Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 263
Filtrar
1.
Clin Genet ; 105(6): 661-665, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38361102

RESUMO

Familial exudative vitreoretinopathy (FEVR) is linked to disruption of the Norrin/Frizzled-4 signaling pathway, which plays an important role in retinal angiogenesis. Severe or complete knock-down of proteins in the pathway also causes syndromic forms of the condition. Both heterozygous and biallelic pathogenic variants in the FZD4 gene, encoding the pathway's key protein frizzled-4, are known to cause FEVR. However, it is not clear what effect different FZD4 variants have, and whether extraocular features should be expected in those with biallelic pathogenic FZD4 variants. Biallelic FZD4 variants were found in a young boy with isolated, severe FEVR. His parents were heterozygous for one variant each and reported normal vision. In-vitro studies of the two variants, demonstrated that it was the combination of the two which led to severe inhibition of the Norrin/Frizzled-4 pathway. Our observations demonstrate that biallelic FZD4-variants are associated with a severe form of FEVR, which does not necessarily include extraocular features. In addition, variants causing severe FEVR in combination, may have no or minimal effect in heterozygous parents as non-penetrance is also a major feature in dominant FZD4-FEVR disease. This underscores the importance of genetic testing of individuals and families with FEVR.


Assuntos
Alelos , Vitreorretinopatias Exsudativas Familiares , Receptores Frizzled , Humanos , Masculino , Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/patologia , Vitreorretinopatias Exsudativas Familiares/genética , Receptores Frizzled/genética , Predisposição Genética para Doença , Heterozigoto , Mutação/genética , Linhagem , Fenótipo , Doenças Retinianas/genética , Doenças Retinianas/patologia , Lactente , Pré-Escolar
2.
Am J Med Genet A ; 194(5): e63542, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38234180

RESUMO

Axenfeld-Rieger Syndrome (ARS) type 1 is a rare autosomal dominant condition characterized by anterior chamber anomalies, umbilical defects, dental hypoplasia, and craniofacial anomalies, with Meckel's diverticulum in some individuals. Here, we describe a clinically ascertained female of childbearing age with ARS for whom clinical targeted sequencing and deletion/duplication analysis followed by clinical exome and genome sequencing resulted in no pathogenic variants or variants of unknown significance in PITX2 or FOXC1. Advanced bioinformatic analysis of the genome data identified a complex, balanced rearrangement disrupting PITX2. This case is the first reported intrachromosomal rearrangement leading to ARS, illustrating that for patients with compelling clinical phenotypes but negative genomic testing, additional bioinformatic analysis are essential to identify subtle genomic abnormalities in target genes.


Assuntos
Segmento Anterior do Olho , Anormalidades do Olho , Oftalmopatias Hereditárias , Proteína Homeobox PITX2 , Feminino , Humanos , Segmento Anterior do Olho/anormalidades , Anormalidades do Olho/diagnóstico , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Oftalmopatias Hereditárias/diagnóstico , Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/patologia , Fatores de Transcrição Forkhead/genética , Proteínas de Homeodomínio/genética
6.
Neuroradiology ; 64(9): 1773-1780, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35420309

RESUMO

The clinical and neuroimaging findings of a family with a variant ACTA2 gene (c351C > G), presenting with smooth muscle dysfunction in structures of neural crest derivation, are discussed. The combination of aortic abnormalities, patent ductus arteriosus, congenital mydriasis and distinctive cerebrovascular and brain morphological abnormalities characterise this disorder. Two sisters, heterozygous for the variant, and their mother, a mosaic, are presented. Brain parenchymal changes are detailed for the first time in a non-Arg179His variant. Radiological features of the petrous canal and external carotid are highlighted. We explore the potential underlying biological and embryological mechanisms.


