Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Genet Metab Rep ; 26: 100712, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33552906

RESUMEN

Congenital corneal opacities (CCO) are a group of blinding corneal disorders, where the underlying molecular mechanisms are poorly understood. Phenotyping through specialized imaging and histopathology analysis, together with assessment of key transcriptomic changes (including glycosaminoglycan metabolic enzymes) in cornea(s) with CCO from a case of Fanconi anemia is the approach taken in this study to identify causal mechanisms. Based on our findings, we propose a novel mechanism and two key players contributing to CCO.

2.
Ophthalmic Genet ; 39(6): 735-740, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30444160

RESUMEN

BACKGROUND: Posterior column ataxia retinitis pigmentosa (PCARP) with feline leukemia virus subgroup C cellular receptor 1 (FLVCR1) gene mutation is a rare disorder with significant ophthalmic features. MATERIALS AND METHODS: We conducted a retrospective case series study of patients diagnosed with PCARP and genetic testing positive for FLVCR1 mutation between 1 January 2015 and 1 October 2017 at the Children's Hospital of Pittsburgh. Clinical charts, visual fields, fundus autofluorescence, and spectral-domain optical coherence tomography (SD-OCT) were reviewed. RESULTS: Seven patients from three families were identified to have PCARP and FLVCR1 mutation. The median age at presentation was 13 years (range, 7-28 years). Common clinical exam findings were astigmatism, cataracts, and vitreous syneresis. Funduscopy on all patients revealed bull's eye maculopathy, retinal vessels attenuation, and bone spicule changes in the peripheral retina. Fundus autofluorescence showed bilateral hyperautofluorescent rings. SD-OCT demonstrated morphological changes, which differed based on age. The youngest sibling family exhibited peripheral loss, but subfoveal preservation of the outer retinal layers. These layers were lost in the oldest sibling family. Visual fields loss paralleled SD-OCT findings. CONCLUSION: There is limited published ophthalmic data on FLVCR1-related PCARP. We describe clinical and retinal imaging features in the one of the largest cohorts of affected patients in the literature. Given the availability of genetic testing for this phenotype, testing for FLVCR1 mutations should be considered in pediatric and adult patients with sensory ataxia and retinitis pigmentosa.


Asunto(s)
Ataxia/diagnóstico por imagen , Ataxia/genética , Proteínas de Transporte de Membrana/genética , Mutación , Receptores Virales/genética , Retinitis Pigmentosa/diagnóstico por imagen , Retinitis Pigmentosa/genética , Tomografía de Coherencia Óptica , Adolescente , Adulto , Astigmatismo/diagnóstico , Astigmatismo/genética , Niño , Femenino , Humanos , Hiperopía/diagnóstico , Hiperopía/genética , Masculino , Biología Molecular , Imagen Multimodal , Miopía/diagnóstico , Miopía/genética , Imagen Óptica , Estudios Retrospectivos , Pruebas del Campo Visual , Campos Visuales/fisiología , Adulto Joven
3.
Am J Ophthalmol Case Rep ; 10: 172-179, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29780932

RESUMEN

PURPOSE: To investigate the association between novel PAX6 mutations to bilateral anterior pyramidal congenital cataracts (APyC), complete and intact irides, and nystagmus. OBSERVATIONS: This is a retrospective observational case series in a multi-center setting with genetic testing. Three female patients were diagnosed with bilateral APyC, intact irides and nystagmus. Genetic testing identified the three patients had novel missense mutations in PAX6 - c.128C > T; p.Ser43Phe (S43F), c. 197T > A; p.Ile66Asn (I66N) and c.781C > G; p.Arg261Gly (R261G). CONCLUSIONS AND IMPORTANCE: This study demonstrates a novel phenotype of bilateral APyC, intact irides, and nystagmus in whom genetic testing for PAX6 identified novel missense mutations (S43F, I66N, R261G) in highly conserved DNA-binding domains. Implications of understanding why the iris is present in these cases is discussed.

4.
Elife ; 62017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29035697

RESUMEN

The underground environment imposes unique demands on life that have led subterranean species to evolve specialized traits, many of which evolved convergently. We studied convergence in evolutionary rate in subterranean mammals in order to associate phenotypic evolution with specific genetic regions. We identified a strong excess of vision- and skin-related genes that changed at accelerated rates in the subterranean environment due to relaxed constraint and adaptive evolution. We also demonstrate that ocular-specific transcriptional enhancers were convergently accelerated, whereas enhancers active outside the eye were not. Furthermore, several uncharacterized genes and regulatory sequences demonstrated convergence and thus constitute novel candidate sequences for congenital ocular disorders. The strong evidence of convergence in these species indicates that evolution in this environment is recurrent and predictable and can be used to gain insights into phenotype-genotype relationships.


Asunto(s)
Adaptación Biológica , Evolución Biológica , Ecosistema , Ojo , Mamíferos , Fenómenos Fisiológicos Oculares , Animales
5.
Expert Rev Ophthalmol ; 11(2): 135-144, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-30100922

RESUMEN

Congenital aniridia manifests as total or partial absence of the iris caused most commonly by mutations in PAX6, FOXC1, PITX2, and CYP1B1. Recently two new genes, FOXD3 and TRIM44, have also been implicated in isolated studies. We discuss the genotype-phenotype correlations for the main implicated genes. Classic aniridia is a panocular condition, which includes aniridia, cataract, corneal pannus, foveal, and optic nerve hypoplasia associated with mutations in the PAX6 gene. Classical aniridia is due to PAX6 mutations, while other genes contribute to aniridia-like phenotypes. We review the challenges involved in the management of aniridia, and discuss various surgical interventions. The clinical importance of defining the genotype in cases of congenital aniridia has become acutely apparent with the advent of possible therapies for classical aniridia, which are discussed.

6.
Plast Reconstr Surg Glob Open ; 3(6): e427, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26180728

RESUMEN

Craniofrontonasal syndrome (CFNS) is a rare X-linked disorder that shows greater severity in females and is largely attributed to mutations in EFNB1. A 7-year-old boy presented with hypertelorism, broad nasal root, midfacial hypoplasia, mandibular prognathia, ptosis, and scaphocephaly was clinically diagnosed with CFNS. Three-dimensional computed tomographic scans confirmed the isolated sagittal synostosis. His mother also showed clinical features of CFNS, but less severe. Genetic tests uncovered a novel C to T mutation at nucleotide 466 (c.466C>T) in exon 1 of EFNB1 for both. To the best of our knowledge, this is the only reported incident of CFNS in a male child exhibiting isolated sagittal synostosis.

7.
Proc Natl Acad Sci U S A ; 108(45): 18289-94, 2011 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22021442

RESUMEN

Epithelial bending is a central feature of morphogenesis in animals. Here we show that mutual antagonism by the small Rho GTPases Rac1 and RhoA determines cell shape, tissue curvature, and invagination activity in the model epithelium of the developing mouse lens. The epithelial cells of the invaginating lens placode normally elongate and change from a cylindrical to an apically constricted, conical shape. RhoA mutant lens placode cells are both longer and less apically constricted than control cells, thereby reducing epithelial curvature and invagination. By contrast, Rac1 mutant lens placode cells are shorter and more apically restricted than controls, resulting in increased epithelial curvature and precocious lens vesicle closure. Quantification of RhoA- and Rac1-dependent pathway markers over the apical-basal axis of lens pit cells showed that in RhoA mutant epithelial cells there was a Rac1 pathway gain of function and vice versa. These findings suggest that mutual antagonism produces balanced activities of RhoA-generated apical constriction and Rac1-dependent cell elongation that controls cell shape and thus curvature of the invaginating epithelium. The ubiquity of the Rho family GTPases suggests that these mechanisms are likely to apply generally where epithelial morphogenesis occurs.


Asunto(s)
Forma de la Célula/fisiología , Células Epiteliales/citología , Morfogénesis , Proteína de Unión al GTP rac1/fisiología , Proteína de Unión al GTP rhoA/fisiología , Animales , Movimiento Celular , Cristalino/metabolismo , Ratones , Mutación , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rhoA/genética
8.
Development ; 138(23): 5177-88, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22031541

RESUMEN

Epithelial invagination is a common feature of embryogenesis. An example of invagination morphogenesis occurs during development of the early eye when the lens placode forms the lens pit. This morphogenesis is accompanied by a columnar-to-conical cell shape change (apical constriction or AC) and is known to be dependent on the cytoskeletal protein Shroom3. Because Shroom3-induced AC can be Rock1/2 dependent, we hypothesized that during lens invagination, RhoA, Rock and a RhoA guanine nucleotide exchange factor (RhoA-GEF) would also be required. In this study, we show that Rock activity is required for lens pit invagination and that RhoA activity is required for Shroom3-induced AC. We demonstrate that RhoA, when activated and targeted apically, is sufficient to induce AC and that RhoA plays a key role in Shroom3 apical localization. Furthermore, we identify Trio as a RhoA-GEF required for Shroom3-dependent AC in MDCK cells and in the lens pit. Collectively, these data indicate that a Trio-RhoA-Shroom3 pathway is required for AC during lens pit invagination.


Asunto(s)
Forma de la Célula/fisiología , Células Epiteliales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Cristalino/embriología , Proteínas de Microfilamentos/metabolismo , Morfogénesis/fisiología , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Proteínas de Unión al GTP rho/metabolismo , Animales , Línea Celular , Embrión de Pollo , Crioultramicrotomía , Perros , Electroporación , Técnica del Anticuerpo Fluorescente , Ratones , Análisis de Regresión , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
9.
Dev Biol ; 360(1): 30-43, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21945075

RESUMEN

Morphogenesis and shape of the ocular lens depend on epithelial cell elongation and differentiation into fiber cells, followed by the symmetric and compact organization of fiber cells within an enclosed extracellular matrix-enriched elastic capsule. The cellular mechanisms orchestrating these different events however, remain obscure. We investigated the role of the Rac1 GTPase in these processes by targeted deletion of expression using the conditional gene knockout (cKO) approach. Rac1 cKO mice were derived from two different Cre (Le-Cre and MLR-10) transgenic mice in which lens-specific Cre expression starts at embryonic day 8.75 and 10.5, respectively, in both the lens epithelium and fiber cells. The Le-Cre/Rac1 cKO mice exhibited an early-onset (E12.5) and severe lens phenotype compared to the MLR-10/Rac1 cKO (E15.5) mice. While the Le-Cre/Rac1 cKO lenses displayed delayed primary fiber cell elongation, lenses from both Rac1 cKO strains were characterized by abnormal shape, impaired secondary fiber cell migration, sutural defects and thinning of the posterior capsule which often led to rupture. Lens fiber cell N-cadherin/ß-catenin/Rap1/Nectin-based cell-cell junction formation and WAVE-2/Abi-2/Nap1-regulated actin polymerization were impaired in the Rac1 deficient mice. Additionally, the Rac1 cKO lenses were characterized by a shortened epithelial sheet, reduced levels of extracellular matrix (ECM) proteins and increased apoptosis. Taken together, these data uncover the essential role of Rac1 GTPase activity in establishment and maintenance of lens shape, suture formation and capsule integrity, and in fiber cell migration, adhesion and survival, via regulation of actin cytoskeletal dynamics, cell adhesive interactions and ECM turnover.


Asunto(s)
Cristalino/embriología , Neuropéptidos/deficiencia , Proteínas de Unión al GTP rac/deficiencia , Actinas/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Comunicación Celular/genética , Comunicación Celular/fisiología , Movimiento Celular/genética , Movimiento Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Citoesqueleto/metabolismo , Células Epiteliales/patología , Células Epiteliales/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Cápsula del Cristalino/anomalías , Cápsula del Cristalino/citología , Cápsula del Cristalino/embriología , Cápsula del Cristalino/fisiología , Cristalino/anomalías , Cristalino/citología , Cristalino/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Neuropéptidos/genética , Neuropéptidos/fisiología , Fenotipo , Embarazo , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/fisiología , Proteína de Unión al GTP rac1
10.
J Biol Chem ; 286(17): 15132-7, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21454503

RESUMEN

RhoA, the founding member of mammalian Rho GTPase family, is thought to be essential for actomyosin regulation. To date, the physiologic function of RhoA in mammalian cell regulation has yet to be determined genetically. Here we have created RhoA conditional knock-out mice. Mouse embryonic fibroblasts deleted of RhoA showed no significant change in actin stress fiber or focal adhesion complex formation in response to serum or LPA, nor any detectable change in Rho-kinase signaling activity. Concomitant knock-out or knockdown of RhoB and RhoC in the RhoA(-/-) cells resulted in a loss of actin stress fiber and focal adhesion similar to that of C3 toxin treatment. Proliferation of RhoA(-/-) cells was impaired due to a complete cell cycle block during mitosis, an effect that is associated with defective cytokinesis and chromosome segregation and can be readily rescued by exogenous expression of RhoA. Furthermore, RhoA deletion did not affect the transcriptional activity of Stat3, NFκB, or serum response factor, nor the expression of the cell division kinase inhibitor p21(Cip)1 or p27(Kip1). These genetic results demonstrate that in primary mouse embryonic fibroblasts, RhoA is uniquely required for cell mitosis but is redundant with related RhoB and RhoC GTPases in actomyosin regulation.


Asunto(s)
Actomiosina/metabolismo , Fibroblastos/citología , Mitosis , Proteínas de Unión al GTP rho/fisiología , Animales , Ciclo Celular , Células Cultivadas , Adhesiones Focales , Ratones , Fibras de Estrés , Transcripción Genética , Proteínas ras , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA , Proteína rhoC de Unión a GTP
11.
Proc Natl Acad Sci U S A ; 108(18): 7607-12, 2011 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21502507

RESUMEN

The organization of neural progenitors in the developing mammalian neuroepithelium is marked by cadherin-based adherens junctions. Whereas RhoA, a founding member of the small Rho GTPase family, has been shown to play important roles in epithelial adherens junctions, its physiological roles in neural development remain uncertain due to the lack of specific loss-of-function studies. Here, we show that RhoA protein accumulates at adherens junctions in the developing mouse brain and colocalizes to the cadherin-catenin complex. Conditional deletion of RhoA in midbrain and forebrain neural progenitors using Wnt1-Cre and Foxg1-Cre mice, respectively, disrupts apical adherens junctions and causes massive dysplasia of the brain. Furthermore, RhoA-deficient neural progenitor cells exhibit accelerated proliferation, reduction of cell- cycle exit, and increased expression of downstream target genes of the hedgehog pathway. Consequently, both lines of conditional RhoA-deficient embryos exhibit expansion of neural progenitor cells and exencephaly-like protrusions. These results demonstrate a critical role of RhoA in the maintenance of apical adherens junctions and the regulation of neural progenitor proliferation in the developing mammalian brain.


Asunto(s)
Uniones Adherentes/metabolismo , Encéfalo/embriología , Proliferación Celular , Células-Madre Neurales/metabolismo , Proteína de Unión al GTP rhoA/deficiencia , Animales , Bromodesoxiuridina , Inmunohistoquímica , Inmunoprecipitación , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Indoles , Ratones , Ratones Mutantes , Microscopía Confocal , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína de Unión al GTP rhoA/metabolismo
12.
Epigenetics Chromatin ; 3(1): 21, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21118511

RESUMEN

BACKGROUND: Brahma-related gene 1 (Brg1, also known as Smarca4 and Snf2ß) encodes an adenosine-5'-triphosphate (ATP)-dependent catalytical subunit of the (switch/sucrose nonfermentable) (SWI/SNF) chromatin remodeling complexes. SWI/SNF complexes are recruited to chromatin through multiple mechanisms, including specific DNA-binding factors (for example, heat shock transcription factor 4 (Hsf4) and paired box gene 6 (Pax6)), chromatin structural proteins (for example, high-mobility group A1 (HMGA1)) and/or acetylated core histones. Previous studies have shown that a single amino acid substitution (K798R) in the Brg1 ATPase domain acts via a dominant-negative (dn) mechanism. Genetic studies have demonstrated that Brg1 is an essential gene for early (that is, prior implantation) mouse embryonic development. Brg1 also controls neural stem cell maintenance, terminal differentiation of multiple cell lineages and organs including the T-cells, glial cells and limbs. RESULTS: To examine the roles of Brg1 in mouse lens development, a dnBrg1 transgenic construct was expressed using the lens-specific αA-crystallin promoter in postmitotic lens fiber cells. Morphological studies revealed abnormal lens fiber cell differentiation in transgenic lenses resulting in cataract. Electron microscopic studies showed abnormal lens suture formation and incomplete karyolysis (that is, denucleation) of lens fiber cells. To identify genes regulated by Brg1, RNA expression profiling was performed in embryonic day 15.5 (E15.5) wild-type and dnBrg1 transgenic lenses. In addition, comparisons between differentially expressed genes in dnBrg1 transgenic, Pax6 heterozygous and Hsf4 homozygous lenses identified multiple genes coregulated by Brg1, Hsf4 and Pax6. DNase IIß, a key enzyme required for lens fiber cell denucleation, was found to be downregulated in each of the Pax6, Brg1 and Hsf4 model systems. Lens-specific deletion of Brg1 using conditional gene targeting demonstrated that Brg1 was required for lens fiber cell differentiation, for expression of DNase IIß, for lens fiber cell denucleation and indirectly for retinal development. CONCLUSIONS: These studies demonstrate a cell-autonomous role for Brg1 in lens fiber cell terminal differentiation and identified DNase IIß as a potential direct target of SWI/SNF complexes. Brg1 is directly or indirectly involved in processes that degrade lens fiber cell chromatin. The presence of nuclei and other organelles generates scattered light incompatible with the optical requirements for the lens.

13.
Development ; 136(21): 3657-67, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19820184

RESUMEN

The vertebrate lens provides an excellent model with which to study the mechanisms required for epithelial invagination. In the mouse, the lens forms from the head surface ectoderm. A domain of ectoderm first thickens to form the lens placode and then invaginates to form the lens pit. The epithelium of the lens placode remains in close apposition to the epithelium of the presumptive retina as these structures undergo a coordinated invagination. Here, we show that F-actin-rich basal filopodia that link adjacent presumptive lens and retinal epithelia function as physical tethers that coordinate invagination. The filopodia, most of which originate in the presumptive lens, form at E9.5 when presumptive lens and retinal epithelia first come into close contact, and have retracted by E11.5 when invagination is complete. At E10.5--the lens pit stage--there is approximately one filopodium per epithelial cell. Formation of filopodia is dependent on the Rho family GTPase Cdc42 and the Cdc42 effector IRSp53 (Baiap2). Loss of filopodia results in reduced lens pit invagination. Pharmacological manipulation of the actin-myosin contraction pathway showed that the filopodia can respond rapidly in length to change inter-epithelial distance. These data suggest that the lens-retina inter-epithelial filopodia are a fine-tuning mechanism to assist in lens pit invagination by transmitting the forces between presumptive lens and retina. Although invagination of the archenteron in sea urchins and dorsal closure in Drosophila are known to be partly dependent on filopodia, this mechanism of morphogenesis has not previously been identified in vertebrates.


Asunto(s)
Cristalino/embriología , Seudópodos/metabolismo , Retina/embriología , Actinas/metabolismo , Animales , Quinasa 1 de Adhesión Focal/metabolismo , Cristalino/citología , Ratones , Miosinas/metabolismo , Retina/citología , Organismos Libres de Patógenos Específicos
14.
EMBO J ; 25(10): 2107-18, 2006 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-16675956

RESUMEN

Pax6 and c-Maf regulate multiple stages of mammalian lens development. Here, we identified novel distal control regions (DCRs) of the alphaA-crystallin gene, a marker of lens fiber cell differentiation induced by FGF-signaling. DCR1 stimulated reporter gene expression in primary lens explants treated with FGF2 linking FGF-signaling with alphaA-crystallin synthesis. A DCR1/alphaA-crystallin promoter (including DCR2) coupled with EGFP virtually recapitulated the expression pattern of alphaA-crystallin in lens epithelium and fibers. In contrast, the DCR3/alphaA/EGFP reporter was expressed only in 'late' lens fibers. Chromatin immunoprecipitations showed binding of Pax6 to DCR1 and the alphaA-crystallin promoter in lens chromatin and demonstrated that high levels of alphaA-crystallin expression correlate with increased binding of c-Maf and CREB to the promoter and of CREB to DCR3, a broad domain of histone H3K9-hyperacetylation extending from DCR1 to DCR3, and increased abundance of chromatin remodeling enzymes Brg1 and Snf2h at the alphaA-crystallin locus. Our data demonstrate a novel mechanism of Pax6, c-Maf and CREB function, through regulation of chromatin-remodeling enzymes, and suggest a multistage model for the activation of alphaA-crystallin during lens differentiation.


Asunto(s)
Cromatina/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Cristalino/crecimiento & desarrollo , Factores de Transcripción Paired Box/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Proteínas Represoras/metabolismo , Cadena A de alfa-Cristalina/metabolismo , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Diferenciación Celular/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , ADN Helicasas , Proteínas del Ojo/genética , Genes Reporteros , Histonas/metabolismo , Proteínas de Homeodominio/genética , Humanos , Cristalino/anatomía & histología , Cristalino/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-maf/genética , ARN Polimerasa II/metabolismo , Proteínas Represoras/genética , Transducción de Señal/fisiología , Técnicas de Cultivo de Tejidos , Factores de Transcripción/metabolismo , Transcripción Genética , Cadena A de alfa-Cristalina/genética
15.
BMC Ophthalmol ; 5: 14, 2005 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-15969763

RESUMEN

BACKGROUND: Paralemmin (Palm) is a prenyl-palmitoyl anchored membrane protein that can drive membrane and process formation in neurons. Earlier studies have shown brain preferred Palm expression, although this protein is a major water insoluble protein in chicken lens fiber cells and the Palm gene may be regulated by Pax6. METHODS: The expression profile of Palm protein in the embryonic, newborn and adult mouse eye as well as dissociated retinal neurons was determined by confocal immunofluorescence. The relative mRNA levels of Palm, Palmdelphin (PalmD) and paralemmin2 (Palm2) in the lens and retina were determined by real time rt-PCR. RESULTS: In the lens, Palm is already expressed at 9.5 dpc in the lens placode, and this expression is maintained in the lens vesicle throughout the formation of the adult lens. Palm is largely absent from the optic vesicle but is detectable at 10.5 dpc in the optic cup. In the developing retina, Palm expression transiently upregulates during the formation of optic nerve as well as in the formation of both the inner and outer plexiform layers. In short term dissociated chick retinal cultures, Palm protein is easily detectable, but the levels appear to reduce sharply as the cultures age. Palm mRNA was found at much higher levels relative to Palm2 or PalmD in both the retina and lens. CONCLUSION: Palm is the major paralemmin family member expressed in the retina and lens and its expression in the retina transiently upregulates during active neurite outgrowth. The expression pattern of Palm in the eye is consistent with it being a Pax6 responsive gene. Since Palm is known to be able to drive membrane formation in brain neurons, it is possible that this molecule is crucial for the increase in membrane formation during lens fiber cell differentiation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Cristalino/embriología , Proteínas de la Membrana/genética , Fosfoproteínas/genética , Retina/embriología , Animales , Animales Recién Nacidos , Embrión de Pollo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Cristalino/crecimiento & desarrollo , Cristalino/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Técnicas de Cultivo de Órganos , Fosfoproteínas/metabolismo , ARN Mensajero/metabolismo , Retina/crecimiento & desarrollo , Retina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
16.
J Neurosci ; 25(7): 1701-10, 2005 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-15716406

RESUMEN

Norrie disease is an X-linked retinal dysplasia that presents with congenital blindness, sensorineural deafness, and mental retardation. Norrin, the protein product of the Norrie disease gene (NDP), is a secreted protein of unknown biochemical function. Norrie disease (Ndp(y/-)) mutant mice that are deficient in norrin develop blindness, show a distinct failure in retinal angiogenesis, and completely lack the deep capillary layers of the retina. We show here that the transgenic expression of ectopic norrin under control of a lens-specific promoter restores the formation of a normal retinal vascular network in Ndp(y/-) mutant mice. The improvement in structure correlates with restoration of neuronal function in the retina. In addition, lenses of transgenic mice with ectopic expression of norrin show significantly more capillaries in the hyaloid vasculature that surrounds the lens during development. In vitro, lenses of transgenic mice in coculture with microvascular endothelial cells induce proliferation of the cells. Transgenic mice with ectopic expression of norrin show more bromodeoxyuridine-labeled retinal progenitor cells at embryonic day 14.5 and thicker retinas at postnatal life than wild-type littermates, indicating a putative direct neurotrophic effect of norrin. These data provide direct evidence that norrin induces growth of ocular capillaries and that pharmacologic modulation of norrin might be used for treatment of the vascular abnormalities associated with Norrie disease or other vascular disorders of the retina.


Asunto(s)
Proteínas del Ojo/fisiología , Neovascularización Fisiológica/fisiología , Proteínas del Tejido Nervioso/fisiología , Vasos Retinianos/crecimiento & desarrollo , Animales , Capilares/citología , Capilares/crecimiento & desarrollo , Recuento de Células , División Celular , Células Cultivadas , Pollos , Técnicas de Cocultivo , Electrorretinografía , Células Endoteliales/citología , Endotelio Vascular/citología , Proteínas del Ojo/genética , Regulación de la Expresión Génica , Humanos , Cristalino/citología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Retina/fisiopatología , Retina/ultraestructura , Células Ganglionares de la Retina/patología , Piel/irrigación sanguínea , beta-Cristalinas/genética
17.
Int J Dev Biol ; 48(8-9): 829-44, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15558475

RESUMEN

Lens development is an excellent model for genetic and biochemical studies of embryonic induction, cell cycle regulation, cellular differentiation and signal transduction. Differentiation of lens is characterized by lens-preferred expression and accumulation of water-soluble proteins, crystallins. Crystallins are required for light transparency, refraction and maintenance of lens integrity. Here, we review mechanisms of lens-preferred expression of crystallin genes by employing synergism between developmentally regulated DNA-binding transcription factors: Pax6, c-Maf, MafA/L-Maf, MafB, NRL, Sox2, Sox1, RARbeta/RXRbeta, RORalpha, Prox1, Six3, gammaFBP-B and HSF2. These factors are differentially expressed in lens precursor cells, lens epithelium and primary and secondary lens fibers. They exert their function in combination with ubiquitously expressed factors (e.g. AP-1, CREB, pRb, TFIID and USF) and co-activators/chromatin remodeling proteins (e.g. ASC-2 and CBP/p300). A special function belongs to Pax6, a paired domain and homeodomain-containing protein, which is essential for lens formation. Pax6 is expressed in lens progenitor cells before the onset of crystallin expression and it serves as an important regulatory factor required for expression of c-Maf, MafA/L-Maf, Six3, Prox1 and retinoic acid signaling both in lens precursor cells and the developing lens. The roles of these factors are illustrated by promoter studies of mouse alphaA-, alphaB-, gammaF- and guinea pig zeta-crystallins. Pax6 forms functional complexes with a number of transcription factors including the retinoblastoma protein, pRb, MafA, Mitf and Sox2. We present novel data showing that pRb antagonizes Pax6-mediated activation of the alphaA-crystallin promoter likely by inhibiting binding of Pax6 to DNA.


Asunto(s)
Proteínas del Ojo/fisiología , Ojo/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/fisiología , Proteínas Represoras/fisiología , Animales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Cromatina/metabolismo , Cristalinas/química , ADN/metabolismo , Heterocigoto , Humanos , Cristalino/embriología , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box , Unión Proteica , Estructura Terciaria de Proteína , Proteína de Retinoblastoma/metabolismo , Distribución Tisular , Factores de Transcripción/metabolismo , Transcripción Genética , Activación Transcripcional , Transgenes
18.
J Mol Biol ; 344(2): 351-68, 2004 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-15522290

RESUMEN

Mammalian alphaB-crystallin is highly expressed both in lens epithelium and lens fibers. In contrast, gammaF-crystallin is highly expressed in the lens fiber cells. Crystallin gene expression in lens is regulated at the level of transcription by a sparse number of specific DNA-binding transcription factors. Here, we report studies on transcriptional regulation of mouse alphaB- and gammaF-crystallin promoters by specific combinations of Pax6/Pax6(5a), large Mafs (MafA, MafB, c-Maf, and NRL), Sox1, Sox2, Six3, and RARbeta/RXRbeta. Two sets of these factors, co-expressed both in lens epithelium and in lens fibers, were tested in co-transfection assays using cultured lens and non-lens cells. Regulation of alphaB-crystallin was studied in the presence of lens epithelial-factors Pax6, MafB, and RARbeta/RXRbeta, and lens fiber-factors Pax6, MafA, c-Maf, and NRL. Pax6 proteins activated the alphaB-crystallin promoter (-162 to +45) with any combination of Mafs. Addition of RARbeta/RXRbeta further increased its promoter activity. Gel shift assays using lens nuclear extracts demonstrated interactions of Pax6, Maf, and retinoic acid nuclear receptor proteins with two lens-specific regions, the distal LSR1 (-147/-118) and proximal LSR2 (-78/-40), of the alphaB-crystallin promoter. In contrast, Pax6 proteins acted as repressors of gammaF-crystallin promoter activity elicited by a combination of large Mafs, Sox, and RARbeta/RXRbeta proteins in transiently transfected lens and non-lens cells. The results show that Pax6 conversely regulates these two lens crystallin promoters. We propose that the opposite roles of Pax6 in crystallin gene regulation are results of different promoter architectures of the alphaB- and gammaF-crystallin genes, developmentally regulated association of transcription factors with the corresponding cis-regulatory sites, and specific recruitment of transcriptional co-activators and co-repressors by Pax6.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Cristalino/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , alfa-Cristalinas/genética , gamma-Cristalinas/genética , Animales , Células CHO , Diferenciación Celular , Línea Celular , Células Cultivadas , Cricetinae , Cricetulus , Ensayo de Cambio de Movilidad Electroforética , Células Epiteliales/metabolismo , Proteínas del Ojo , Vectores Genéticos , Humanos , Cristalino/citología , Ratones , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box , Plásmidos , Proteínas Represoras , Factores de Transcripción/genética
19.
Nucleic Acids Res ; 32(5): 1696-709, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15020706

RESUMEN

Pax6 is essential for development of the eye, olfactory system, brain and pancreas. Haploinsufficiency of Pax6 causes abnormal eye development. Two forms of Pax6 protein, PAX6 and PAX6(5a), differ in a 14 amino acid insertion encoded by an alternatively spliced exon 5a in the N-terminal DNA-binding paired domain (PD), and they are simultaneously expressed. Here, we show that PAX6 and PAX6(5a) together synergistically activate transcription from promoters recognized by Pax6 PD and PD5a, but not by their homeodomain. This synergism promotes activation of transcription by c-Maf and MafA on the alphaB-crystallin promoter, and is required for transcriptional co-activation by RARbeta/RXRbeta and PAX6/PAX6(5a) on the gammaF-crystallin promoter. To determine the role of this synergism in haploinsufficiency, we tested four human missense (G18W, R26G, G64V and R128C) and one nonsense (R317X) mutants, with reporters driven by Pax6 PD consensus binding sites and the alphaB-crystallin promoter. The simultaneous activity of Pax6 proteins [PAX6, mutated PAX6, PAX6(5a) and mutated PAX6(5a)] modeling haploinsufficiency yielded results not predicted by properties of individual PAX6 or PAX6(5a). Taken together, these results indicate that complex ocular phenotypes due to Pax6 haploinsufficiency originate, at least partially, from functional interactions between alternatively spliced PAX6 and PAX6(5a) variants and other factors, e.g. MafA/c-Maf.


Asunto(s)
Cristalinas/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Empalme Alternativo , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas del Ojo , Heterocigoto , Proteínas de Homeodominio/genética , Humanos , Ratones , Mutación , Factor de Transcripción PAX5 , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box , Proteínas Represoras , Factores de Transcripción/genética , Cadena B de alfa-Cristalina/biosíntesis , Cadena B de alfa-Cristalina/genética , gamma-Cristalinas/biosíntesis , gamma-Cristalinas/genética
20.
Invest Ophthalmol Vis Sci ; 45(2): 385-92, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14744876

RESUMEN

PURPOSE: Pax6 is essential for development of the eye, brain, and pancreas. Two major products of PAX6 are specific DNA-binding proteins, PAX6 and PAX6(5a). PAX6(5a) contains a short insertion influencing its DNA-binding activity. Heterozygous mutations in PAX6 result in abnormal eye development implicating haploinsufficiency. Deletions of one PAX6 allele result in aniridia characterized by severe ocular phenotypes. Approximately 10% of PAX6 mutations encode missense mutations. These mutations usually cause less severe abnormalities than does aniridia. The moderate phenotypes raise the possibility that different ocular tissues are differently sensitive to specific mutations. To test this hypothesis, we probed functional properties of individual mutated Pax6 proteins in a variety of conditions. METHODS: Mutations in PAX6 and PAX6(5a) were introduced by site-directed mutagenesis and tested by transfections in four cell lines using reporters containing three different Pax6 binding sites. Pax6 binding to DNA was studied by electrophoretic mobility shift assays. RESULTS: Functional studies of PAX6 and PAX6(5a) and their eight natural missense (G18W, R26G, A33P, S43P, G64V, I87R, V126D and R128C) and two nonsense (R317X and S353X) disease-causing mutants revealed unexpected pleiotropic effects in gene regulation, not predicted by the PAX6-DNA crystal structure. Transactivation by PAX6 and PAX6(5a) was dependent on the location of mutation, type of DNA-binding site, and cellular environment. CONCLUSIONS: This work provides evidence that activation by PAX6 and PAX6(5a) is modulated by specific cellular environments. It is likely that moderate phenotypes associated with PAX6 missense mutations originate from abnormal protein function in a restricted number of ocular cell types.


Asunto(s)
Proteínas del Ojo/fisiología , Proteínas de Homeodominio/fisiología , Mutación Missense/fisiología , Factores de Transcripción/fisiología , Animales , Western Blotting , Células CHO/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Transformada , Codón sin Sentido , Cricetinae , Ensayo de Cambio de Movilidad Electroforética , Células Epiteliales/metabolismo , Humanos , Cristalino/citología , Mutagénesis Sitio-Dirigida , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box , Unión Proteica , Conejos , Proteínas Represoras , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...