Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Invest Ophthalmol Vis Sci ; 63(5): 15, 2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35575904

RESUMO

Purpose: To investigate how cataract-linked mutations affect the gradient refractive index (GRIN) and lens opacification in mouse lenses and whether there is any effect on the optics of the lens from treatment with an oxysterol compound. Methods: A total of 35 mice including wild-type and knock-in mutants (Cryaa-R49C and Cryab-R120G) were used in these experiments: 26 mice were treated with topical VP1-001, an oxysterol, in one eye and vehicle in the other, and nine mice were untreated controls. Slit lamp biomicroscopy was used to analyze the lens in live animals and to provide apparent cataract grades. Refractive index in the lenses of 64 unfixed whole mouse eyes was calculated from measurements with X-ray phase tomography based on X-ray Talbot interferometry with a synchrotron radiation source. Results: Heterozygous Cryaa-R49C lenses had slightly irregularly shaped contours in the center of the GRIN and distinct disturbances of the gradient index at the anterior and posterior poles. Contours near the lens surface were denser in homozygous Cryab-R120G lenses. Treatment with topical VP1-001, an oxysterol, showed an improvement in refractive index profiles in 61% of lenses and this was supported by a reduction in apparent lens opacity grade by 1.0 in 46% of live mice. Conclusions: These results indicate that α-crystallin mutations alter the refractive index gradient of mouse lenses in distinct ways and suggest that topical treatment with VP1-001 may improve lens transparency and refractive index contours in some lenses with mutations.


Assuntos
Catarata , Cristalinas , Cristalino , Oxisteróis , Animais , Catarata/genética , Cristalinas/genética , Modelos Animais de Doenças , Cristalino/metabolismo , Cristalino/fisiologia , Camundongos , Oxisteróis/farmacologia
2.
PLoS One ; 16(9): e0257098, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34520490

RESUMO

αB-crystallin is a small heat shock protein that forms a heterooligomeric complex with αA-crystallin in the ocular lens. It is also widely distributed in tissues throughout the body and has been linked with neurodegenerative diseases such as Alzheimer's, where it is associated with amyloid fibrils. Crystallins can form amorphous aggregates in cataracts as well as more structured amyloid-like fibrils. The arginine 120 to glycine (R120G) mutation in αB-crystallin (Cryab-R120G) results in high molecular weight crystallin protein aggregates and loss of the chaperone activity of the protein in vitro, and it is associated with human hereditary cataracts and myopathy. Characterizing the amorphous (unstructured) versus the highly ordered (amyloid fibril) nature of crystallin aggregates is important in understanding their role in disease and important to developing pharmacological treatments for cataracts. We investigated protein secondary structure in wild-type (WT) and Cryab-R120G knock-in mutant mouse lenses using two-dimensional infrared (2DIR) spectroscopy, which has been used to detect amyloid-like fibrils in human lenses and measure UV radiation-induced changes in porcine lenses. Our goal was to compare the aggregated proteins in this mouse lens model to human lenses and evaluate the protein structural relevance of the Cryab-R120G knock-in mouse model to general age-related cataract disease. In the 2DIR spectra, amide I diagonal peak frequencies were red-shifted to smaller wavenumbers in mutant mouse lenses as compared to WT mouse lenses, consistent with an increase in ordered secondary structure. The cross peak frequency and intensity indicated the presence of amyloid in the mutant mouse lenses. While the diagonal and cross peak changes in location and intensity from the 2DIR spectra indicated significant structural differences between the wild type and mutant mouse lenses, these differences were smaller than those found in human lenses; thus, the Cryab-R120G knock-in mouse lenses contain less amyloid-like secondary structure than human lenses. The results of the 2DIR spectroscopy study confirm the presence of amyloid-like secondary structure in Cryab-R120G knock-in mice with cataracts and support the use of this model to study age-related cataract.


Assuntos
Amiloide/metabolismo , Catarata/genética , Técnicas de Introdução de Genes , Espectrofotometria Infravermelho , Cadeia B de alfa-Cristalina/química , Cadeia B de alfa-Cristalina/genética , Animais , Modelos Animais de Doenças , Formaldeído , Humanos , Cristalino/metabolismo , Camundongos Endogâmicos C57BL , Proteínas Mutantes/metabolismo , Inclusão em Parafina , Estrutura Secundária de Proteína , Fixação de Tecidos
3.
PLoS One ; 15(8): e0238081, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32833997

RESUMO

Cataracts are a major cause of blindness worldwide and commonly occur in individuals over 70 years old. Cataracts can also appear earlier in life due to genetic mutations. The lens proteins, αA- and αB-crystallins, are chaperone proteins that have important roles maintaining protein solubility to prevent cataract formation. Mutations in the CRYAA and CRYAB crystallin genes are associated with autosomal dominant early onset human cataracts. Although studies about the proteomic and genomic changes that occur in cataracts have been reported, metabolomics studies are very limited. Here, we directly investigated cataract metabolism using gas-chromatography-mass spectrometry (GC-MS) to analyze the metabolites in adult Cryaa-R49C and Cryab-R120G knock-in mouse lenses. The most abundant metabolites were myo-inositol, L-(+)-lactic acid, cholesterol, phosphate, glycerol phosphate, palmitic and 9-octadecenoic acids, α-D-mannopyranose, and ß-D-glucopyranose. Cryaa-R49C knock-in mouse lenses had a significant decrease in the number of sugars and minor sterols, which occurred in concert with an increase in lactic acid. Cholesterol composition was unchanged. In contrast, Cryab-R120G knock-in lenses exhibited increased total amino acid content including valine, alanine, serine, leucine, isoleucine, glycine, and aspartic acid. Minor sterols, including cholest-7-en-3-ol and glycerol phosphate were decreased. These studies indicate that lenses from Cryaa-R49C and Cryab-R120G knock-in mice, which are models for human cataracts, have unique amino acid and metabolite profiles.


Assuntos
Catarata/genética , Catarata/metabolismo , Cristalino/metabolismo , Mutação , alfa-Cristalinas/genética , Animais , Modelos Animais de Doenças , Humanos , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL
4.
BMC Res Notes ; 13(1): 315, 2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-32616056

RESUMO

OBJECTIVE: Understanding the mechanisms of cataract formation is important for age-related and hereditary cataracts caused by mutations in lens protein genes. Lens proteins of the crystallin gene families α-, ß-, and γ-crystallin are the most abundant proteins in the lens. Single point mutations in crystallin genes cause autosomal dominant cataracts in multigenerational families. Our previous proteomic and RNAseq studies identified genes and proteins altered in the early stages of cataract formation in mouse models. Histones H2A, H2B, and H4 increase in abundance in αA- and αB-crystallin mutant mouse lenses and in cultured cells expressing the mutant form of αA-crystallin linked with hereditary cataracts. RESULTS: In this study of histones in mutant lenses, we extracted histones from adult mouse lenses from cryaa-R49C and cryab-R120G mutant knock-in mice. We characterized the histones using matrix-assisted laser desorption/ionization time of flight (MALDI-TOF)-mass spectrometric analysis and gel electrophoresis and characterized the lens nucleus morphology using electron microscopy (EM). The relative abundance of histone H3 protein decreased in lenses from cryaa-R49C mutant mice and the relative abundance of histone H2 increased in these lenses. Electron microscopy of nuclei from cryaa-R49C-homozygous mutant mouse lenses revealed a pronounced alteration in the distribution of heterochromatin.


Assuntos
Catarata/genética , Heterocromatina/ultraestrutura , Histonas/metabolismo , Cristalino/metabolismo , Cadeia A de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/genética , Animais , Catarata/metabolismo , Técnicas de Introdução de Genes , Cristalino/ultraestrutura , Camundongos , Mutação
5.
Biochem Biophys Rep ; 22: 100748, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32154391

RESUMO

Creatine kinase (CK) is an energy storage enzyme that plays an important role in energy metabolism. CK/phosphocreatine functions as an energy buffer and links ATP production sites with ATP utilization sites. Several key mutations in the αA-crystallin (cryaa) and αB-crystallin (cryab) genes have been linked with autosomal-dominant, hereditary human cataracts. The cryaa-R49C mutation was identified in a four-generation Caucasian family. We previously identified an increase in the quantity of CK complexed with α-crystallin in the lenses of knock-in mice expressing the cryaa-R49C mutation using proteomic analyses. Increased levels of CK in postnatal cataractous lenses may indicate increased ATP requirements during early cataract development. To gain a further understanding of the relationship between CK and α-crystallin, we investigated whether α-crystallin interacts with and forms complexes with CK, in vitro. Isothermal titration calorimetry (ITC) showed that each CK dimer bound to 28 α-crystallin subunits, with a Kd of 3.3 × 10-7 M, and that the interaction between α-crystallin and CK was endothermic, thermodynamically favorable, and entropy-driven. High-salt concentrations did not affect the interaction between CK and α-crystallin, suggesting that the interaction between CK and α-crystallin is primarily hydrophobic. Gel permeation chromatography (GPC) detected water-soluble α-crystallin and CK complexes, as determined by increased light scattering after complex formation. In addition, CK and α-crystallin formed partially-water-insoluble, high-molecular-mass complexes. Enzyme-linked immunosorbent assay (ELISA)-based enzymatic activity analyses of lens homogenates showed a 17-fold increase in CK activity in the postnatal lenses of cryaa-R49C knock-in mice. These studies indicate that the interaction between α-crystallin and CK is functionally important and that increased CK levels may be necessary to meet the increased ATP demands of ATP-dependent functions in cataractous lenses.

6.
Invest Ophthalmol Vis Sci ; 60(10): 3320-3331, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31369034

RESUMO

Purpose: We previously identified an oxysterol, VP1-001 (also known as compound 29), that partially restores the transparency of lenses with cataracts. To understand the mechanism of VP1-001, we tested the ability of its enantiomer, ent-VP1-001, to bind and stabilize αB-crystallin (cryAB) in vitro and to produce a similar therapeutic effect in cryAB(R120G) mutant and aged wild-type mice with cataracts. VP1-001 and ent-VP1-001 have identical physicochemical properties. These experiments are designed to critically evaluate whether stereoselective binding to cryAB is required for activity. Methods: We compared the binding of VP1-001 and ent-VP1-001 to cryAB using in silico docking, differential scanning fluorimetry (DSF), and microscale thermophoresis (MST). Compounds were delivered by six topical administrations to mouse eyes over 2 weeks, and the effects on cataracts and lens refractive measures in vivo were examined. Additionally, lens epithelial and fiber cell morphologies were assessed via transmission electron microscopy. Results: Docking studies suggested greater binding of VP1-001 into a deep groove in the cryAB dimer compared with ent-VP1-001. Consistent with this prediction, DSF and MST experiments showed that VP1-001 bound cryAB, whereas ent-VP1-001 did not. Accordingly, topical treatment of lenses with ent-VP1-001 had no effect, whereas VP1-001 produced a statistically significant improvement in lens clarity and favorable changes in lens morphology. Conclusions: The ability of VP1-001 to bind native cryAB dimers is important for its ability to reverse lens opacity in mouse models of cataracts.


Assuntos
Catarata/tratamento farmacológico , Oxisteróis/farmacologia , Cadeia B de alfa-Cristalina/metabolismo , Administração Oftálmica , Animais , Catarata/metabolismo , Catarata/patologia , Cromatografia em Gel , Modelos Animais de Doenças , Fluorometria , Cristalino/efeitos dos fármacos , Cristalino/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Soluções Oftálmicas , Oxisteróis/metabolismo , Agregação Patológica de Proteínas/tratamento farmacológico , Lâmpada de Fenda
7.
Biochem Biophys Rep ; 15: 7-12, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30023439

RESUMO

The aggregation of crystallins in lenses is associated with cataract formation. We previously reported that mutant crystallins are associated with an increased abundance of histones in knock-in and knockout mouse models. However, very little is known about the specific interactions between lens crystallins and histones. Here, we performed in vitro analyses to determine whether α-crystallin interacts with histones directly. Isothermal titration calorimetry revealed a strong histone-α-crystallin binding with a Kd of 4 × 10-7 M, and the thermodynamic parameters suggested that the interaction was both entropy and enthalpy driven. Size-exclusion chromatography further showed that histone-α-crystallin complexes are water soluble but become water insoluble as the concentration of histones is increased. Right-angle light scattering measurements of the water-soluble fractions of histone-α-crystallin mixtures showed a decrease in the oligomeric molecular weight of α-crystallin, indicating that histones alter the oligomerization of α-crystallin. Taken together, these findings reveal for the first time that histones interact with and affect the solubility and aggregation of α-crystallin, indicating that the interaction between α-crystallin and histones in the lens is functionally important.

8.
PLoS One ; 13(1): e0190817, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29338044

RESUMO

The mammalian eye lens expresses a high concentration of crystallins (α, ß and γ-crystallins) to maintain the refractive index essential for lens transparency. Crystallins are long-lived proteins that do not turnover throughout life. The structural destabilization of crystallins by UV exposure, glycation, oxidative stress and mutations in crystallin genes leads to protein aggregation and development of cataracts. Several destabilizing mutations in crystallin genes are linked with human autosomal dominant hereditary cataracts. To investigate the mechanism by which the α-crystallin mutations Cryaa-R49C and Cryab-R120G lead to cataract formation, we determined whether these mutations cause an altered expression of specific transcripts in the lens at an early postnatal age by RNA-seq analysis. Using knock-in mouse models previously generated in our laboratory, in the present work, we identified genes that exhibited altered abundance in the mutant lenses, including decreased transcripts for Clic5, an intracellular water channel in Cryaa-R49C heterozygous mutant lenses, and increased transcripts for Eno1b in Cryab-R120G heterozygous mutant lenses. In addition, RNA-seq analysis revealed increased histones H2B, H2A, and H4 gene expression in Cryaa-R49C mutant lenses, suggesting that the αA-crystallin mutation regulates histone expression via a transcriptional mechanism. Additionally, these studies confirmed the increased expression of histones H2B, H2A, and H4 by proteomic analysis of Cryaa-R49C knock-in and Cryaa;Cryab gene knockout lenses reported previously. Taken together, these findings offer additional insight into the early transcriptional changes caused by Cryaa and Cryab mutations associated with autosomal dominant human cataracts, and indicate that the transcript levels of certain genes are affected by the expression of mutant α-crystallin in vivo.


Assuntos
Catarata/genética , Mutação , Cadeia A de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/genética , Animais , Carboxipeptidases/genética , Carboxipeptidases/metabolismo , Catarata/metabolismo , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Introdução de Genes , Histonas/genética , Histonas/metabolismo , Humanos , Cristalino/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Proteínas/genética , Proteínas/metabolismo , Proteômica , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Cadeia A de alfa-Cristalina/metabolismo , Cadeia B de alfa-Cristalina/metabolismo
9.
Biochim Biophys Acta ; 1860(1 Pt B): 234-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26071686

RESUMO

BACKGROUND: Knock-in mice provide useful models of congenital and age-related cataracts caused by α-crystallin mutations. R49C αA-crystallin and R120G αB-crystallin mutations are linked with hereditary cataracts. Knock-in αA-R49C+/- heterozygotes develop cataracts by 1-2months, whereas homozygote mice have cataracts at birth. The R49C mutation drastically reduces lens protein water solubility and causes cell death in knock-in mouse lenses. Mutant crystallin cannot function as a chaperone, which leads to protein aggregation and lens opacity. Protein aggregation disrupts the lens fiber cell structure and normal development and causes cell death in epithelial and fiber cells. We determined what aspects of the wild-type phenotype are age-dependently altered in the mutant lens. METHODS: Wild-type, heterozygote (αA-R49C+/-), and homozygote (αA-R49C+/+) mouse lenses were assessed pre- and postnatally for lens morphology (electron microscopy, immunohistochemistry), and autophagy or unfolded protein response markers (immunoblotting). RESULTS: Morphology was altered by embryonic day 17 in R49C+/+ lenses; R49C+/- lens morphology was unaffected at this stage. Active autophagy in the lens epithelium of mutant lenses was indicated by the presence of autophagosomes using electron microscopy. Protein p62 levels, which are degraded specifically by autophagy, increased in αA-R49C mutant versus wild-type lenses, suggesting autophagy inhibition in the mutant lenses. The unfolded protein response marker XBP-1 was upregulated in adult lenses of αB-R120G+/+ mice, suggesting its role in lens opacification. CONCLUSIONS: Mutated crystallins alter lens morphology, autophagy, and stress responses. GENERAL SIGNIFICANCE: Therapeutic modulation of autophagic pathways may improve protein degradation in cataractous lenses and reduce lens opacity. This article is part of a Special Issue entitled Crystallin Biochemistry in Health and Disease.


Assuntos
Envelhecimento/genética , Catarata/genética , Catarata/patologia , Cristalinas/genética , Cristalino/patologia , Resposta a Proteínas não Dobradas/genética , Animais , Autofagia/genética , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Cristalino/metabolismo , Camundongos , Mutação
10.
Science ; 350(6261): 674-7, 2015 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-26542570

RESUMO

Cataracts reduce vision in 50% of individuals over 70 years of age and are a common form of blindness worldwide. Cataracts are caused when damage to the major lens crystallin proteins causes their misfolding and aggregation into insoluble amyloids. Using a thermal stability assay, we identified a class of molecules that bind α-crystallins (cryAA and cryAB) and reversed their aggregation in vitro. The most promising compound improved lens transparency in the R49C cryAA and R120G cryAB mouse models of hereditary cataract. It also partially restored protein solubility in the lenses of aged mice in vivo and in human lenses ex vivo. These findings suggest an approach to treating cataracts by stabilizing α-crystallins.


Assuntos
Catarata/tratamento farmacológico , Hidroxicolesteróis/farmacologia , Cadeia A de alfa-Cristalina/química , Cadeia B de alfa-Cristalina/química , Amiloide/antagonistas & inibidores , Amiloide/química , Animais , Varredura Diferencial de Calorimetria , Catarata/genética , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Hidroxicolesteróis/química , Hidroxicolesteróis/uso terapêutico , Camundongos , Conformação Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Cadeia A de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/genética
11.
PLoS One ; 9(4): e95507, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24760011

RESUMO

αA-crystallin and αB-crystallin are members of the small heat shock protein family and function as molecular chaperones and major lens structural proteins. Although numerous studies have examined their chaperone-like activities in vitro, little is known about the proteins they protect in vivo. To elucidate the relationships between chaperone function, substrate binding, and human cataract formation, we used proteomic and mass spectrometric methods to analyze the effect of mutations associated with hereditary human cataract formation on protein abundance in αA-R49C and αB-R120G knock-in mutant lenses. Compared with age-matched wild type lenses, 2-day-old αA-R49C heterozygous lenses demonstrated the following: increased crosslinking (15-fold) and degradation (2.6-fold) of αA-crystallin; increased association between αA-crystallin and filensin, actin, or creatine kinase B; increased acidification of ßB1-crystallin; increased levels of grifin; and an association between ßA3/A1-crystallin and αA-crystallin. Homozygous αA-R49C mutant lenses exhibited increased associations between αA-crystallin and ßB3-, ßA4-, ßA2-crystallins, and grifin, whereas levels of ßB1-crystallin, gelsolin, and calpain 3 decreased. The amount of degraded glutamate dehydrogenase, α-enolase, and cytochrome c increased more than 50-fold in homozygous αA-R49C mutant lenses. In αB-R120G mouse lenses, our analyses identified decreased abundance of phosphoglycerate mutase, several ß- and γ-crystallins, and degradation of αA- and αB-crystallin early in cataract development. Changes in the abundance of hemoglobin and histones with the loss of normal α-crystallin chaperone function suggest that these proteins also play important roles in the biochemical mechanisms of hereditary cataracts. Together, these studies offer a novel insight into the putative in vivo substrates of αA- and αB-crystallin.


Assuntos
Cristalino/metabolismo , Cadeia A de alfa-Cristalina/metabolismo , Cadeia B de alfa-Cristalina/metabolismo , Animais , Calpaína/genética , Calpaína/metabolismo , Cristalinas/genética , Cristalinas/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Gelsolina/genética , Gelsolina/metabolismo , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Knockout , Camundongos Mutantes , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Cadeia A de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/genética
12.
Exp Eye Res ; 115: 263-73, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23872361

RESUMO

The formation of cataracts is associated with the accumulation of protein aggregates in the ocular lens, suggesting that defective protein degradation plays a role in cataract pathogenesis. Accumulation of the p62 protein has recently been identified as a marker for impaired autophagy in a variety of tissues; however, little information exists on its expression in the ocular lens and in cataracts. In the present study we examined the expression of p62 in the mouse lens and compared its expression in wild-type lenses with that in lenses from knock-in mice with an arginine to glycine mutation in αB-crystallin (αB-R120G) that is known to cause human hereditary cataract. Immunohistochemical, immunoblotting, and transmission electron microscopic analyses of wild type and αB-R120G mutant mice were performed. To assess the effect of increased protein aggregation on autophagy, immunohistochemical staining was performed with an anti-p62 antibody, revealing the presence of p62-positive punctate staining in a band of denucleated cortical fiber cells. The number and size of p62 puncta were significantly greater in αB-R120G homozygous mutant lenses than in wild type and heterozygous mutant lenses. p62 staining was also abundant in lens epithelial cells and was concentrated around the nuclear membrane. Double-membraned structures similar to autophagosomes containing cellular cytoplasmic content were detected in lens epithelial cells by transmission electron microscopy. The autophagosomes in lens epithelial cells from αB-R120G homozygous mutant mice were larger than those in wild type mice. Double-membraned structures that are probably autophagosomes were also detected in cortical fiber cells and were more abundant in the αB-R120G homozygous mutant lens than the wild type lens. This study demonstrates p62 distribution as speckles in the lens fiber cells, altered levels of p62 expression, and the presence of autophagosomes in the ocular lens of αB-R120G mutant mice. We propose that autophagy is inhibited in the αB-R120G mutant lenses because of a defect in protein degradation after autophagosome formation. Further work is necessary to determine the relationship between αB-crystallin function, autophagy, and cataract formation.


Assuntos
Autofagia/genética , Catarata/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica/fisiologia , Fatores de Transcrição/genética , Cadeia B de alfa-Cristalina/genética , Animais , Catarata/metabolismo , Catarata/patologia , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Técnica Indireta de Fluorescência para Anticorpo , Genótipo , Immunoblotting , Cristalino/metabolismo , Cristalino/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Fagossomos/ultraestrutura , Mutação Puntual , Reação em Cadeia da Polimerase , Fator de Transcrição TFIIH , Fatores de Transcrição/metabolismo , Cadeia B de alfa-Cristalina/metabolismo
13.
Biochemistry ; 52(17): 2933-48, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23590631

RESUMO

Mice with deletion of genes for small heat shock proteins αA- and αB-crystallin (αA/αB(-/-)) develop cataracts. We used proteomic analysis to identify lens proteins that change in abundance after deletion of these α-crystallin genes. Wild-type (WT) and αA/αB(-/-) knockout (DKO) mice were compared using two-dimensional difference gel electrophoresis and mass spectrometric analysis, and protein identifications were validated by Mascot proteomic software. The abundance of histones H2A, H4, and H2B fragment, and a low molecular weight ß1-catenin increased 2-3-fold in postnatal day 2 lenses of DKO lenses compared with WT lenses. Additional major increases were observed in abundance of ßB2-crystallin and vimentin in 30-day-old lenses of DKO animals compared with WT animals. Lenses of DKO mice were comprised of nine protein spots containing ßB2-crystallin at 10-40-fold higher abundance and three protein spots containing vimentin at ≥2-fold higher abundance than in WT lenses. Gel permeation chromatography identified a unique 328 kDa protein in DKO lenses, containing ß-crystallin, demonstrating aggregation of ß-crystallin in the absence of α-crystallins. Together, these changes provide biochemical evidence for possible functions of specific cell adhesion proteins, cytoskeletal proteins, and crystallins in lens opacities caused by the absence of the major chaperones, αA- and αB-crystallins.


Assuntos
Cristalinas/genética , Proteínas de Choque Térmico/genética , Proteômica , beta Catenina/genética , Fatores Etários , Animais , Western Blotting , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Cristalinas/metabolismo , Eletroforese em Gel Bidimensional , Proteínas de Choque Térmico/metabolismo , Histonas/metabolismo , Espectrometria de Massas , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vimentina/metabolismo , beta Catenina/metabolismo
14.
Invest Ophthalmol Vis Sci ; 52(11): 8330-41, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21873653

RESUMO

PURPOSE: To determine whether class 1 UV-blocking contact lenses protect against UVB radiation-induced damage in a human lens epithelial cell line (HLE B-3) and postmortem human lenses using a proteomics approach. METHODS: HLE B-3 cells were exposed to 6.4 mW/cm(2) UVB radiation at 302 nm for 2 minutes (768 mJ/cm(2)) with or without covering by senofilcon A class 1 UV-blocking contact lenses or lotrafilcon A non-UV-blocking (lotrafilcon A has some UV-blocking ability, albeit minimal) contact lenses. Control cells were not exposed to UVB radiation. Four hours after treatment, cells were analyzed by two-dimensional difference gel electrophoresis and tandem mass spectrometry, and changes in protein abundance were quantified. F-actin and microtubule cytoskeletons were examined by fluorescence staining. In addition, human donor lenses were exposed to UVB radiation at 302 nm for 4 minutes (1536 mJ/cm(2)). Cortical and epithelial cell proteins were scraped from lens surfaces and subjected to the same protein analyses. RESULTS: Senofilcon A lenses were beneficial for protecting HLE B-3 cells against UVB radiation-induced changes in caldesmon 1 isoform, lamin A/C transcript variant 1, DEAD (Asp-Glu-Ala-Asp) box polypeptide, ß-actin, glyceraldehyde 3-phosphate dehydrogenase (G3PDH), annexin A2, triose phosphate isomerase, and ubiquitin B precursor. These contact lenses also prevented actin and microtubule cytoskeleton changes typically induced by UVB radiation. Conversely, non-UV-blocking contact lenses were not protective. UVB-irradiated human lenses showed marked reductions in αA-crystallin, αB-crystallin, aldehyde dehydrogenase 1, ßS-crystallin, ßB2-crystallin, and G3PDH, and UV-absorbing contact lenses significantly prevented these alterations. CONCLUSIONS: Senofilcon A class 1 UV-blocking contact lenses largely prevented UVB-induced changes in protein abundance in lens epithelial cells and in human lenses.


Assuntos
Lentes de Contato , Células Epiteliais/efeitos da radiação , Cristalino/efeitos da radiação , Proteômica/métodos , Raios Ultravioleta/efeitos adversos , Catarata/prevenção & controle , Linhagem Celular Transformada , Eletroforese em Gel Bidimensional , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Proteínas do Olho/metabolismo , Humanos , Cristalino/citologia , Espectrometria de Massas , Pessoa de Meia-Idade , Lesões por Radiação/patologia , Lesões por Radiação/fisiopatologia , Lesões por Radiação/prevenção & controle
15.
Photochem Photobiol ; 87(6): 1321-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21770952

RESUMO

Crystallin proteins are responsible for maintaining lens transparency and allowing the lens to focus light undistorted onto the retina. The α-crystallins are the major lens crystallins, and function as both structural proteins and chaperones to protect all lens proteins from damage leading to lens deterioration. Because lens crystallin proteins do not turn over, the damage they accumulate can lead to cataracts, the world's leading cause of blindness. Photosensitizing porphyrins can accumulate in the eye through either endogenous metabolism or through therapeutic or diagnostic procedures. Porphyrin buildup exacerbates lens aging through increased levels of singlet oxygen, resulting in protein polymerization and amino acid residue alteration. Tryptophans oxidize to kynurenine and N-formylkynurenine (NFK) causing irreversible changes in the refractive index of the normally transparent lens, leading to development of cataracts. Additionally, NFK is itself a photosensitizer, and its presence exacerbates lens deterioration. This work uses anti-NFK antiserum to study porphyrin-facilitated photooxidation of α-crystallin tryptophan residues. In vitro experiments show that four biologically interesting porphyrins mediate α-crystallin polymerization and accumulation of both protein radicals and NFK. Confocal microscopy of cultured human lens epithelial cells indicates that while all four porphyrins photosensitize cellular proteins, not all oxidize the tryptophans of cellular α-crystallin to NFK.


Assuntos
Cinurenina/análogos & derivados , Fotoquímica , Porfirinas/química , alfa-Cristalinas/química , Linhagem Celular , Humanos , Cinurenina/análise , Microscopia Confocal , Oxirredução , Refratometria
16.
J Ocul Pharmacol Ther ; 27(3): 265-71, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21574867

RESUMO

PURPOSE: The purpose of this study was to compare the in vitro effects of triamcinolone acetonide (TA) and dexamethasone sodium phosphate (DEX) on human lens epithelial cells (HLE B-3). METHODS: HLE B-3 cells were exposed for 24 h to commercially available TA (c-TA) and dimethylsulfoxide-solubilized TA (s-TA). The cells were treated with 1,000 (clinical dose), 750, 500, 200, and 100 µg/mL concentrations of c-TA, s-TA, and supernatant for 24 h. The cells were also treated with DEX at 2, 1, 0.5, 0.2, 0.1 (clinical dose), and 0.05 mg/mL. Cell viability, caspase-3/7 activity, and DNA fragmentation analyses were performed. RESULTS: The mean cell viabilities of HLE B-3 after exposure to c-TA at 1,000, 750, 500, 200, and 100 µg/mL were significantly reduced compared with control untreated cells. The s-TA also significantly reduced cell viability at 1,000, 750, and 500 µg/mL compared with dimethylsulfoxide control. The supernatant did not reduce cell viability. Caspase-3/7 activity significantly increased after treatment with c-TA and s-TA. DNA laddering revealed bands at 200 bp intervals with both c-TA at≥100 µg/mL and s-TA at ≥500 µg/mL. The cell viabilities of HLE B-3 after 24 h exposure to DEX were significantly reduced at 2 and 1 mg/mL but not at lower concentrations tested. Caspase-3/7 activities in HLE B-3 cells were not increased significantly after treatment with DEX at any dose tested. DNA laddering did not reveal any band at any dose tested. CONCLUSION: This study showed that TA at its clinical dose (1,000 µg/mL) in both commercial preparation and solubilized forms decrease HLE B-3 cell viability through an apoptotic pathway. DEX at its clinical dose (0.1 mg/mL) does not decrease cell viability or cause any increase of caspase-3/7 activity. This study suggests that for long-term sustained-release devices, DEX may be less damaging to human lens cells than TA.


Assuntos
Anti-Inflamatórios/toxicidade , Dexametasona/análogos & derivados , Células Epiteliais/efeitos dos fármacos , Glucocorticoides/toxicidade , Cristalino/efeitos dos fármacos , Triancinolona Acetonida/toxicidade , Anti-Inflamatórios/química , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Preparações de Ação Retardada/química , Preparações de Ação Retardada/toxicidade , Dexametasona/toxicidade , Dimetil Sulfóxido/química , Glucocorticoides/química , Humanos , Concentração Osmolar , Veículos Farmacêuticos/química , Solubilidade , Triancinolona Acetonida/química
17.
PLoS One ; 6(3): e17671, 2011 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-21445271

RESUMO

An autosomal dominant missense mutation in αB-crystallin (αB-R120G) causes cataracts and desmin-related myopathy, but the underlying mechanisms are unknown. Here, we report the development of an αB-R120G crystallin knock-in mouse model of these disorders. Knock-in αB-R120G mice were generated and analyzed with slit lamp imaging, gel permeation chromatography, immunofluorescence, immunoprecipitation, histology, and muscle strength assays. Wild-type, age-matched mice were used as controls for all studies. Both heterozygous and homozygous mutant mice developed myopathy. Moreover, homozygous mutant mice were significantly weaker than wild-type control littermates at 6 months of age. Cataract severity increased with age and mutant gene dosage. The total mass, precipitation, and interaction with the intermediate filament protein vimentin, as well as light scattering of αB-crystallin, also increased in mutant lenses. In skeletal muscle, αB-R120G co-aggregated with desmin, became detergent insoluble, and was ubiquitinated in heterozygous and homozygous mutant mice. These data suggest that the cataract and myopathy pathologies in αB-R120G knock-in mice share common mechanisms, including increased insolubility of αB-crystallin and co-aggregation of αB-crystallin with intermediate filament proteins. These knock-in αB-R120G mice are a valuable model of the developmental and molecular biological mechanisms that underlie the pathophysiology of human hereditary cataracts and myopathy.


Assuntos
Catarata/genética , Cristalinas/genética , Modelos Animais de Doenças , Predisposição Genética para Doença , Doenças Musculares/genética , Mutação , Animais , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Mutantes
18.
Biochem Biophys Res Commun ; 401(2): 192-6, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20833134

RESUMO

αA-crystallin is a lens chaperone that plays an essential role in the transparency and refractive properties of the lens. Mutations in αA-crystallin have been associated with the development of hereditary cataracts. The R49C mutation of αA-crystallin (αA-R49C) was identified in a four-generation Caucasian family with hereditary cataracts. The αA-R49C protein forms larger-than-normal oligomers in the lens and has decreased solubility. This aberrant αA-R49C oligomerization suggests that protein folding is altered. However, whether activation of the unfolded protein response (UPR) occurs during crystallin mutation-induced cataract formation and whether the UPR causes cell death under these conditions is unclear. We investigated UPR activation in an in vivo mouse model of αA-R49C using immunoblot analysis of lens extracts. We found that expression of the endoplasmic reticulum (ER) chaperone, BiP, was 5-fold higher in homozygous αA-R49C lenses than in wild type lenses. Analysis of proteins typically expressed during the UPR revealed that ATF-4 and CHOP levels were also higher in homozygous lenses than in wild type lenses, while the opposite was true of ATF-6 and XBP-1. Taken together, these findings show that mutation of αA-crystallin induces activation of the UPR during cataract formation. They also suggest that the UPR is an important mediator of cell death observed in homozygous αA-R49C lenses.


Assuntos
Catarata/metabolismo , Resposta a Proteínas não Dobradas , Cadeia A de alfa-Cristalina/metabolismo , Animais , Catarata/genética , Camundongos , Camundongos Mutantes , Mutação , Cadeia A de alfa-Cristalina/genética
19.
Exp Eye Res ; 91(5): 640-51, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20709056

RESUMO

α-Crystallins are small heat-shock proteins important to lens transparency that provide the lens with its refractive properties. In their role as molecular chaperones, these crystallins also prevent protein aggregation, affect cytoskeletal remodeling, enhance resistance to cell stress, and provide lens cells with protection against apoptosis. While many of the functions assigned to αA-crystallin are attributable to its presence in the cytoplasm of lens cells, αA-crystallin also has been detected at the lens plasma membrane. However, how αA-crystallin becomes linked to the plasma membrane or what its functions are at this site has remained unknown. In this study, we examined the mechanisms by which αA-crystallin becomes associated with the lens membrane, focusing specifically on its interaction with membrane receptors, and the differentiation-specificity of these interactions. We also determined how the long-term absence of αA-crystallin alters receptor-linked signaling pathways. αA-crystallin association with membrane receptors was determined by co-immunoprecipitation analysis; its membrane localization was examined by confocal imaging; and the effect of αA-crystallin loss-of-function on the activation state of signaling molecules in pathways linked to membrane receptors was determined by immunoblot analysis. The results show that, in lens epithelial cells, plasma membrane αA-crystallin was primarily localized to apicolateral borders, reflecting the association of αA-crystallin with E-cadherin complexes. These studies also provide the first evidence that αA-crystallin maintained its association with the plasma membrane in lens cortical fiber cells, where it was localized to lateral interfaces, and further show that this association was mediated, in part, by αA-crystallin interaction with α6 integrin receptor complexes. We report that the absence of αA-crystallin led to constitutive activation of the stress kinases p38 and JNK, classical inducers of apoptotic cell death, and the loss of the phospho-Bad pro-survival signal, effects that were greatest in differentiating lens fiber cells. Concurrent with this, activation of FAK and ERK kinases was increased, demonstrating that these receptor-linked pathways also were dysregulated in the absence of αA-crystallin. These data link αA-crystallin plasma membrane association to its differentiation-state-specific interaction with E-cadherin and α6 integrin receptor complexes. The changes in cell signaling in αA-crystallin-null lenses suggest that dysregulation of receptor-linked cell-signaling pathways that accompany the failure of αA-crystallin to associate with membrane receptors may be responsible for the induction of apoptosis. The observed changes in lens cell signaling likely reflect long-term functional adaptations to the absence of the αA-crystallin chaperone/small heat-shock protein.


Assuntos
Células Epiteliais/metabolismo , Integrina alfa6/metabolismo , Cristalino/metabolismo , Receptores de Laminina/metabolismo , Transdução de Sinais/fisiologia , Cadeia A de alfa-Cristalina/metabolismo , Animais , Apoptose , Caderinas/metabolismo , Catarata/metabolismo , Membrana Celular/metabolismo , Sobrevivência Celular , Embrião de Galinha , Técnica Indireta de Fluorescência para Anticorpo , Immunoblotting , Imunoprecipitação , Cristalino/embriologia , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Knockout , Codorniz , Transfecção , Cadeia A de alfa-Cristalina/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Mol Vis ; 16: 1041-6, 2010 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-20606707

RESUMO

The disrupted morphology of lenses in mouse models for cataracts precludes accurate in vitro assessment of lens growth by weight. To overcome this limitation, we developed morphometric methods to assess defects in eye lens growth and shape in mice expressing the alphaA-crystallin R49C (alphaA-R49C) mutation. Our morphometric methods determine quantitative shape and dry weight of the whole lens from histological sections of the lens. This method was then used to quantitatively compare the biometric growth patterns of lenses of different genotypes of mice from birth to 12 months. The wild type dry lens weights determined using the morphometric method were comparable to previously reported weights. Next we applied the method to assessing the lenses of alphaA-R49C knock-in mice, which exhibit decreased alphaA-crystallin protein solubility, resulting in a variety of growth abnormalities including early cataract formation, decreased eye and lens size, failure to form the equatorial bow region, and continued lens cell death, sometimes resulting in the entire loss of the lens and eye. Our morphometric methods reproducibly quantified these defects by combining histology, microscopy, and image analysis. The volume measurement accurately represented the total growth of the lens, whereas the geometric shape of the lens more accurately quantified the differences between the growth of the mutant and wild-type lenses. These methods are robust tools for measuring dry lens weight and quantitatively comparing the growth of small lenses that are difficult to weigh accurately such as those from very young mice and mice with developmental lens defects.


Assuntos
Biometria/métodos , Cristalino/anormalidades , Cristalino/patologia , Fenótipo , Envelhecimento , Animais , Arginina , Catarata/genética , Cisteína , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Genótipo , Heterozigoto , Homozigoto , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Tamanho do Órgão , Solubilidade , Cadeia A de alfa-Cristalina/química , Cadeia A de alfa-Cristalina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA