Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(19)2023 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-37833870

RESUMEN

Pigmentary glaucoma has recently been associated with missense mutations in PMEL that are dominantly inherited and enriched in the protein's fascinating repeat domain. PMEL pathobiology is intriguing because PMEL forms functional amyloid in healthy eyes, and this PMEL amyloid acts to scaffold melanin deposition. This is an informative contradistinction to prominent neurodegenerative diseases where amyloid formation is neurotoxic and mutations cause a toxic gain of function called "amyloidosis". Preclinical animal models have failed to model this PMEL "dysamyloidosis" pathomechanism and instead cause recessively inherited ocular pigment defects via PMEL loss of function; they have not addressed the consequences of disrupting PMEL's repetitive region. Here, we use CRISPR to engineer a small in-frame mutation in the zebrafish homolog of PMEL that is predicted to subtly disrupt the protein's repetitive region. Homozygous mutant larvae displayed pigmentation phenotypes and altered eye morphogenesis similar to presumptive null larvae. Heterozygous mutants had disrupted eye morphogenesis and disrupted pigment deposition in their retinal melanosomes. The deficits in the pigment deposition of these young adult fish were not accompanied by any detectable glaucomatous changes in intraocular pressure or retinal morphology. Overall, the data provide important in vivo validation that subtle PMEL mutations can cause a dominantly inherited pigment pathology that aligns with the inheritance of pigmentary glaucoma patient pedigrees. These in vivo observations help to resolve controversy regarding the necessity of PMEL's repeat domain in pigmentation. The data foster an ongoing interest in an antithetical dysamyloidosis mechanism that, akin to the amyloidosis of devastating dementias, manifests as a slow progressive neurodegenerative disease.


Asunto(s)
Glaucoma de Ángulo Abierto , Enfermedades Neurodegenerativas , Animales , Humanos , Adulto Joven , Amiloide/metabolismo , Ojo/metabolismo , Glaucoma de Ángulo Abierto/metabolismo , Antígeno gp100 del Melanoma/genética , Melanosomas/genética , Melanosomas/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Pez Cebra
2.
Molecules ; 26(12)2021 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-34207849

RESUMEN

The ancient paralogs premelanosome protein (PMEL) and glycoprotein nonmetastatic melanoma protein B (GPNMB) have independently emerged as intriguing disease loci in recent years. Both proteins possess common functional domains and variants that cause a shared spectrum of overlapping phenotypes and disease associations: melanin-based pigmentation, cancer, neurodegenerative disease and glaucoma. Surprisingly, these proteins have yet to be shown to physically or genetically interact within the same cellular pathway. This juxtaposition inspired us to compare and contrast this family across a breadth of species to better understand the divergent evolutionary trajectories of two related, but distinct, genes. In this study, we investigated the evolutionary history of PMEL and GPNMB in clade-representative species and identified TMEM130 as the most ancient paralog of the family. By curating the functional domains in each paralog, we identified many commonalities dating back to the emergence of the gene family in basal metazoans. PMEL and GPNMB have gained functional domains since their divergence from TMEM130, including the core amyloid fragment (CAF) that is critical for the amyloid potential of PMEL. Additionally, the PMEL gene has acquired the enigmatic repeat domain (RPT), composed of a variable number of imperfect tandem repeats; this domain acts in an accessory role to control amyloid formation. Our analyses revealed the vast variability in sequence, length and repeat number in homologous RPT domains between craniates, even within the same taxonomic class. We hope that these analyses inspire further investigation into a gene family that is remarkable from the evolutionary, pathological and cell biology perspectives.


Asunto(s)
Melanocitos/metabolismo , Glicoproteínas de Membrana/metabolismo , Mutación , Enfermedades Neurodegenerativas/patología , Antígeno gp100 del Melanoma/metabolismo , Secuencia de Aminoácidos , Proteínas Amiloidogénicas/metabolismo , Animales , Biología Computacional/métodos , Humanos , Glicoproteínas de Membrana/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Filogenia , Pigmentación , Dominios Proteicos , Homología de Secuencia , Antígeno gp100 del Melanoma/genética
3.
Artículo en Inglés | MEDLINE | ID: mdl-33688250

RESUMEN

PURPOSE: Forkhead box Q1 (FOXQ1) has been shown to contribute to the development and progression of cancers, including ovarian and breast cancer (BC). However, research exploring FOXQ1 expression, copy number variation (CNV), and prognostic value across different BC subtypes is limited. Our purpose was to evaluate FOXQ1 mRNA expression, CNV, and prognostic value across BC subtypes. MATERIALS AND METHODS: We determined FOXQ1 expression and CNV in BC patient tumors using RT-qPCR and qPCR, respectively. We also analyzed FOXQ1 expression and CNV in BC cell lines in the CCLE database using K-means clustering. The prognostic value of FOXQ1 expression in the TCGA-BRCA database was assessed using univariate and multivariate Cox's regression analysis as well as using the online tools OncoLnc, GEPIA, and UALCAN. RESULTS: Our analyses reveal that FOXQ1 mRNA is differentially expressed between different subtypes of BC and is significantly decreased in luminal BC and HER2 patients when compared to normal breast tissue samples. Furthermore, analysis of BC cell lines showed that FOXQ1 mRNA expression was independent of CNV. Moreover, patients with low FOXQ1 mRNA expression had significantly poorer overall survival compared to those with high FOXQ1 mRNA expression. Finally, low FOXQ1 expression had a critical impact on the prognostic values of BC patients and was an independent predictor of overall survival when it was adjusted for BC subtypes and to two other FOX genes, FOXF2 and FOXM1. CONCLUSION: Our study reveals for the first time that FOXQ1 is differentially expressed across BC subtypes and that low expression of FOXQ1 is indicative of poor prognosis in patients with BC.

4.
Am J Med Genet C Semin Med Genet ; 184(3): 535-537, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32864823

RESUMEN

In this special issue of the American Journal of Medical Genetics, Part C, we explore the ever-expanding field of Ophthalmic Genetics. The eye is unique among organs for its accessibility to physical examination, permitting exploration of every tissue by slit lamp microscopy, ophthalmoscopy, and imaging including color and autofluorescent photography, ultrasound, optical coherence tomography (OCT), electrophysiology, and adaptive optics confocal and scanning laser ophthalmoscopy. This accessibility permits a variety of surgical and nonsurgical treatments, including the first FDA-approved gene therapy, voretigene neparvovec-rzyl for RPE65-associated Leber Congenital Amaurosis. In this issue, we sought to provide a survey highlighting how heritable ophthalmic disorders are recognizable and accessible to clinical geneticists as well as ophthalmologists.


Asunto(s)
Oftalmopatías/terapia , Terapia Genética , Oftalmología/tendencias , Tomografía de Coherencia Óptica/métodos , Oftalmopatías/diagnóstico por imagen , Oftalmopatías/genética , Oftalmopatías/patología , Humanos
5.
Am J Med Genet C Semin Med Genet ; 184(3): 860-868, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32896097

RESUMEN

Current genetic screening methods for inherited eye diseases are concentrated on the coding exons of known disease genes (gene panels, clinical exome). These tests have a variable and often limited diagnostic rate depending on the clinical presentation, size of the gene panel and our understanding of the inheritance of the disorder (with examples described in this issue). There are numerous possible explanations for the missing heritability of these cases including undetected variants within the relevant gene (intronic, up/down-stream and structural variants), variants harbored in genes outside the targeted panel, intergenic variants, variants undetectable by the applied technology, complex/non-Mendelian inheritance, and nongenetic phenocopies. In this article we further explore and review methods to investigate these sources of missing heritability.


Asunto(s)
Enfermedades Hereditarias del Ojo/diagnóstico , Enfermedades Hereditarias del Ojo/genética , Genoma Humano/genética , Genómica/métodos , Enfermedades Hereditarias del Ojo/epidemiología , Enfermedades Hereditarias del Ojo/terapia , Humanos , Oftalmología/tendencias
6.
Exp Eye Res ; 189: 107815, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31560925

RESUMEN

Aniridia and Axenfeld-Rieger Syndrome are related, human ocular disorders that are typically inherited in an autosomal dominant manner. Both result from incorrect development of the eye and have, as their most serious consequences, elevated risk to develop the blinding condition glaucoma. This review will focus on describing the clinical presentations of Aniridia and Axenfeld-Rieger Syndrome as well as the molecular genetics and current and emerging therapies used to treat patients.


Asunto(s)
Anomalías Múltiples , Aniridia/genética , Segmento Anterior del Ojo/anomalías , Anomalías del Ojo/genética , Enfermedades Hereditarias del Ojo/genética , Proteínas de Homeodominio/genética , Aniridia/diagnóstico , Aniridia/metabolismo , Segmento Anterior del Ojo/metabolismo , Anomalías del Ojo/diagnóstico , Anomalías del Ojo/metabolismo , Enfermedades Hereditarias del Ojo/diagnóstico , Enfermedades Hereditarias del Ojo/metabolismo , Genotipo , Proteínas de Homeodominio/metabolismo , Humanos , Mutación
7.
Hum Mol Genet ; 28(8): 1286-1297, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30561639

RESUMEN

Molecular mechanisms governing the development of the human cochlea remain largely unknown. Through genome sequencing, we identified a homozygous FOXF2 variant c.325A>T (p.I109F) in a child with profound sensorineural hearing loss (SNHL) associated with incomplete partition type I anomaly of the cochlea. This variant is not found in public databases or in over 1000 ethnicity-matched control individuals. I109 is a highly conserved residue in the forkhead box (Fox) domain of FOXF2, a member of the Fox protein family of transcription factors that regulate the expression of genes involved in embryogenic development as well as adult life. Our in vitro studies show that the half-life of mutant FOXF2 is reduced compared to that of wild type. Foxf2 is expressed in the cochlea of developing and adult mice. The mouse knockout of Foxf2 shows shortened and malformed cochleae, in addition to altered shape of hair cells with innervation and planar cell polarity defects. Expressions of Eya1 and Pax3, genes essential for cochlear development, are reduced in the cochleae of Foxf2 knockout mice. We conclude that FOXF2 plays a major role in cochlear development and its dysfunction leads to SNHL and developmental anomalies of the cochlea in humans and mice.


Asunto(s)
Cóclea/embriología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/fisiología , Adulto , Animales , Niño , Cóclea/metabolismo , Cóclea/fisiología , Desarrollo Embrionario , Femenino , Células Ciliadas Auditivas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Organogénesis , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX3/fisiología , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal/genética , Secuenciación Completa del Genoma
8.
Hum Mol Genet ; 28(8): 1298-1311, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30561643

RESUMEN

Pigmentary glaucoma (PG) is a common glaucoma subtype that results from release of pigment from the iris, called pigment dispersion syndrome (PDS), and its deposition throughout the anterior chamber of the eye. Although PG has a substantial heritable component, no causative genes have yet been identified. We used whole exome sequencing of two independent pedigrees to identify two premelanosome protein (PMEL) variants associated with heritable PDS/PG. PMEL encodes a key component of the melanosome, the organelle essential for melanin synthesis, storage and transport. Targeted screening of PMEL in three independent cohorts (n = 394) identified seven additional PDS/PG-associated non-synonymous variants. Five of the nine variants exhibited defective processing of the PMEL protein. In addition, analysis of PDS/PG-associated PMEL variants expressed in HeLa cells revealed structural changes to pseudomelanosomes indicating altered amyloid fibril formation in five of the nine variants. Introduction of 11-base pair deletions to the homologous pmela in zebrafish by the clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 method caused profound pigmentation defects and enlarged anterior segments in the eye, further supporting PMEL's role in ocular pigmentation and function. Taken together, these data support a model in which missense PMEL variants represent dominant negative mutations that impair the ability of PMEL to form functional amyloid fibrils. While PMEL mutations have previously been shown to cause pigmentation and ocular defects in animals, this research is the first report of mutations in PMEL causing human disease.


Asunto(s)
Glaucoma de Ángulo Abierto/genética , Antígeno gp100 del Melanoma/genética , Antígeno gp100 del Melanoma/fisiología , Adulto , Amiloide/metabolismo , Animales , Femenino , Células HeLa , Humanos , Iris/metabolismo , Masculino , Melanosomas/genética , Persona de Mediana Edad , Mutación Missense/genética , Linaje , Pigmentación/genética , Secuenciación del Exoma/métodos , Adulto Joven , Pez Cebra
9.
J Ophthalmol ; 2018: 5926906, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29780638

RESUMEN

We explore the ideas and advances surrounding the genetic basis of pigment dispersion syndrome (PDS) and pigmentary glaucoma (PG). As PG is the leading cause of nontraumatic blindness in young adults and current tailored interventions have proven ineffective, a better understanding of the underlying causes of PDS, PG, and their relationship is essential. Despite PDS being a subclinical disease, a large proportion of patients progress to PG with associated vision loss. Decades of research have supported a genetic component both for PDS and conversion to PG. We review the body of evidence supporting a genetic basis in humans and animal models and reevaluate classical mechanisms of PDS/PG considering this new evidence.

10.
Invest Ophthalmol Vis Sci ; 59(6): 2548-2554, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29847662

RESUMEN

Purpose: This study examines the effect of FOXC1 on the prostaglandin pathway in order to explore FOXC1's role in the prostaglandin-resistant glaucoma phenotype commonly seen in Axenfeld-Rieger syndrome. Methods: Binding and transcriptional activity of FOXC1 to the gene coding for the EP3 prostaglandin receptor (PTGER3) were evaluated through ChIP-qPCR and luciferase-based assays. Immortalized trabecular meshwork cells (TM1) and HeLa cells had FOXC1 mRNA reduced via siRNA interference. qPCR and Western blot experiments were conducted to examine the changes in prostaglandin receptor expression brought about by lowered FOXC1. TM1 cells were then treated with 10 µM latanoprost acid and/or an siRNA for FOXC1. The expression of fibronectin and matrix metalloproteinase 9 were evaluated via qPCR in each treatment condition. Results: ChIP-qPCR and luciferase experiments confirmed that FOXC1 binds to and activates transcription of the EP3 gene prostaglandin receptor. qPCR and Western experiments in HeLa and TM1 cells showed that FOXC1 siRNA knockdown results in significantly lowered EP3 levels (protein and RNA). In addition, RNA levels of the other prostaglandin receptor genes EP1 (PTGER1), EP2 (PTGER2), EP4 (PTGER4), and FP (PTGFR) were altered when FOXC1 was knocked down in TM1 and HeLa cells. Analysis of fibronectin expression in TM1 cells after treatment with 10 µM latanoprost acid showed a statistically significant increase in expression; this increase was abrogated by cotreatment with a siRNA for FOXC1. Conclusions: We show the abrogation of latanoprost signalling when FOXC1 is knocked down via siRNA in a trabecular meshwork cell line. We propose that the lower levels of active FOXC1 in Axenfeld-Rieger syndrome patients with glaucoma account for the lack of response to prostaglandin-based medications.


Asunto(s)
Antihipertensivos/farmacología , Factores de Transcripción Forkhead/fisiología , Regulación de la Expresión Génica/fisiología , Latanoprost/farmacología , Subtipo EP3 de Receptores de Prostaglandina E/genética , Malla Trabecular/efectos de los fármacos , Segmento Anterior del Ojo/anomalías , Western Blotting , Anomalías del Ojo/tratamiento farmacológico , Enfermedades Hereditarias del Ojo/tratamiento farmacológico , Silenciador del Gen/fisiología , Glaucoma/tratamiento farmacológico , Células HeLa/efectos de los fármacos , Células HeLa/metabolismo , Humanos , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Malla Trabecular/metabolismo
11.
PLoS One ; 13(4): e0195971, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29664915

RESUMEN

Mutations in PITX2 have been implicated in several genetic disorders, particularly Axenfeld-Rieger syndrome. In order to determine the most reliable bioinformatics tools to assess the likely pathogenicity of PITX2 variants, the results of bioinformatics predictions were compared to the impact of variants on PITX2 structure and function. The MutPred, Provean, and PMUT bioinformatic tools were found to have the highest performance in predicting the pathogenicity effects of all 18 characterized missense variants in PITX2, all with sensitivity and specificity >93%. Applying these three programs to assess the likely pathogenicity of 13 previously uncharacterized PITX2 missense variants predicted 12/13 variants as deleterious, except A30V which was predicted as benign variant for all programs. Molecular modeling of the PITX2 homoedomain predicts that of the 31 known PITX2 variants, L54Q, F58L, V83F, V83L, W86C, W86S, and R91P alter PITX2's structure. In contrast, the remaining 24 variants are not predicted to change PITX2's structure. The results of molecular modeling, performed on all the PITX2 missense mutations located in the homeodomain, were compared with the findings of eight protein stability programs. CUPSAT was found to be the most reliable in predicting the effect of missense mutations on PITX2 stability. Our results showed that for PITX2, and likely other members of this homeodomain transcription factor family, MutPred, Provean, PMUT, molecular modeling, and CUPSAT can reliably be used to predict PITX2 missense variants pathogenicity.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Simulación por Computador , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Mutación , Factores de Transcripción/química , Factores de Transcripción/genética , Alelos , Secuencia de Aminoácidos , Bases de Datos Genéticas , Proteínas de Homeodominio/metabolismo , Humanos , Modelos Moleculares , Fenotipo , Conformación Proteica , Estabilidad Proteica , Relación Estructura-Actividad , Factores de Transcripción/metabolismo , Proteína del Homeodomínio PITX2
12.
Oncotarget ; 9(8): 8165-8178, 2018 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-29487724

RESUMEN

In recent years, rapidly accumulating evidence implicates forkhead box C1 (FOXC1) in cancer, especially in studies of basal-like breast cancer (BLBC). Other studies have followed suit, demonstrating that FOXC1 is not only a major player in this breast cancer subtype, but also in hepatocellular carcinoma (HCC), endometrial cancer, Hodgkin's lymphoma (HL), and non-Hodgkin's lymphoma (NHL). The FOXC1 gene encodes a transcription factor that is crucial to mesodermal, neural crest, and ocular development, and mutations found in FOXC1 have been found to cause dominantly inherited Axenfeld-Rieger Syndrome (ARS). Interestingly, while FOXC1 missense mutations that are associated with ARS usually reduce gene activity, increased FOXC1 function now appears to be often linked to more aggressive cancer phenotypes in BLBC, HCC, HL, and NHL. This review discusses not only the role of FOXC1 in cancer cell progression, proliferation, differentiation, and metastasis, but also the underlying mechanisms of how FOXC1 can contribute to aggressive cancer phenotypes.

13.
Hum Mol Genet ; 26(18): 3630-3638, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28911203

RESUMEN

Mutations in FOXC1 and PITX2 constitute the most common causes of ocular anterior segment dysgenesis (ASD), and confer a high risk for secondary glaucoma. The genetic causes underlying ASD in approximately half of patients remain unknown, despite many of them being screened by whole exome sequencing. Here, we performed whole genome sequencing on DNA from two affected individuals from a family with dominantly inherited ASD and glaucoma to identify a 748-kb deletion in a gene desert that contains conserved putative PITX2 regulatory elements. We used CRISPR/Cas9 to delete the orthologous region in zebrafish in order to test the pathogenicity of this structural variant. Deletion in zebrafish reduced pitx2 expression during development and resulted in shallow anterior chambers. We screened additional patients for copy number variation of the putative regulatory elements and found an overlapping deletion in a second family and in a potentially-ancestrally-related index patient with ASD and glaucoma. These data suggest that mutations affecting conserved non-coding elements of PITX2 may constitute an important class of mutations in patients with ASD for whom the molecular cause of their disease have not yet been identified. Improved functional annotation of the human genome and transition to sequencing of patient genomes instead of exomes will be required before the magnitude of this class of mutations is fully understood.


Asunto(s)
Anomalías del Ojo/genética , Glaucoma/genética , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Animales , Segmento Anterior del Ojo/metabolismo , Secuencia Conservada , Variaciones en el Número de Copia de ADN , Modelos Animales de Enfermedad , Anomalías del Ojo/metabolismo , Eliminación de Gen , Glaucoma/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Intrones , Músculos , Mutación , Linaje , Eliminación de Secuencia , Factores de Transcripción/metabolismo , Pez Cebra/genética , Proteína del Homeodomínio PITX2
14.
PLoS One ; 12(6): e0178518, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28575017

RESUMEN

The neurodegenerative disease glaucoma is one of the leading causes of blindness in the world. Glaucoma is characterized by progressive visual field loss caused by retinal ganglion cell (RGC) death. Both surgical glaucoma treatments and medications are available, however, they only halt glaucoma progression and are unable to reverse damage. Furthermore, many patients do not respond well to treatments. It is therefore important to better understand the mechanisms involved in glaucoma pathogenesis. Patients with Axenfeld-Rieger syndrome (ARS) offer important insight into glaucoma progression. ARS patients are at 50% risk of developing early onset glaucoma and respond poorly to treatments, even when surgical treatments are combined with medications. Mutations in the transcription factor FOXC1 cause ARS. Alterations in FOXC1 levels cause ocular malformations and disrupt stress response in ocular tissues, thereby contributing to glaucoma progression. In this study, using biochemical and molecular techniques, we show that FOXC1 regulates the expression of RAB3GAP1, RAB3GAP2 and SNAP25, three genes with central roles in both exocytosis and endocytosis, responsible for extracellular trafficking. FOXC1 positively regulates RAB3GAP1 and RAB3GAP2, while either increase or decrease in FOXC1 levels beyond its normal range results in decreased SNAP25. In addition, we found that FOXC1 regulation of RAB3GAP1, RAB3GAP2 and SNAP25 affects secretion of Myocilin (MYOC), a protein associated with juvenile onset glaucoma and steroid-induced glaucoma. The present work reveals that FOXC1 is an important regulator of exocytosis and establishes a new link between FOXC1 and MYOC-associated glaucoma.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Exocitosis , Proteínas del Ojo/metabolismo , Factores de Transcripción Forkhead/fisiología , Glicoproteínas/metabolismo , Proteína 25 Asociada a Sinaptosomas/fisiología , Proteínas de Unión al GTP rab3/fisiología , Factores de Transcripción Forkhead/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Luciferasas/genética , ARN Mensajero/genética , Proteína 25 Asociada a Sinaptosomas/genética , Activación Transcripcional , Proteínas de Unión al GTP rab3/genética
15.
Hum Mutat ; 38(2): 169-179, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27804176

RESUMEN

Mutations in the forkhead box C1 gene (FOXC1) cause Axenfeld-Rieger syndrome (ARS). Here, we investigated the effect of four ARS missense variants on FOXC1 structure and function, and examined the predictive value of four in silico programs for all 31 FOXC1 missense variants identified to date. Molecular modeling of the FOXC1 forkhead domain predicts that c.402G> A (p.C135Y) alters FOXC1's structure. In contrast, c.378A> G (p.H128R) and c.481A> G (p.M161V) are not predicted to change FOXC1's structure. Functional analysis indicates that p.H128R reduced DNA binding, transactivation, nuclear localization, and has a longer protein half-life than normal. p.C135Y significantly disrupts FOXC1's DNA binding, transactivation, and nuclear localization. p.M161V reduces transactivation capacity without affecting other FOXC1 functions. C.1103C> A (p.T368N) is indistinguishable from wild-type FOXC1 in all tests, consistent with being a rare benign variant. Comparison of these four variants, plus 18 previously characterized FOXC1 missense variants, with predictions from four commonly used in silico bioinformatics programs indicated that sorting intolerant from tolerant (SIFT), polymorphism phenotyping (PolyPhen-2), and MutPred can sensitively identify as pathogenic only FOXC1 mutations with significant functional defects. This information was used to predict, as disease-causing, nine additional FOXC1 missense variations. Importantly, our results indicate SIFT, PolyPhen-2, and MutPred can reliably be used to predict missense variant pathogenicity for forkhead transcription factors.


Asunto(s)
Segmento Anterior del Ojo/anomalías , Biología Computacional , Anomalías del Ojo/genética , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Modelos Moleculares , Mutación , Alelos , Secuencia de Aminoácidos , Biología Computacional/métodos , Anomalías del Ojo/diagnóstico , Enfermedades Hereditarias del Ojo , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Genotipo , Células HeLa , Humanos , Mutación Missense , Conformación Proteica , Programas Informáticos , Relación Estructura-Actividad , Transactivadores/metabolismo
16.
Ophthalmic Genet ; 38(2): 108-116, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27070211

RESUMEN

Prostaglandins are small pro-inflammatory molecules derived from arachidonic acid that play roles in a multitude of biological processes including, but not limited to, inflammation, pain modulation, allergies, and bone formation. Prostaglandin analogues are the front-line medications for the treatment of glaucoma, a condition resulting in blindness due to the death of retinal ganglion cells. These drugs act by lowering intraocular pressure (IOP), a major risk factor for glaucoma. The currently used prostaglandin analogues (latanoprost, bimatoprost, tafluprost, and travoprost) mimic PGF2 and target one of the prostaglandin receptors (FP), though research into harnessing the other receptors using compounds like Sulprostone (EP3 receptor), or Iloprost (IP receptor) are currently ongoing. In this review, we summarize the research into each of the prostaglandin molecules (PGD2, PGE2, PGF2, PGI2, TXA2) and their respective receptors (DP, EP1, 2, 3, 4, FP, IP). We examine the modes of action of each of these receptors, their expression, their role in aqueous humour production and outflow within the eye, as well as their roles as medications for the treatment of glaucoma.


Asunto(s)
Ojo/metabolismo , Glaucoma/metabolismo , Prostaglandinas Sintéticas/uso terapéutico , Prostaglandinas/fisiología , Glaucoma/tratamiento farmacológico , Humanos , Presión Intraocular/efectos de los fármacos , Receptores de Prostaglandina/metabolismo
17.
Oncotarget ; 7(34): 54228-54239, 2016 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-27276711

RESUMEN

Dominant mutations in the FOXC2 gene cause a form of lymphedema primarily of the limbs that usually develops at or after puberty. In 90-95% of patients, lymphedema is accompanied by distichiasis. FOXC2 is a member of the forkhead/winged-helix family of transcription factors and plays essential roles in different developmental pathways and physiological processes. We previously described six unrelated families with primary lymphedema-distichiasis in which patients showed different FOXC2 mutations located outside of the forkhead domain. Of those, four were missense mutations, one a frameshift mutation, and the last a stop mutation. To assess their pathogenic potential, we have now examined the subcellular localization and the transactivation activity of the mutated FOXC2 proteins. All six FOXC2 mutant proteins were able to localize into the nucleus; however, the frameshift truncated protein appeared to be sequestered into nuclear aggregates. A reduction in the ability to activate FOXC1/FOXC2 response elements was detected in 50% of mutations, while the remaining ones caused an increase of protein transactivation activity. Our data reveal that either a complete loss or a significant gain of FOXC2 function can cause a perturbation of lymphatic vessel formation leading to lymphedema.


Asunto(s)
Pestañas/anomalías , Factores de Transcripción Forkhead/genética , Linfedema/genética , Mutación , Adulto , Anciano , Femenino , Factores de Transcripción Forkhead/fisiología , Humanos , Linfedema/etiología , Masculino , Persona de Mediana Edad
18.
Acta Ophthalmol ; 94(7): e571-e579, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27009473

RESUMEN

PURPOSE: Mutations in the bicoid-like transcription factor PITX2 gene often result in Axenfeld-Rieger syndrome (ARS), an autosomal-dominant inherited disorder. We report here the discovery and characterization of novel PITX2 deletions in a small kindred with ARS. METHODS: Two familial patients (father and son) from a consanguineous family were examined in the present study. Patient DNA samples were screened for PITX2 mutations by DNA sequencing and for copy number variation by SYBR Green quantitative polymerase chain reaction (PCR) analysis. RESULTS: We report a novel deletion involving the coding region of PITX2 in both patients. The minimum size of the deletion is 1 421 914 bp that spans one upstream regulatory element (CE4), PITX2 and a minimum of 13 neighbouring genes. The maximum size of the deletion is 3 789 983 bp. The proband (son) additionally possesses a novel 2-bp deletion in a non-coding exon of the remaining PITX2 allele predicted to alter correct splicing. CONCLUSION: Our findings implicate a novel deletion of the PITX2 gene in the pathogenesis of ARS in the affected family. This ARS family presented with an atypical and extremely severe phenotype that resulted in four miscarriages and the death at 10 months of age of a sib of the proband. As the phenotypic manifestations in the proband are more severe than that of the father, we hypothesize that the deletion of the entire PITX2 allele plus a novel 2-bp deletion (observed in the proband) within the remaining PITX2 allele together contributed to the atypical ARS presentation in this family. This is the first study reporting on bi-allelic changes of PITX2 potentially contributing to a more severe ARS phenotype.


Asunto(s)
Segmento Anterior del Ojo/anomalías , Anomalías del Ojo/genética , Proteínas de Homeodominio/genética , Mutación , Factores de Transcripción/genética , Adulto , Preescolar , Consanguinidad , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Exones/genética , Enfermedades Hereditarias del Ojo , Humanos , Masculino , Sistemas de Lectura Abierta/genética , Linaje , Reacción en Cadena en Tiempo Real de la Polimerasa , Eliminación de Secuencia , Proteína del Homeodomínio PITX2
19.
Surv Ophthalmol ; 60(4): 310-26, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25907525

RESUMEN

Glaucoma, a progressive degenerative condition that results in the death of retinal ganglion cells, is one of the leading causes of blindness, affecting millions worldwide. The mechanisms underlying glaucoma are not well understood, although years of studies have shown that the largest risk factors are elevated intraocular pressure, age, and genetics. Eleven genes and multiple loci have been identified as contributing factors. These genes act by a number of mechanisms, including mechanical stress, ischemic/oxidative stress, and neurodegeneration. We summarize the recent advances in the understanding of glaucoma and propose a unified hypothesis for glaucoma pathogenesis. Glaucoma does not result from a single pathological mechanism, but rather a combination of pathways that are influenced by genes, age, and environment. In particular, we hypothesize that, in the presence of genetic risk factors, exposure to environment stresses results in an earlier age of onset for glaucoma. This hypothesis is based upon the overlap of the molecular pathways in which glaucoma genes are involved. Because of the interactions between these processes, it is likely that there are common therapies that may be effective for different subtypes of glaucoma.


Asunto(s)
Edad de Inicio , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad/etiología , Glaucoma/etiología , Células Ganglionares de la Retina/patología , Humanos , Estrés Oxidativo , Factores de Riesgo
20.
Lab Invest ; 94(7): 726-39, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24840332

RESUMEN

The forkhead box (Fox) superfamily of transcription factors has essential roles in organogenesis and tissue differentiation. Foxa1 and Foxa2 are expressed during prostate budding and ductal morphogenesis, whereas Foxa1 expression is retained in adult prostate epithelium. Previous characterization of prostatic tissue rescued from embryonic Foxa1 knockout mice revealed Foxa1 to be essential for ductal morphogenesis and epithelial maturation. However, it is unknown whether Foxa1 is required to maintain the differentiated status in adult prostate epithelium. Here, we employed the PBCre4 transgenic system and determined the impact of prostate-specific Foxa1 deletion in adult murine epithelium. PBCre4/Foxa1(loxp/loxp) mouse prostates showed progressive florid hyperplasia with extensive cribriform patterning, with the anterior prostate being most affected. Immunohistochemistry studies show mosaic Foxa1 KO consistent with PBCre4 activity, with Foxa1 KO epithelial cells specifically exhibiting altered cell morphology, increased proliferation, and elevated expression of basal cell markers. Castration studies showed that, while PBCre4/Foxa1(loxp/loxp) prostates did not exhibit altered sensitivity in response to hormone ablation compared with control prostates, the number of Foxa1-positive cells in mosaic Foxa1 KO prostates was significantly reduced compared with Foxa1-negative cells following castration. Unexpectedly, gene expression profile analyses revealed that Foxa1 deletion caused abnormal expression of seminal vesicle-associated genes in KO prostates. In summary, these results indicate Foxa1 expression is required for the maintenance of prostatic cellular differentiation.


Asunto(s)
Diferenciación Celular/genética , Epitelio/metabolismo , Factor Nuclear 3-alfa del Hepatocito/genética , Hiperplasia Prostática/genética , Animales , Epitelio/patología , Factor Nuclear 3-alfa del Hepatocito/deficiencia , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Inmunohistoquímica , Integrasas/genética , Integrasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Próstata/metabolismo , Próstata/patología , Hiperplasia Prostática/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vesículas Seminales/metabolismo , Transcriptoma/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...