Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Vis ; 23: 228-241, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28465655

RESUMO

PURPOSE: To examine the effects of autophagy deficiency induced by RPE-specific deletion of Atg5 or Atg7 in mice as a function of age. METHODS: Conditional knockout mice with a floxed allele of Atg5 or Atg7 were crossed with inducible VMD2-rtTA/Cre transgenic mice. VMD2-directed RPE-specific Cre recombinase expression was induced with doxycycline feeding in the resulting mice. Cre-mediated deletion of floxed Atg5 or Atg7 resulted in RPE-specific inactivation of the Atg5 or Atg7 gene. Plastic and thin retinal sections were analyzed with light and electron microscopy for histological changes. Photoreceptor outer segment (POS) thickness in plastic sections was measured using the Adobe Photoshop CS4 extended ruler tool. Autophagic adaptor p62/SQSTM1 and markers for oxidatively damaged lipids, proteins, and DNA were examined with immunofluorescence staining of cryosections. Fluorescence signals were quantified using Image J software. RESULTS: Accumulation of p62/SQSTM1 reflecting autophagy deficiency was observed in the RPE of the Atg5ΔRPE and Atg7ΔRPE mice. 3-nitrotyrosine, advanced glycation end products (AGEs), and 8-hydroxy-2'-deoxyguanosine (8-OHdG), markers for oxidatively damaged proteins and DNA, were also found to accumulate in the RPE of these mice. We observed retinal degeneration in 35% of the Atg5ΔRPE mice and 45% of the Atg7ΔRPE mice at 8 to 24 months old. Degeneration severity and the number of mice with degeneration increased with age. The mean POS thickness of these mice was 25 µm at 8-12 months, 15 µm at 13-18 months, and 3 µm at 19-24 months, compared to 35 µm, 30 µm, and 24 µm in the wild-type mice, respectively. Early age-related macular degeneration (AMD)-like RPE defects were found in all the Atg5ΔRPE and Atg7ΔRPE mice 13 months old or older, including vacuoles, uneven RPE thickness, diminished basal infoldings, RPE hypertrophy/hypotrophy, pigmentary irregularities, and necrosis. The severity of the RPE defects increased with age and in the mice with retinal degeneration. RPE atrophy and choroidal neovascularization (CNV) were occasionally observed in the Atg5ΔRPE and Atg7ΔRPE mice with advanced age. CONCLUSIONS: Autophagy deficiency induced by RPE-specific deletion of Atg5 or Atg7 predisposes but does not necessarily drive the development of AMD-like phenotypes or retinal degeneration.


Assuntos
Proteína 5 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/genética , Autofagia , Deleção de Genes , Degeneração Macular/genética , Degeneração Retiniana/genética , Epitélio Pigmentado da Retina/patologia , Alelos , Animais , Biomarcadores/metabolismo , Eletrorretinografia , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Degeneração Macular/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Degeneração Retiniana/patologia , Epitélio Pigmentado da Retina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
2.
Biochim Biophys Acta ; 1842(12 Pt B): 2579-83, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25108282

RESUMO

Adenylyl cyclases (ACs) are a family of enzymes which convert ATP to cAMP, an essential intermediate in many signal transduction pathways. Of the 10 AC genes in man, 9 fall into the category of transmembrane ACs (tmACs), which associate with G-protein coupled receptors (GPCRs) and are activated by forskolin. The 10th AC, termed soluble AC (sAC) is neither activated by forskolin nor does it interact with GPCRs. Rather, sAC can be found in many compartments within the cell and is activated by bicarbonate. As such, sAC is considered a major sensor of bicarbonate in many tissues. The pathways involving sAC vary in different tissues and organ systems, and are as diverse as facilitating sperm capacitation and regulating pressure in the eye. The role of sAC in the eye has only recently begun to receive significant attention. Here we summarize what is known about the roles of sAC in the eye. This article is part of a Special Issue entitled: The role of soluble adenylyl cyclase in health and disease.


Assuntos
Adenilil Ciclases/metabolismo , Olho/enzimologia , Animais , Humanos
3.
Hum Mol Genet ; 22(23): 4688-97, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23825107

RESUMO

Mutations in bestrophin-1 (Best1) cause Best vitelliform macular dystrophy (BVMD), a dominantly inherited retinal degenerative disease. Best1 is a homo-oligomeric anion channel localized to the basolateral surface of retinal pigment epithelial (RPE) cells. A number of Best1 mutants mislocalize in Madin-Darby canine kidney (MDCK) cells. However, many proteins traffic differently in MDCK and RPE cells, and MDCK cells do not express endogenous Best1. Thus, effects of Best1 mutations on localization in MDCK cells may not translate to RPE cells. To determine whether BVMD causing mutations affect Best1 localization, we compared localization and oligomerization of Best1 with Best1 mutants V9M, W93C, and R218C. In MDCK cells, Best1 and Best1(R218C) were basolaterally localized. Best1(W93C) and Best1(V9M) accumulated in cells. In cultured fetal human retinal pigment epithelium cells (fhRPE) expressing endogenous Best1, Best1(R218C) and Best1(W93C) were basolateral. Best1(V9M) was intracellular. All three mutants exhibited similar fluorescence resonance energy transfer (FRET) efficiencies to, and co-immunoprecipitated with Best1, indicating unimpaired oligomerization. When human Best1 was expressed in RPE in mouse eyes it was basolaterally localized. However, Best1(V9M) accumulated in intracellular compartments in mouse RPE. Co-expression of Best1 and Best1(W93C) in MDCK cells resulted in basolateral localization of both Best1 and Best1(W93C), but co-expression of Best1 with Best1(V9M) resulted in mislocalization of both proteins. We conclude that different mutations in Best1 cause differential effects on its localization and that this effect varies with the presence or absence of wild-type (WT) Best1. Furthermore, MDCK cells can substitute for RPE when examining the effects of BVMD causing mutations on Best1 localization if co-expressed with WT Best1.


Assuntos
Canais de Cloreto/metabolismo , Proteínas do Olho/metabolismo , Canais Iônicos/metabolismo , Distrofia Macular Viteliforme/patologia , Animais , Bestrofinas , Sinalização do Cálcio , Membrana Celular/metabolismo , Células Cultivadas , Canais de Cloreto/genética , Olho/metabolismo , Proteínas do Olho/genética , Regulação da Expressão Gênica , Humanos , Canais Iônicos/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Transporte Proteico/genética , Distrofia Macular Viteliforme/genética
4.
Mol Vis ; 21: 347-59, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25878489

RESUMO

PURPOSE: Mutations in BEST1, encoding Bestrophin-1 (Best1), cause Best vitelliform macular dystrophy (BVMD) and other inherited retinal degenerative diseases. Best1 is an integral membrane protein localized to the basolateral plasma membrane of the retinal pigment epithelium (RPE). Data from numerous in vitro and in vivo models have demonstrated that Best1 regulates intracellular Ca2+ levels. Although it is known from in vitro and crystal structure data that Best1 is also a calcium-activated anion channel, evidence for Best1 functioning as a channel in human RPE is lacking. To assess Best1-associated channel activity in the RPE, we examined the transepithelial electrical properties of fetal human RPE (fhRPE) cells, which express endogenous Best1. METHODS: Using adenovirus-mediated gene transfer, we overexpressed Best1 and the BVMD mutant Best1W93C in fhRPE cells and assessed resting transepithelial potential (TEP), transepithelial resistance, short circuit current (Isc), and intracellular Ca2+ levels. Cl- currents were directly measured in transfected HEK293 cells using whole-cell patch clamp. RESULTS: Best1W93C showed ablated Cl- currents and, when co-expressed, suppressed the channel activity of Best1 in HEK293 cells. In fhRPE, overexpression of Best1 increased TEP and Isc, while Best1W93C diminished TEP and Isc. Substitution of Cl- in the bath media resulted in a significant reduction of Isc in monolayers overexpressing Best1, but no significant Isc change in monolayers expressing Best1W93C. We removed Ca2+ as a limit on transepithelial electrical properties by treating cells with ionomycin, and found that changes in Isc and TEP for monolayers expressing Best1 were absent in monolayers expressing Best1W93C. Similarly, inhibition of calcium-activated anion channels with niflumic acid reduced both Isc and TEP of control and Best1 monolayers, but did not notably affect Best1W93C monolayers. Stimulation with extracellular ATP induced an increase in TEP in control monolayers that was greater than that observed in those expressing Best1(W93C). Examination of [Ca2+]i following ATP stimulation demonstrated that the expression of Best1W93C impaired intracellular Ca2+ signaling. CONCLUSIONS: These data indicate that Best1 activity strongly influences electrophysiology and Ca2+ signaling in RPE cells, and that a common BVMD mutation disrupts both of these parameters. Our findings support the hypothesis that Best1 functions as an anion channel in human RPE.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Membrana Celular/metabolismo , Canais de Cloreto/metabolismo , Células Epiteliais/metabolismo , Proteínas do Olho/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Trifosfato de Adenosina/farmacologia , Adenovírus Humanos/genética , Bestrofinas , Membrana Celular/efeitos dos fármacos , Canais de Cloreto/genética , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Proteínas do Olho/genética , Feto , Expressão Gênica , Vetores Genéticos , Células HEK293 , Humanos , Transporte de Íons/efeitos dos fármacos , Ionomicina/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Mutação , Ácido Niflúmico/farmacologia , Técnicas de Patch-Clamp , Cultura Primária de Células , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/efeitos dos fármacos , Transfecção , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo , Distrofia Macular Viteliforme/patologia
5.
Exp Eye Res ; 121: 74-85, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24560797

RESUMO

BEST1 encodes Bestrophin-1 (Best1), a homo-oligomeric, integral membrane protein localized to the basolateral plasma membrane of the retinal pigment epithelium. Mutations in BEST1 cause five distinct retinal degenerative diseases, including adult vitelliform macular dystrophy (AVMD), autosomal recessive bestrophinopathy (ARB), autosomal dominant vitreoretinochoroidopathy (ADVIRC), and retinitis pigmentosa (RP). The mechanisms underlying these diseases and why mutations cause one disease over another are, for the most part, unknown. To gain insights into these four diseases, we expressed 28 Best1 mutants fused to YFP in polarized MDCK monolayers and, via confocal microscopy and immunofluorescence, live-cell FRET, and reciprocal co-immunoprecipitation experiments, screened these mutants for defects in localization and oligomerization. All 28 mutants exhibited comparable FRET efficiencies to and co-immunoprecipitated with WT Best1, indicating unimpaired oligomerization. RP- and ADVIRC-associated mutants were properly localized to the basolateral plasma membrane of cells, while two AVMD and most ARB mutants were mislocalized. When co-expressed, all mislocalized mutants caused mislocalization of WT Best1 to intracellular compartments. Our current and past results indicate that mislocalization of Best1 is not an absolute feature of any individual bestrophinopathy, occurring in AVMD, BVMD, and ARB. Furthermore, some ARB mutants that do not also cause dominant disease cause mislocalization of Best1, indicating that mislocalization is not a cause of disease, and that absence of Best1 activity from the plasma membrane is tolerated. Lastly, we find that the ARB truncation mutants L174Qfs*57 and R200X can form oligomers with WT Best1, indicating that the first ∼174 amino acids of Best1 are sufficient for oligomerization to occur.


Assuntos
Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Oftalmopatias Hereditárias/genética , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Mutação de Sentido Incorreto , Multimerização Proteica/fisiologia , Doenças Retinianas/genética , Adenoviridae/genética , Animais , Proteínas de Bactérias/metabolismo , Bestrofinas , Western Blotting , Doenças da Coroide/genética , Doenças da Coroide/metabolismo , Cães , Eletrofisiologia , Oftalmopatias Hereditárias/metabolismo , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/metabolismo , Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Proteínas Luminescentes/metabolismo , Células Madin Darby de Rim Canino/metabolismo , Microscopia Confocal , Técnicas de Patch-Clamp , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Doenças Retinianas/metabolismo , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Transfecção , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo
6.
J Biol Chem ; 286(48): 41353-41358, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-21994938

RESUMO

Glaucoma is a leading cause of blindness affecting as many as 2.2 million Americans. All current glaucoma treatment strategies aim to reduce intraocular pressure (IOP). IOP results from the resistance to drainage of aqueous humor (AH) produced by the ciliary body in a process requiring bicarbonate. Once secreted into the anterior chamber, AH drains from the eye via two pathways: uveoscleral and pressure-dependent or conventional outflow (C(t)). Modulation of "inflow" and "outflow" pathways is thought to occur via distinct, local mechanisms. Mice deficient in the bicarbonate channel bestrophin-2 (Best2), however, exhibit a lower IOP despite an increase in AH production. Best2 is expressed uniquely in nonpigmented ciliary epithelial (NPE) cells providing evidence for a bicarbonate-dependent communicative pathway linking inflow and outflow. Here, we show that bicarbonate-sensitive soluble adenylyl cyclase (sAC) is highly expressed in the ciliary body in NPE cells, but appears to be absent from drainage tissues. Pharmacologic inhibition of sAC in mice causes a significant increase in IOP due to a decrease in C(t) with no effect on inflow. In mice deficient in sAC IOP is elevated, and C(t) is decreased relative to wild-type mice. Pharmacologic inhibition of sAC did not alter IOP or C(t) in sAC-deficient mice. Based on these data we propose that the ciliary body can regulate C(t) and that sAC serves as a critical sensor of bicarbonate in the ciliary body regulating the secretion of substances into the AH that govern outflow facility independent of pressure.


Assuntos
Adenilil Ciclases/metabolismo , Bicarbonatos/metabolismo , Corpo Ciliar/enzimologia , Glaucoma/enzimologia , Pressão Intraocular , Adenilil Ciclases/genética , Animais , Humor Aquoso/enzimologia , Bestrofinas , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Glaucoma/epidemiologia , Glaucoma/genética , Humanos , Camundongos , Camundongos Knockout , Suínos
7.
Hum Mol Genet ; 19(6): 1108-18, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20053664

RESUMO

Mutations in BEST1, encoding bestrophin-1 (Best1), cause Best vitelliform macular dystrophy (BVMD), a dominantly inherited macular degeneration characterized by a diminished electrooculogram light peak (LP), lipofuscin in retinal pigment epithelial cells (RPE), and fluid- and debris-filled retinal detachments. To understand the pathogenesis of BVMD we generated knock-in mice carrying the BVMD-causing mutation W93C in Best1. Both Best1(+/W93C)and Best1(W93C/W93C) mice had normal ERG a- and b-waves, but exhibited an altered LP luminance response reminiscent of that observed in BVMD patients. Morphological analysis identified fluid- and debris-filled retinal detachments in mice as young as 6 months of age. By 18-24 months of age Best1(+/W93C)and Best1(W93C/W93C) mice exhibited enhanced accumulation of lipofuscin in the RPE, and a significant deposition of debris composed of unphagocytosed photoreceptor outer segments and lipofuscin granules in the subretinal space. Although Best1 is thought to function as a Ca(2+)-activated Cl(-) channel, RPE cells from Best1(W93C) mice exhibited normal Cl(-) conductances. We have previously shown that Best1(-/-) mice exhibit increased [Ca(2+)](i) in response to ATP stimulation. However, ATP-stimulated changes in [Ca(2+)](i) in RPE cells from Best1(+/W93C) and Best1(W93C/W93C) mice were suppressed relative to Best1(+/+) littermates. Based on these data we conclude that mice carrying the Best1(W93C) mutation are a valid model for BVMD. Furthermore, these data suggest that BVMD is not because of Best1 deficiency, as the phenotypes of Best1(+/W93C) and Best1(W93C/W93C) mice are distinct from that of Best1(-/-) mice with regard to lipofuscin accumulation, and changes in the LP and ATP Ca(2+) responses.


Assuntos
Sinalização do Cálcio , Modelos Animais de Doenças , Degeneração Macular/metabolismo , Trifosfato de Adenosina/farmacologia , Substituição de Aminoácidos/genética , Animais , Bestrofinas , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/efeitos da radiação , Cloretos/metabolismo , Eletroculografia , Eletrorretinografia , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Técnicas de Introdução de Genes , Genótipo , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/efeitos da radiação , Canais Iônicos , Luz , Degeneração Macular/genética , Degeneração Macular/patologia , Degeneração Macular/fisiopatologia , Camundongos , Proteínas Mutantes/metabolismo , Mutação/genética , Epitélio Pigmentado Ocular/metabolismo , Epitélio Pigmentado Ocular/patologia , Epitélio Pigmentado Ocular/efeitos da radiação , Epitélio Pigmentado Ocular/ultraestrutura
8.
Circ Res ; 106(3): 583-92, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20019329

RESUMO

RATIONALE: Loss of fibulin-4 during embryogenesis results in perinatal lethality because of aneurysm rupture, and defective elastic fiber assembly has been proposed as an underlying cause for the aneurysm phenotype. However, aneurysms are never seen in mice deficient for elastin, or for fibulin-5, which absence also leads to compromised elastic fibers. OBJECTIVE: We sought to determine the mechanism of aneurysm development in the absence of fibulin-4 and establish the role of fibulin-4 in aortic development. METHODS AND RESULTS: We generated germline and smooth muscle cell (SMC)-specific deletion of the fibulin-4 gene in mice (Fbln4(GKO) and Fbln4(SMKO), respectively). Fbln4(GKO) and Fbln4(SMKO) aortic walls fail to fully differentiate, exhibiting reduced expression of SM-specific contractile genes and focal proliferation of SMCs accompanied by degenerative changes of the medial wall. Marked upregulation of extracellular signal-regulated kinase 1/2 signaling pathway was observed in the aneurysmal wall of Fbln4(GKO) and Fbln4(SMKO) mice and both mutants developed aneurysm predominantly in the ascending thoracic aorta. In vitro, Fbln4(GKO) SMCs exhibit an immature SMC phenotype with a marked reduction of SM-myosin heavy chain and increased proliferative capacity. CONCLUSIONS: The vascular phenotype in Fbln4 mutant mice is remarkably similar to a subset of human thoracic aortic aneurysms caused by mutations in SMC contractile genes. Our study provides a potential link between the intrinsic properties of SMCs and aneurysm progression in vivo and supports the dual role of fibulin-4 in the formation of elastic fibers as well as terminal differentiation and maturation of SMCs in the aortic wall.


Assuntos
Aorta/patologia , Aneurisma Aórtico/genética , Proteínas da Matriz Extracelular/deficiência , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Animais , Aorta/embriologia , Aneurisma Aórtico/patologia , Diferenciação Celular , Cruzamentos Genéticos , Tecido Elástico/patologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/fisiologia , Feminino , Mutação em Linhagem Germinativa , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Especificidade de Órgãos , Túnica Média/patologia
9.
Proc Natl Acad Sci U S A ; 106(45): 19029-34, 2009 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-19855011

RESUMO

Great arteries, as well as lungs and skin, contain elastic fibers as important components to maintain their physiological functions. Although recent studies have revealed that a glycoprotein fibulin-4 (FBLN4) is indispensable for the assembly of mature elastic fibers, it remains to be elucidated how FBLN4 takes part in elastogenesis. Here, we report a dose-dependent requirement for FBLN4 in the development of the elastic fibers in arteries, and a specific role of FBLN4 in recruiting the elastin-cross-linking enzyme, lysyl oxidase (LOX). Reduced expression of Fbln4, which was achieved with a smooth muscle-specific Cre-mediated gene deletion, caused arterial stiffness. Electron-microscopic examination revealed disorganized thick elastic laminae with aberrant deposition of elastin. Aneurysmal dilation of the ascending aorta was found when the Fbln4 expression level was reduced to an even lower level, whereas systemic Fbln4 null mice died perinatally from rupture of the diaphragm. We also found a specific interaction between FBLN4 and the propeptide of LOX, which efficiently promotes assembly of LOX onto tropoelastin. These data suggest a mechanism of elastogenesis, in which a sufficient amount of FBLN4 is essential for tethering LOX to tropoelastin to facilitate cross-linking.


Assuntos
Artérias/metabolismo , Elastina/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Animais , Artérias/ultraestrutura , Proteínas da Matriz Extracelular/genética , Deleção de Genes , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Reação em Cadeia da Polimerase , Ligação Proteica
10.
Trends Genet ; 23(5): 225-31, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17368622

RESUMO

Macular degenerations (MD), age-related or inherited, interfere with the ability to read, drive and recognize faces. Understanding this class of diseases has been challenging because the mouse, the mammal most amenable to genetic manipulation, lacks a macula. Here we discuss whether we can model MD in the mouse, present criteria for an 'ideal' mouse model of MD and discuss how mouse models have contributed to our knowledge of MD by contrasting how well they meet the 'ideal' criteria with how informative they have actually been. By modeling MD in mice, we can learn about aspects of MD that an animal with a macula would be unable to teach us.


Assuntos
Modelos Animais de Doenças , Degeneração Macular/genética , Envelhecimento/patologia , Animais , Proteínas da Matriz Extracelular/genética , Proteínas do Olho/genética , Previsões , Proteínas de Filamentos Intermediários/genética , Degeneração Macular/patologia , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Periferinas , Proteínas Recombinantes/genética , Degeneração Retiniana/genética , Degeneração Retiniana/patologia
11.
Am J Pathol ; 174(1): 206-15, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19095964

RESUMO

Fibulin-5 is crucial for normal elastic fiber synthesis in the vaginal wall; more than 90% of fibulin-5-knockout mice develop pelvic organ prolapse by 20 weeks of age. In contrast, fibulin-1 and -2 deficiencies do not result in similar pathologies, and fibulin-4-knockout mice die shortly after birth. EFEMP1 encodes fibulin-3, an extracellular matrix protein important in the maintenance of abdominal fascia. Herein, we evaluated the role of fibulin-3 in pelvic organ support. Pelvic organ support was impaired significantly in female Efemp1 knockout mice (Fbln3(-[supi]/-)), and overt vaginal, perineal, and rectal prolapse occurred in 26.9% of animals. Prolapse severity increased with age but not parity. Fibulin-5 was up-regulated in vaginal tissues from Fbln3(-[supi]/-) mice regardless of prolapse. Despite increased expression of fibulin-5 in the vaginal wall, pelvic organ support failure occurred in Fbln3(-[supi]/-) animals, suggesting that factors related to aging led to prolapse. Elastic fiber abnormalities in vaginal tissues from young Fbln3(-[supi]/-) mice progressed to severe elastic fiber disruption with age, and vaginal matrix metalloprotease activity was increased significantly in Fbln3(-[supi]/-) animals with prolapse compared with Fbln3(-[supi]/-) mice without prolapse. Overall, these results indicate that both fibulin-3 and -5 are important in maintaining pelvic organ support in mice. We suggest that increased vaginal protease activity and abnormal elastic fibers in the vaginal wall are important components in the pathogenesis of pelvic organ prolapse.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Prolapso Uterino/metabolismo , Animais , Desmosina/metabolismo , Tecido Elástico/metabolismo , Tecido Elástico/patologia , Proteínas da Matriz Extracelular/genética , Feminino , Immunoblotting , Imuno-Histoquímica , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Proteínas Recombinantes/metabolismo , Prolapso Retal/genética , Prolapso Retal/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Prolapso Uterino/genética
12.
Mol Cell Biol ; 26(5): 1700-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16478991

RESUMO

Elastic fibers provide tissues with elasticity which is critical to the function of arteries, lungs, skin, and other dynamic organs. Loss of elasticity is a major contributing factor in aging and diseases. However, the mechanism of elastic fiber development and assembly is poorly understood. Here, we show that lack of fibulin-4, an extracellular matrix molecule, abolishes elastogenesis. fibulin-4-/- mice generated by gene targeting exhibited severe lung and vascular defects including emphysema, artery tortuosity, irregularity, aneurysm, rupture, and resulting hemorrhages. All the homozygous mice died perinatally. The earliest abnormality noted was a uniformly narrowing of the descending aorta in fibulin-4-/- embryos at embryonic day 12.5 (E12.5). Aorta tortuosity and irregularity became noticeable at E15.5. Histological analysis demonstrated that fibulin-4-/- mice do not develop intact elastic fibers but contain irregular elastin aggregates. Electron microscopy revealed that the elastin aggregates are highly unusual in that they contain evenly distributed rod-like filaments, in contrast to the amorphous appearance of normal elastic fibers. Desmosine analysis indicated that elastin cross-links in fibulin-4-/- tissues were largely diminished. However, expression of tropoelastin or lysyl oxidase mRNA was unaffected in fibulin-4-/- mice. In addition, fibulin-4 strongly interacts with tropoelastin and colocalizes with elastic fibers in culture. These results demonstrate that fibulin-4 plays an irreplaceable role in elastogenesis.


Assuntos
Tecido Elástico/fisiologia , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Morte Fetal/genética , Animais , Aorta/anormalidades , Aorta/embriologia , Células Cultivadas , Desmosina/metabolismo , Tecido Elástico/anormalidades , Elastina/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Inativação Gênica , Humanos , Pulmão/anormalidades , Pulmão/embriologia , Pulmão/patologia , Camundongos , Camundongos Mutantes , Proteína-Lisina 6-Oxidase/metabolismo , Tropoelastina/metabolismo
13.
Invest Ophthalmol Vis Sci ; 49(4): 1563-70, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18385076

RESUMO

PURPOSE: The bestrophin family of proteins has been demonstrated to generate or regulate Ca2+-activated Cl(-) conductances. Mutations in bestrophin-1 (Best1) cause several blinding eye diseases, but little is known about other bestrophin family members. This study involved disruption of the Best2 gene in mice. METHODS: The mouse Best2 gene was disrupted by replacing exons 1, 2, and part of exon 3 with a Lac Z. The expression profile of Bestrophin-2 (Best2) was examined using RT-PCR, X-gal staining, and immunohistochemistry. Intraocular pressure (IOP) was measured by anterior chamber cannulation. RESULTS: RT-PCR of mouse tissues revealed Best2 mRNA in eye, colon, nasal epithelia, trachea, brain, lung, and kidney. X-gal staining, confirmed expression in colon epithelia and in the eye, in the nonpigmented epithelia (NPE). Best2 was not expressed in RPE cells. Best2 protein was observed only in NPE and colon epithelia. The absence of Best2 had no obvious deleterious effect on the mice. However, the Best2-/- mice were found to have significantly (P < 0.02) diminished IOP with respect to the Best2+/+ and Best2+/- littermates. The Best2-/- and Best2+/- mice responded better to the carbonic anhydrase inhibitor brinzolamide than did their Best2+/+ littermates, although the beta-blocker timolol brought IOP to the same level, regardless of genotype. CONCLUSIONS: Best2 plays a role in the generation of IOP by regulating formation of aqueous humor, and inhibition of Best2 function represents an attractive new avenue for regulating IOP in individuals with glaucoma.


Assuntos
Canais de Cloreto/fisiologia , Pressão Intraocular/fisiologia , Animais , Humor Aquoso/metabolismo , Bestrofinas , Western Blotting , Corpo Ciliar/citologia , Células Epiteliais/metabolismo , Expressão Gênica/fisiologia , Perfilação da Expressão Gênica , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
14.
Sci Rep ; 8(1): 4487, 2018 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-29540715

RESUMO

Autosomal recessive bestrophinopathy (ARB) is caused by mutations in the gene BEST1 which encodes bestrophin 1 (Best1), an anion channel expressed in retinal pigment epithelial (RPE) cells. It has been hypothesized that ARB represents the human null phenotype for BEST1 and that this occurs due to nonsense mediated decay (NMD). To test this hypothesis, we generated induced pluripotent stem cells (iPSCs) from a patient with ARB and her parents. After differentiation to retinal pigment epithelial (iPSC-RPE) cells, both BEST1 mRNA and Best1 protein expression were compared to controls. BEST1 mRNA expression levels, determined by quantitative PCR, were similar in ARB iPSC-RPE, parental cells, and genetically unrelated controls. Western blotting revealed that CRALBP and RPE65 were expressed within the range delineated by unrelated controls in iPSC-RPE from the ARB donor and her parents. Best1 protein was detected in different clones of ARB iPSC-RPE, but at reduced levels compared to all controls. When tested for the ability to phagocytose photoreceptor outer segments, ARB iPSC-RPE exhibited impaired internalization. These data suggest that impaired phagocytosis is a trait common to the bestrophinopathies. Furthermore, ARB is not universally the result of NMD and ARB, in this patient, is not due to the absence of Best1.


Assuntos
Bestrofinas/genética , Oftalmopatias Hereditárias/genética , Expressão Gênica , Genes Recessivos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação , Fagocitose/genética , Doenças Retinianas/genética , Adolescente , Alelos , Bestrofinas/metabolismo , Diferenciação Celular , Linhagem Celular , Oftalmopatias Hereditárias/diagnóstico , Feminino , Angiofluoresceinografia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fenótipo , Doenças Retinianas/diagnóstico , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo
15.
Biochim Biophys Acta ; 1758(2): 241-7, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16600174

RESUMO

Bestrophin-1 (Best-1) is an integral membrane protein, defects in which cause Best vitelliform macular dystrophy. Best-1 is proposed to function as a Cl- channel and/or a regulator of Ca++ channels. A tetrameric (or pentameric) stoichiometry has been reported for recombinant best-1. Using a combination of gel exclusion chromatography and velocity sedimentation we examined the quaternary structure of native best-1 and found that it migrates as a single species with a Stokes radius of 7.3 nm, sedimentation coefficient (S20,w) of 4.9, and partial specific volume (nu) of 0.80 ml/g. The mass of the protein-detergent complex is calculated to be 206 kDa, with the protein component estimated to be approximately 138 kDa. Given a monomeric mass of 68 kDa, we conclude that native best-1 solubilized with Triton X-100 is a homodimer. The differences between this observation and a prior report were examined by comparing recombinant best-1 with tissue derived best-1 using gel exclusion chromatography. Much of the recombinant best-1 eluted in the column void (Vo) fraction, unlike that extracted from RPE cells. We conclude that the minimal functional unit of best-1 is dimeric. This stoichiometry differs from that previously measured for recombinant best-1, suggesting that further studies are necessary to determine the stoichiometry of functional best-1 in RPE membranes.


Assuntos
Canais de Cloreto/química , Animais , Linhagem Celular , Fenômenos Químicos , Físico-Química , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Distrofias Hereditárias da Córnea/genética , Detergentes , Proteínas do Olho/química , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Humanos , Técnicas In Vitro , Masculino , Peso Molecular , Octoxinol , Epitélio Pigmentado Ocular/química , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sus scrofa , Distribuição Tecidual , Transfecção
16.
J Gen Physiol ; 127(5): 577-89, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16636205

RESUMO

Mutations in VMD2, encoding bestrophin (best-1), cause Best vitelliform macular dystrophy (BMD), adult-onset vitelliform macular dystrophy (AVMD), and autosomal dominant vitreoretinochoroidopathy (ADVIRC). BMD is distinguished from AVMD by a diminished electrooculogram light peak (LP) in the absence of changes in the flash electroretinogram. Although the LP is thought to be generated by best-1, we find enhanced LP luminance responsiveness with normal amplitude in Vmd2-/- mice and no differences in cellular Cl- currents in comparison to Vmd2+/+ littermates. The putative Ca2+ sensitivity of best-1, and our recent observation that best-1 alters the kinetics of voltage-dependent Ca2+ channels (VDCC), led us to examine the role of VDCCs in the LP. Nimodipine diminished the LP, leading us to survey VDCC beta-subunit mutant mice. Lethargic mice, which harbor a loss of function mutation in the beta4 subunit of VDCCs, exhibited a significant shift in LP luminance response, establishing a role for Ca2+ in LP generation. When stimulated with ATP, which increases [Ca++]I, retinal pigment epithelial cells derived from Vmd2-/- mice exhibited a fivefold greater response than Vmd2+/+ littermates, indicating that best-1 can suppress the rise in [Ca2+]I associated with the LP. We conclude that VDCCs regulated by a beta4 subunit are required to generate the LP and that best-1 antagonizes the LP luminance response potentially via its ability to modulate VDCC function. Furthermore, we suggest that the loss of vision associated with BMD is not caused by the same pathologic process as the diminished LP, but rather is caused by as yet unidentified effects of best-1 on other cellular processes.


Assuntos
Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/fisiologia , Eletrorretinografia/métodos , Proteínas do Olho/fisiologia , Luz , Trifosfato de Adenosina/farmacologia , Animais , Bestrofinas , Cálcio/análise , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cloreto/fisiologia , Eletrofisiologia , Proteínas do Olho/genética , Imuno-Histoquímica , Canais Iônicos , Camundongos , Camundongos Mutantes , Mutação , Nimodipina/farmacologia , Técnicas de Patch-Clamp , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Invest Ophthalmol Vis Sci ; 58(3): 1455-1461, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28264101

RESUMO

Purpose: EFEMP1 (fibulin-3) is mutated in Malattia Leventinese/Doyne's honeycomb retinal dystrophy (ML/DHRD), an inherited macular dystrophy similar to AMD. Both ML/DHRD and AMD are characterized by the presence of sub-RPE deposits. Efemp1 knockout mice do not develop sub-RPE deposits. This study was to test whether sub-RPE deposits can be induced in Efemp1 knockout mice by experimentally applied stress conditions that cause wild-type mice to develop sub-RPE deposits. Methods: Efemp1 knockout and control mice at 6, 18, or 24 months old were fed with a synthetic high-fat diet (HFD). Beginning 1 month after starting the HFD, one group of mice was exposed to cigarette smoke daily for 1 month, and another group of mice was subjected to photochemical injury every other day for 2 weeks from a 488-nm argon laser. After the treatments, histologic analysis was performed to assess whether sub-RPE deposits were induced. Results: Basal laminar deposits (BLamDs), a form of sub-RPE deposits, were observed in the 18- and 24-month-old wild-type mice but not in Efemp1 knockout mice in any age groups after exposure to HFD and cigarette smoke or laser injury. Conclusions: Mice lacking fibulin-3 do not develop sub-RPE deposits. Environmental oxidative stressors (HFD/cigarette smoke or HFD/laser) known to cause BLamD formation in wild-type mice failed to induce BLamD formation in Efemp1 knockout mice. These results suggest that fibulin-3 is a central player in the development of BLamD, and deletion of fibulin-3 is protective against the development of BLamD.


Assuntos
DNA/genética , Proteínas da Matriz Extracelular/genética , Degeneração Macular/genética , Mutação , Epitélio Pigmentado Ocular/metabolismo , Animais , Análise Mutacional de DNA , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/metabolismo , Seguimentos , Deleção de Genes , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Camundongos , Camundongos Knockout , Estresse Oxidativo , Epitélio Pigmentado Ocular/patologia , Fatores de Tempo
18.
Invest Ophthalmol Vis Sci ; 58(7): 3046-3054, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28622396

RESUMO

Purpose: The mutation R345W in EFEMP1 (fibulin-3) causes macular degeneration. This study sought to determine whether proteoglycan content and diffusion across Bruch's membrane are altered in Efemp1ki/ki mice carrying this mutation or in Efemp1-/- mice. Methods: Proteoglycans in mouse Bruch's membranes were stained with Cupromeronic Blue (CB). Heparan sulfated proteoglycan (HSPG) and chondroitin/dermatan sulfate proteoglycan (C/DSPG) distributions were visualized following treatments with chondroitinase ABC (C-ABC) or nitrous acid. Total sulfated glycosaminoglycans (sGAGs) in Bruch's membrane/choroid (BrM/Ch) were measured with dimethylmethylene blue (DMMB). Matrix metalloprotease (MMP)-2, MMP-9, and tissue inhibitor of metalloproteinase (TIMP)-3 were examined by immunofluorescence and quantified using Image J. Molecules with different Stokes radius (Rs) were allowed simultaneously to diffuse through mouse BrM/Ch mounted in a modified Ussing chamber. Samples were quantified using gel exclusion chromatography. Results: HSPGs and C/DSPGs were markedly increased in Efemp1ki/ki Bruch's membrane, and MMP-2 and MMP-9 were decreased, but TIMP-3 was increased. Diffusion across Efemp1ki/ki Bruch's membrane was impaired. In contrast, the proteoglycan amount in Efemp1-/- Bruch's membrane was not significantly different, but the size of proteoglycans was much larger. MMP-2, MMP-3, and TIMP-3 levels were similar to that of Efemp1+/+ mice, but they were localized diffusely in retinal pigment epithelium (RPE) cells instead of Bruch's membrane. Diffusion across Efemp1-/- Bruch's membrane was enhanced. Conclusions: Mutant fibulin-3 causes proteoglycan accumulation, reduction of MMP-2 and MMP-9, but increase of TIMP-3, and impairs diffusion across Bruch's membrane. Fibulin-3 ablation results in altered sizes of proteoglycans, altered distributions of MMP-2, MMP-9, and TIMP-3, and enhances diffusion across Bruch's membrane.


Assuntos
Lâmina Basilar da Corioide/metabolismo , DNA/genética , Proteínas da Matriz Extracelular/genética , Degeneração Macular/genética , Mutação , Proteoglicanas/metabolismo , Envelhecimento/genética , Animais , Lâmina Basilar da Corioide/patologia , Análise Mutacional de DNA , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/metabolismo , Degeneração Macular/diagnóstico , Degeneração Macular/metabolismo , Camundongos , Camundongos Mutantes
19.
Prog Retin Eye Res ; 58: 45-69, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28153808

RESUMO

Mutations in the gene BEST1 are causally associated with as many as five clinically distinct retinal degenerative diseases, which are collectively referred to as the "bestrophinopathies". These five associated diseases are: Best vitelliform macular dystrophy, autosomal recessive bestrophinopathy, adult-onset vitelliform macular dystrophy, autosomal dominant vitreoretinochoroidopathy, and retinitis pigmentosa. The most common of these is Best vitelliform macular dystrophy. Bestrophin 1 (Best1), the protein encoded by the gene BEST1, has been the subject of a great deal of research since it was first identified nearly two decades ago. Today we know that Best1 functions as both a pentameric anion channel and a regulator of intracellular Ca2+ signaling. Best1 is an integral membrane protein which, within the eye, is uniquely expressed in the retinal pigment epithelium where it predominantly localizes to the basolateral plasma membrane. Within the brain, Best1 expression has been documented in both glial cells and astrocytes where it functions in both tonic GABA release and glutamate transport. The crystal structure of Best1 has revealed critical information about how Best1 functions as an ion channel and how Ca2+ regulates that function. Studies using animal models have led to critical insights into the physiological roles of Best1 and advances in stem cell technology have allowed for the development of patient-derived, "disease in a dish" models. In this article we review our knowledge of Best1 and discuss prospects for near-term clinical trials to test therapies for the bestrophinopathies, a currently incurable and untreatable set of diseases.


Assuntos
Bestrofinas/genética , DNA/genética , Mutação , Doenças Retinianas/genética , Animais , Bestrofinas/metabolismo , Análise Mutacional de DNA , Modelos Animais de Doenças , Humanos , Camundongos , Doenças Retinianas/metabolismo
20.
Arthritis Rheumatol ; 69(3): 576-585, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27780308

RESUMO

OBJECTIVE: The EFEMP1 gene encoding fibulin 3 is specifically expressed in the superficial zone (SZ) of articular cartilage. The aims of this study were to examine the expression patterns of fibulin 3 in the knee joints during aging and during osteoarthritis (OA) and to determine the role of fibulin 3 in the pathogenesis of OA. METHODS: Immunohistochemical analysis was performed on normal and OA knee cartilage samples from humans and mice. Experimental OA was induced in wild-type and fibulin 3-/- mice, and the severity of OA was evaluated by histologic scoring. To examine fibulin 3 function, human chondrocyte monolayer cultures were transfected with small interfering RNA (siRNA), followed by quantitative polymerase chain reaction and Western blot analyses. Human bone marrow-derived mesenchymal stem cells (BM-MSCs) were transduced with an EFEMP1 lentivirus and analyzed for markers of chondrogenesis. RESULTS: Fibulin 3 was specifically expressed in the SZ of normal knee joint cartilage from humans and mice, and the expression levels declined with aging. Both aging-related OA and experimental OA were significantly more severe in fibulin 3-/- mice compared with wild-type mice. Fibulin 3 expression was high in undifferentiated human BM-MSCs and decreased during chondrogenesis. Suppression of fibulin 3 by siRNA significantly increased the expression of SOX9, type II collagen, and aggrecan in human articular chondrocytes, while overexpression of fibulin 3 inhibited chondrogenesis in BM-MSCs. CONCLUSION: Fibulin 3 is specifically expressed in the SZ of articular cartilage and its expression is reduced in aging and OA. Fibulin 3 regulates differentiation of adult progenitor cells, and its aging-related decline is an early event in the pathogenesis of OA. Preventing aging-associated loss of fibulin 3 or restoring it to normal levels in SZ chondrocytes has the potential to delay or prevent the onset of OA.


Assuntos
Envelhecimento , Cartilagem Articular , Proteínas da Matriz Extracelular/fisiologia , Osteoartrite do Joelho/etiologia , Adulto , Animais , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Condrócitos , Proteínas da Matriz Extracelular/biossíntese , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Osteoartrite do Joelho/metabolismo , Osteoartrite do Joelho/patologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA