentrations (500 mM). Though, these results shed a light on the effect of sorbitol on α-crystallin structure-function, further studies are required to understand the mechanism of the observed effects and their implication to cataractogenesis.


Assuntos
Catarata , Diabetes Mellitus , Cristalino , alfa-Cristalinas , Animais , Cristalino/metabolismo , Chaperonas Moleculares/metabolismo , Ratos , Sorbitol/farmacologia , alfa-Cristalinas/química , alfa-Cristalinas/metabolismo , alfa-Cristalinas/farmacologia
19.
Biochemistry (Mosc) ; 87(3): 225-241, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35526854

RESUMO

* The article is published as a part of the Special Issue "Protein Misfolding and Aggregation in Cataract Disorders" (Vol. 87, No. 2). ** To whom correspondence should be addressed. Cataract is a major cause of blindness. Due to the lack of protein turnover, lens proteins accumulate age-related and environmental modifications that alter their native conformation, leading to the formation of aggregation-prone intermediates, as well as insoluble and light-scattering aggregates, thus compromising lens transparency. The lens protein, α-crystallin, is a molecular chaperone that prevents protein aggregation, thereby maintaining lens transparency. However, mutations or post-translational modifications, such as oxidation, deamidation, truncation and crosslinking, can render α-crystallins ineffective and lead to the disease exacerbation. Here, we describe such mutations and alterations, as well as their consequences. Age-related modifications in α-crystallins affect their structure, oligomerization, and chaperone function. Mutations in α-crystallins can lead to the aggregation/intracellular inclusions attributable to the perturbation of structure and oligomeric assembly and resulting in the rearrangement of aggregation-prone regions. Such rearrangements can lead to the exposure of hitherto buried aggregation-prone regions, thereby populating aggregation-prone state(s) and facilitating amorphous/amyloid aggregation and/or inappropriate interactions with cellular components. Investigations of the mutation-induced changes in the structure, oligomer assembly, aggregation mechanisms, and interactomes of α-crystallins will be useful in fighting protein aggregation-related diseases.


Assuntos
Catarata , Cristalino , alfa-Cristalinas , Catarata/genética , Humanos , Cristalino/metabolismo , Chaperonas Moleculares/metabolismo , Mutação , Agregados Proteicos , alfa-Cristalinas/química , alfa-Cristalinas/genética , alfa-Cristalinas/metabolismo
20.
Biophys J ; 121(12): 2233-2250, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35619565

RESUMO

Exposure to solar UV irradiation damages γ-crystallin, leading to cataract formation via aggregation. α-Crystallin, as a small heat shock protein, efficiently suppresses this irreversible aggregation by selectively binding the denatured γ-crystallin monomer. In this study, liquid chromatography tandem mass spectrometry was used to evaluate UV-325 nm irradiation-induced photodamage of human γD-crystallin in the presence of bovine α-crystallin, atomic force microscope (AFM) and dynamic light scattering (DLS) techniques were used to detect the quaternary structure changes of the α-crystallin oligomer, and Fourier transform infrared spectroscopy and temperature-jump nanosecond time-resolved IR absorbance difference spectroscopy were used to probe the secondary structure changes of bovine α-crystallin. We find that the thermal-induced subunit dissociation of the α-crystallin oligomer involves the breaking of hydrogen bonds at the dimeric interface, leading to three different spectral components at varied temperature regions as resolved from temperature-dependent IR spectra. Under UV-325 nm irradiation, unfolded γD-crystallin binds to the dissociated α-crystallin subunit to form an αγ-complex, then follows the reassociation of the αγ-complex to the partially dissociated α-crystallin oligomer. This prevents the aggregation of denatured γD-crystallin. The formation of the γD-bound α-crystallin oligomer is further confirmed by AFM and DLS analysis, which reveals an obvious size expansion in the reassociated αγ-oligomers. In addition, UV-325 nm irradiation causes a peptide bond cleavage of γD-crystallin at Ala158 in the presence of α-crystallin. Our results suggest a very effective protection mechanism for subunits dissociated from α-crystallin oligomers against UV irradiation-induced aggregation of γD-crystallin, at the expense of a loss of a short C-terminal peptide in γD-crystallin.


Assuntos
alfa-Cristalinas , gama-Cristalinas , Animais , Bovinos , Humanos , Chaperonas Moleculares/metabolismo , Dobramento de Proteína , Estrutura Secundária de Proteína , alfa-Cristalinas/metabolismo , gama-Cristalinas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...