Assuntos
Permeabilidade do Canal Arterial , Oftalmopatias Hereditárias , Midríase , Actinas , Permeabilidade do Canal Arterial/genética , Permeabilidade do Canal Arterial/patologia , Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/patologia , Feminino , Humanos , Músculo Liso/patologia , Midríase/genética , Midríase/patologia , Neuroimagem
8.
Genes (Basel) ; 12(9)2021 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-34573386

RESUMO

Monogenic syndromic disorders frequently feature ocular manifestations, one of which is glaucoma. In many cases, glaucoma in children may go undetected, especially in those that have other severe systemic conditions that affect other parts of the eye and the body. Similarly, glaucoma may be the first presenting sign of a systemic syndrome. Awareness of syndromes associated with glaucoma is thus critical both for medical geneticists and ophthalmologists. In this review, we highlight six categories of disorders that feature glaucoma and other ocular or systemic manifestations: anterior segment dysgenesis syndromes, aniridia, metabolic disorders, collagen/vascular disorders, immunogenetic disorders, and nanophthalmos. The genetics, ocular and systemic features, and current and future treatment strategies are discussed. Findings from rare diseases also uncover important genes and pathways that may be involved in more common forms of glaucoma, and potential novel therapeutic strategies to target these pathways.


Assuntos
Oftalmopatias Hereditárias/genética , Glaucoma/etiologia , Glaucoma/genética , Aniridia/genética , Aniridia/patologia , Doenças do Colágeno/complicações , Doenças do Colágeno/genética , Doenças do Colágeno/patologia , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Oftalmopatias Hereditárias/patologia , Glaucoma/patologia , Glaucoma de Ângulo Fechado/complicações , Glaucoma de Ângulo Fechado/genética , Glaucoma de Ângulo Fechado/patologia , Humanos , Hiperopia/complicações , Hiperopia/genética , Hiperopia/patologia , Doenças do Sistema Imunitário/complicações , Doenças do Sistema Imunitário/genética , Doenças do Sistema Imunitário/patologia , Doenças Metabólicas/complicações , Doenças Metabólicas/genética , Doenças Metabólicas/patologia , Microftalmia/complicações , Microftalmia/genética , Microftalmia/patologia , Síndrome , Doenças Vasculares/complicações , Doenças Vasculares/genética , Doenças Vasculares/patologia
9.
Int J Mol Sci ; 22(18)2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34576164

RESUMO

Axenfeld-Rieger syndrome (ARS) encompasses a group of developmental disorders that affect the anterior segment of the eye, as well as systemic developmental defects in some patients. Malformation of the ocular anterior segment often leads to secondary glaucoma, while some patients also present with cardiovascular malformations, craniofacial and dental abnormalities and additional periumbilical skin. Genes that encode two transcription factors, FOXC1 and PITX2, account for almost half of known cases, while the genetic lesions in the remaining cases remain unresolved. Given the genetic similarity between zebrafish and humans, as well as robust antisense inhibition and gene editing technologies available for use in these animals, loss of function zebrafish models for ARS have been created and shed light on the mechanism(s) whereby mutations in these two transcription factors cause such a wide array of developmental phenotypes. This review summarizes the published phenotypes in zebrafish foxc1 and pitx2 loss of function models and discusses possible mechanisms that may be used to target pharmaceutical development and therapeutic interventions.


Assuntos
Segmento Anterior do Olho/anormalidades , Anormalidades do Olho/genética , Oftalmopatias Hereditárias/genética , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Segmento Anterior do Olho/patologia , Anormalidades do Olho/patologia , Oftalmopatias Hereditárias/patologia , Fatores de Transcrição Forkhead/genética , Humanos , Fatores de Transcrição/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
10.
Sci Rep ; 11(1): 19032, 2021 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-34561487

RESUMO

The photoreceptor-specific nuclear receptor Nr2e3 is not expressed in Nr2e3rd7/rd7 mice, a mouse model of the recessively inherited retinal degeneration enhanced S-cone sensitivity syndrome (ESCS). We characterized in detail C57BL/6J Nr2e3rd7/rd7 mice in vivo by fundus photography, optical coherence tomography and fluorescein angiography and, post mortem, by histology and immunohistochemistry. White retinal spots and so-called 'rosettes' first appear at postnatal day (P) 12 in the dorsal retina and reach maximal expansion at P21. The highest density in 'rosettes' is observed within a region located between 100 and 350 µM from the optic nerve head. 'Rosettes' disappear between 9 to 12 months. Non-apoptotic cell death markers are detected during the slow photoreceptor degeneration, at a rate of an approximately 3% reduction of outer nuclear layer thickness per month, as observed from 7 to 31 months of age. In vivo analysis of Nr2e3rd7/rd7 Cx3cr1gfp/+ retinas identified microglial cells within 'rosettes' from P21 on. Subretinal macrophages were observed in vivo and by confocal microscopy earliest in 12-months-old Nr2e3rd7/rd7 retinas. At P21, S-opsin expression and the number of S-opsin expressing dorsal cones was increased. The dorso-ventral M-cone gradient was present in Nr2e3rd7/rd7 retinas, but M-opsin expression and M-opsin expressing cones were decreased. Retinal vasculature was normal.


Assuntos
Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/patologia , Receptores Nucleares Órfãos/genética , Células Fotorreceptoras Retinianas Cones/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Transtornos da Visão/genética , Transtornos da Visão/patologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Opsinas/genética , Opsinas/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia
11.
JAMA Ophthalmol ; 139(10): 1089-1095, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34410316

RESUMO

IMPORTANCE: Keratoglobus is a rare corneal disorder characterized by generalized thinning and globular protrusion of the cornea. Affected individuals typically have significantly decreased vision and are at risk of corneal perforation. The genetic basis and inheritance pattern of isolated congenital keratoglobus are currently unknown. OBJECTIVE: To identify the genetic basis of isolated congenital keratoglobus. DESIGN, SETTING, AND PARTICIPANTS: This case series and molecular analysis studied 3 unrelated nonconsanguineous families with keratoglobus at a medical center in Israel. Data were collected from June 2019 to March 2021 and analyzed during the same period. EXPOSURES: Whole-exome sequencing and direct Sanger sequencing, expression analysis by real-time polymerase chain reaction, splice-site variant analysis, immunohistochemical staining, and histological evaluation of a knockout mouse model. MAIN OUTCOMES AND MEASURE: Molecular characteristics associated with keratoglobus. RESULTS: Four pediatric patients (3 male individuals) from 3 families had clinical findings consistent with keratoglobus. These included globular protrusion, corneal thinning more prominent at the periphery, and high astigmatism. Truncating and splice site variants were identified in the TMEM45A gene, which fully segregate with the disorder. All affected individuals were homozygous or compound heterozygous for variants in the TMEM45A gene, while unaffected family members were heterozygous carriers. Expression analysis in healthy controls showed that TMEM45A was expressed 23 times higher in the human cornea compared with peripheral blood. Immunohistochemical staining of the TMEM45A protein in normal corneas confirmed its expression in the corneal stroma and epithelium. A TMEM45A knockout mouse model showed structural features consistent with keratoglobus. CONCLUSIONS AND RELEVANCE: Expression of TMEM45A has been previously shown to result in upregulation of extracellular matrix components and fibrosis. These results suggest that isolated congenital keratoglobus is an autosomal recessively inherited disorder associated with variants in the TMEM45A gene.


Assuntos
Oftalmopatias Hereditárias , Ceratocone , Animais , Criança , Córnea/patologia , Anormalidades do Olho , Oftalmopatias Hereditárias/patologia , Feminino , Doenças Genéticas Ligadas ao Cromossomo X , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos
12.
Sci Rep ; 11(1): 16528, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34400702

RESUMO

Epithelial cysts run a high risk of recurrence and conversion to sheet-like ingrowth after surgical intervention. In this retrospective study, we introduced a modified iridectomy for treatment of secondary epithelial iris cysts (EICs) in the anterior chamber. Twenty-nine patients (29 eyes) aged 2-61 years received "open iridectomy" for EICs between April 1995 and July 2019. After viscodissection, most of the cyst wall was cut using a 20-gauge aspiration cutter via a 2.5-mm clear corneal incision. The residue closely adhering to the iris stroma was remained to avoid photophobia and diplopia. At 3 months, best corrected visual acuity was ≥ 20/100 in 55.5% (15/27, except two pediatric patients with poor cooperation) of patients. Among the eight patients suffering partial corneal edema preoperatively, six patients received surgery treatment at 3-6.5 months, and the cornea in the other two patients became transparent after medication. In a mean follow-up of 47.4 months, recurrence occurred in 3 patients at 7, 37, and 118 months, respectively. The percentage of treatment success was 96%, 87%, and 65% at 1, 5, and 10 years, respectively. "Open iridectomy" was effective for EICs, with a minimal invasion, less damage to the corneal endothelium, and a low recurrence rate.


Assuntos
Oftalmopatias Hereditárias/cirurgia , Iridectomia/métodos , Iris/anormalidades , Epitélio Pigmentado Ocular/anormalidades , Adolescente , Adulto , Assistência ao Convalescente , Câmara Anterior/cirurgia , Criança , Pré-Escolar , Oftalmopatias Hereditárias/etiologia , Oftalmopatias Hereditárias/patologia , Ferimentos Oculares Penetrantes/complicações , Feminino , Seguimentos , Humanos , Iris/patologia , Iris/cirurgia , Masculino , Pessoa de Meia-Idade , Epitélio Pigmentado Ocular/patologia , Epitélio Pigmentado Ocular/cirurgia , Recidiva , Estudos Retrospectivos , Resultado do Tratamento , Adulto Jovem
13.
Sci Rep ; 11(1): 15393, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321574

RESUMO

Many studies have demonstrated that rhegmatogenous retinal detachment (RRD) leads to impaired retinal circulation. However, the involvement of inflammation in the RRD-induced worsening of retinal circulation was obscure. This retrospective observational study included 150 patients with primary RRD (macula-on, n = 63; macula-off, n = 87) who underwent 25-gauge microincision vitrectomy surgery (25G MIVS). Total retinal blood flow was represented by the mean blur rate (MBR) of the optic nerve head vessel, measured by laser speckle flowgraphy preoperatively and until 6 months postoperatively. Aqueous humor samples were obtained during surgery to determine cytokine concentrations by enzyme-linked immunosorbent assay. At 3 and 6 months postoperatively, there were no significant differences between eyes with macula-on RRD and fellow eyes. However, in macula-off RRD, MBR remained significantly lower in RRD eyes 6 months postoperatively (P < 0.05). Log-transformed levels of soluble intercellular adhesion molecule-1 (sICAM-1) were negatively correlated with relative MBR (r-MBR, RRD eye/fellow eye) before surgery (r = - 0.47, P = 0.01) in macula-on, but not macula-off, RRD. Six months postoperatively, r-MBR correlated significantly with sICAM-1 levels (r = - 0.36, P = 0.02) in macula-off RRD. ICAM-1 may play a role in RRD-induced deterioration of retinal circulation.


Assuntos
Oftalmopatias Hereditárias/genética , Molécula 1 de Adesão Intercelular/genética , Macula Lutea/metabolismo , Retina/metabolismo , Descolamento Retiniano/genética , Oftalmopatias Hereditárias/sangue , Oftalmopatias Hereditárias/patologia , Oftalmopatias Hereditárias/cirurgia , Feminino , Humanos , Molécula 1 de Adesão Intercelular/sangue , Macula Lutea/irrigação sanguínea , Macula Lutea/patologia , Macula Lutea/cirurgia , Masculino , Pessoa de Meia-Idade , Disco Óptico/metabolismo , Disco Óptico/patologia , Retina/patologia , Retina/cirurgia , Descolamento Retiniano/sangue , Descolamento Retiniano/patologia , Descolamento Retiniano/cirurgia , Tomografia de Coerência Óptica , Acuidade Visual/genética , Acuidade Visual/fisiologia , Vitrectomia
14.
Am J Hum Genet ; 108(9): 1669-1691, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34314705

RESUMO

Transportin-2 (TNPO2) mediates multiple pathways including non-classical nucleocytoplasmic shuttling of >60 cargoes, such as developmental and neuronal proteins. We identified 15 individuals carrying de novo coding variants in TNPO2 who presented with global developmental delay (GDD), dysmorphic features, ophthalmologic abnormalities, and neurological features. To assess the nature of these variants, functional studies were performed in Drosophila. We found that fly dTnpo (orthologous to TNPO2) is expressed in a subset of neurons. dTnpo is critical for neuronal maintenance and function as downregulating dTnpo in mature neurons using RNAi disrupts neuronal activity and survival. Altering the activity and expression of dTnpo using mutant alleles or RNAi causes developmental defects, including eye and wing deformities and lethality. These effects are dosage dependent as more severe phenotypes are associated with stronger dTnpo loss. Interestingly, similar phenotypes are observed with dTnpo upregulation and ectopic expression of TNPO2, showing that loss and gain of Transportin activity causes developmental defects. Further, proband-associated variants can cause more or less severe developmental abnormalities compared to wild-type TNPO2 when ectopically expressed. The impact of the variants tested seems to correlate with their position within the protein. Specifically, those that fall within the RAN binding domain cause more severe toxicity and those in the acidic loop are less toxic. Variants within the cargo binding domain show tissue-dependent effects. In summary, dTnpo is an essential gene in flies during development and in neurons. Further, proband-associated de novo variants within TNPO2 disrupt the function of the encoded protein. Hence, TNPO2 variants are causative for neurodevelopmental abnormalities.


Assuntos
Deficiências do Desenvolvimento/genética , Proteínas de Drosophila/genética , Oftalmopatias Hereditárias/genética , Deficiência Intelectual/genética , Carioferinas/genética , Anormalidades Musculoesqueléticas/genética , beta Carioferinas/genética , Proteína ran de Ligação ao GTP/genética , Alelos , Sequência de Aminoácidos , Animais , Deficiências do Desenvolvimento/metabolismo , Deficiências do Desenvolvimento/patologia , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Oftalmopatias Hereditárias/metabolismo , Oftalmopatias Hereditárias/patologia , Feminino , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genoma Humano , Humanos , Lactente , Recém-Nascido , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Carioferinas/antagonistas & inibidores , Carioferinas/metabolismo , Masculino , Anormalidades Musculoesqueléticas/metabolismo , Anormalidades Musculoesqueléticas/patologia , Mutação , Neurônios/metabolismo , Neurônios/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Sequenciamento Completo do Genoma , beta Carioferinas/metabolismo , Proteína ran de Ligação ao GTP/metabolismo
15.
Hum Genet ; 140(8): 1143-1156, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33974130

RESUMO

Biallelic STX3 variants were previously reported in five individuals with the severe congenital enteropathy, microvillus inclusion disease (MVID). Here, we provide a significant extension of the phenotypic spectrum caused by STX3 variants. We report ten individuals of diverse geographic origin with biallelic STX3 loss-of-function variants, identified through exome sequencing, single-nucleotide polymorphism array-based homozygosity mapping, and international collaboration. The evaluated individuals all presented with MVID. Eight individuals also displayed early-onset severe retinal dystrophy, i.e., syndromic-intestinal and retinal-disease. These individuals harbored STX3 variants that affected both the retinal and intestinal STX3 transcripts, whereas STX3 variants affected only the intestinal transcript in individuals with solitary MVID. That STX3 is essential for retinal photoreceptor survival was confirmed by the creation of a rod photoreceptor-specific STX3 knockout mouse model which revealed a time-dependent reduction in the number of rod photoreceptors, thinning of the outer nuclear layer, and the eventual loss of both rod and cone photoreceptors. Together, our results provide a link between STX3 loss-of-function variants and a human retinal dystrophy. Depending on the genomic site of a human loss-of-function STX3 variant, it can cause MVID, the novel intestinal-retinal syndrome reported here or, hypothetically, an isolated retinal dystrophy.


Assuntos
Oftalmopatias Hereditárias/genética , Mucosa Intestinal/metabolismo , Síndromes de Malabsorção/genética , Microvilosidades/patologia , Mucolipidoses/genética , Polimorfismo de Nucleotídeo Único , Proteínas Qa-SNARE/genética , Células Fotorreceptoras Retinianas Cones/metabolismo , Distrofias Retinianas/genética , Idoso , Idoso de 80 Anos ou mais , Oxirredutases do Álcool/genética , Oxirredutases do Álcool/metabolismo , Animais , Autopsia , Proteínas Correpressoras/genética , Proteínas Correpressoras/metabolismo , Oftalmopatias Hereditárias/metabolismo , Oftalmopatias Hereditárias/patologia , Feminino , Regulação da Expressão Gênica , Homozigoto , Humanos , Mucosa Intestinal/patologia , Síndromes de Malabsorção/metabolismo , Síndromes de Malabsorção/patologia , Camundongos , Camundongos Knockout , Microvilosidades/genética , Microvilosidades/metabolismo , Mucolipidoses/metabolismo , Mucolipidoses/patologia , Fenótipo , Proteínas Qa-SNARE/deficiência , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Distrofias Retinianas/metabolismo , Distrofias Retinianas/patologia , Rodopsinas Sensoriais/genética , Rodopsinas Sensoriais/metabolismo , Sequenciamento do Exoma
16.
EBioMedicine ; 67: 103360, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33975254

RESUMO

Ocular morphogenesis in vertebrates is a highly organized process, orchestrated largely by intrinsic genetic programs that exhibit stringent spatiotemporal control. Alternations in these genetic instructions can lead to hereditary or nonhereditary congenital disorders, a major cause of childhood visual impairment, and contribute to common late-onset blinding diseases. Currently, limited treatment options exist for clinical phenotypes involving eye development. This review summarizes recent advances in our understanding of early-onset ocular disorders and highlights genetic complexities in development and diseases, specifically focusing on coloboma, congenital glaucoma and Leber congenital amaurosis. We also discuss innovative paradigms for potential therapeutic modalities.


Assuntos
Oftalmopatias Hereditárias/genética , Criança , Olho/embriologia , Olho/metabolismo , Oftalmopatias Hereditárias/patologia , Oftalmopatias Hereditárias/terapia , Terapia Genética/métodos , Humanos , Terapia de Alvo Molecular/métodos , Transplante de Células-Tronco/métodos
17.
Int J Mol Sci ; 22(9)2021 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-33922602

RESUMO

Mutations in GPR179 lead to autosomal recessive complete congenital stationary night blindness (cCSNB). This condition represents a signal transmission defect from the photoreceptors to the ON-bipolar cells. To confirm the phenotype, better understand the pathogenic mechanism in vivo, and provide a model for therapeutic approaches, a Gpr179 knock-out mouse model was genetically and functionally characterized. We confirmed that the insertion of a neo/lac Z cassette in intron 1 of Gpr179 disrupts the same gene. Spectral domain optical coherence tomography reveals no obvious retinal structure abnormalities. Gpr179 knock-out mice exhibit a so-called no-b-wave (nob) phenotype with severely reduced b-wave amplitudes in the electroretinogram. Optomotor tests reveal decreased optomotor responses under scotopic conditions. Consistent with the genetic disruption of Gpr179, GPR179 is absent at the dendritic tips of ON-bipolar cells. While proteins of the same signal transmission cascade (GRM6, LRIT3, and TRPM1) are correctly localized, other proteins (RGS7, RGS11, and GNB5) known to regulate GRM6 are absent at the dendritic tips of ON-bipolar cells. These results add a new model of cCSNB, which is important to better understand the role of GPR179, its implication in patients with cCSNB, and its use for the development of therapies.


Assuntos
Modelos Animais de Doenças , Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/patologia , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Miopia/genética , Miopia/patologia , Cegueira Noturna/genética , Cegueira Noturna/patologia , Receptores Acoplados a Proteínas G/fisiologia , Retina/patologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Fenótipo , Retina/metabolismo , Transdução de Sinais
18.
Eur J Med Genet ; 64(6): 104209, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33766794

RESUMO

We present a male infant with alveolar capillary dysplasia without misalignment of pulmonary veins, hyperinflammation, megalocornea and macrosomia/macrocephaly at birth. Whole-exome sequencing revealed a homozygous 2bp-insertion in the latent transforming growth factor-beta binding protein 2 (LTBP2) (c.278_279dup, p.(Ser94Glyfs*187)). So far, LTBP2-variants have been frequently reported with an eye-restricted phenotype including primary congenital glaucoma and megalocornea/microspherphakia and ectopia lentis with/without secondary glaucoma. Hitherto reported systemic phenotypes showed, among others, features as tall stature, finger anomalies, high-arched palate and cardiovascular anomalies. The main pathophysiological finding of our patient was an alveolar capillary dysplasia (with pulmonary arterial hypertension and right ventricular impairment but without misalignment of pulmonary veins) resulting in almost continuous oxygen demand and prolonged dependence on mechanical ventilation. He died of respiratory failure at the age of seven months. This patient may extend the LTBP2-related phenotype with resulting diagnostic implications.


Assuntos
Oftalmopatias Hereditárias/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Glaucoma/genética , Proteínas de Ligação a TGF-beta Latente/genética , Síndrome da Persistência do Padrão de Circulação Fetal/genética , Fenótipo , Alvéolos Pulmonares/anormalidades , Doença Cardiopulmonar/genética , Oftalmopatias Hereditárias/patologia , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Glaucoma/patologia , Humanos , Lactente , Masculino , Síndrome da Persistência do Padrão de Circulação Fetal/patologia , Alvéolos Pulmonares/patologia , Doença Cardiopulmonar/patologia , Veias Pulmonares/anormalidades
19.
Genes (Basel) ; 12(3)2021 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-33668843

RESUMO

Hemizygous pathogenic variants in CACNA1F lead to defective signal transmission from retinal photoreceptors to bipolar cells and cause incomplete congenital stationary night blindness in humans. Although the primary defect is at the terminal end of first-order neurons (photoreceptors), there is limited knowledge of higher-order neuronal changes (inner retinal) in this disorder. This study aimed to investigate inner retinal changes in CACNA1F-retinopathy by analyzing macular ganglion cell layer-inner plexiform layer (GCL-IPL) thickness and optic disc pallor in 22 subjects with molecularly confirmed CACNA1F-retinopathy. Detailed ocular phenotypic data including distance and color vision, refraction and electroretinogram (ERG) were collected. Distance vision was universally reduced (mean: 0.42 LogMAR), six had abnormal color vision and myopia was common (n = 15; mean: -6.32 diopters). Mean GCL-IPL thickness was significantly lower in patients (55.00 µm) compared to age-matched controls (n = 87; 84.57 µm; p << 0.001). The GCL-IPL thickness correlated with scotopic standard (p = 0.04) and bright-flash (p = 0.014) ERG b/a ratios and photopic b-wave amplitudes (p = 0.05). Twenty-one patients had some degree of disc pallor (bilateral in 19). Fifteen putative disease-causing, including five novel variants were identified. This study establishes macular inner retinal thinning and optic atrophy as characteristic features of CACNA1F-retinopathy, which are independent of myopia and could impact potential future treatment strategies.


Assuntos
Oftalmopatias Hereditárias/diagnóstico por imagem , Doenças Genéticas Ligadas ao Cromossomo X/diagnóstico por imagem , Miopia/diagnóstico por imagem , Cegueira Noturna/diagnóstico por imagem , Atrofia Óptica/patologia , Retina/patologia , Tomografia de Coerência Óptica/métodos , Adolescente , Adulto , Idoso , Criança , Eletrorretinografia , Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/patologia , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Miopia/genética , Miopia/patologia , Cegueira Noturna/genética , Cegueira Noturna/patologia , Atrofia Óptica/diagnóstico por imagem , Refração Ocular , Retina/diagnóstico por imagem , Estudos Retrospectivos , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA