Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Nat Med ; 28(5): 1014-1021, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35379979

RESUMO

CEP290-associated Leber congenital amaurosis type 10 (LCA10) is a retinal disease resulting in childhood blindness. Sepofarsen is an RNA antisense oligonucleotide targeting the c.2991+1655A>G variant in the CEP290 gene to treat LCA10. In this open-label, phase 1b/2 ( NCT03140969 ), 12-month, multicenter, multiple-dose, dose-escalation trial, six adult patients and five pediatric patients received ≤4 doses of intravitreal sepofarsen into the worse-seeing eye. The primary objective was to evaluate sepofarsen safety and tolerability via the frequency and severity of ocular adverse events (AEs); secondary objectives were to evaluate pharmacokinetics and efficacy via changes in functional outcomes. Six patients received sepofarsen 160 µg/80 µg, and five patients received sepofarsen 320 µg/160 µg. Ten of 11 (90.9%) patients developed ocular AEs in the treated eye (5/6 with 160 µg/80 µg; 5/5 with 320 µg/160 µg) versus one of 11 (9.1%) in the untreated eye; most were mild in severity and dose dependent. Eight patients developed cataracts, of which six (75.0%) were categorized as serious (2/3 with 160 µg/80 µg; 4/5 with 320 µg/160 µg), as lens replacement was required. As the 160-µg/80-µg group showed a better benefit-risk profile, higher doses were discontinued or not initiated. Statistically significant improvements in visual acuity and retinal sensitivity were reported (post hoc analysis). The manageable safety profile and improvements reported in this trial support the continuation of sepofarsen development.


Assuntos
Amaurose Congênita de Leber , Adulto , Antígenos de Neoplasias/genética , Cegueira/genética , Proteínas de Ciclo Celular/genética , Criança , Proteínas do Citoesqueleto/metabolismo , Humanos , Amaurose Congênita de Leber/tratamento farmacológico , Amaurose Congênita de Leber/genética , Oligonucleotídeos Antissenso/efeitos adversos , Visão Ocular
2.
Hum Mol Genet ; 31(15): 2560-2570, 2022 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-35253837

RESUMO

Retinitis pigmentosa (RP) is a genetically heterogeneous form of inherited retinal disease that leads to progressive visual impairment. One genetic subtype of RP, RP54, has been linked to mutations in PCARE (photoreceptor cilium actin regulator). We have recently shown that PCARE recruits WASF3 to the tip of a primary cilium, and thereby activates an Arp2/3 complex which results in the remodeling of actin filaments that drives the expansion of the ciliary tip membrane. On the basis of these findings, and the lack of proper photoreceptor development in mice lacking Pcare, we postulated that PCARE plays an important role in photoreceptor outer segment disk formation. In this study, we aimed to decipher the relationship between predicted structural and function amino acid motifs within PCARE and its function. Our results show that PCARE contains a predicted helical coiled coil domain together with evolutionary conserved binding sites for photoreceptor kinase MAK (type RP62), as well as EVH1 domain-binding linear motifs. Upon deletion of the helical domain, PCARE failed to localize to the cilia. Furthermore, upon deletion of the EVH1 domain-binding motifs separately or together, co-expression of mutant protein with WASF3 resulted in smaller ciliary tip membrane expansions. Finally, inactivation of the lipid modification on the cysteine residue at amino acid position 3 also caused a moderate decrease in the sizes of ciliary tip expansions. Taken together, our data illustrate the importance of amino acid motifs and domains within PCARE in fulfilling its physiological function.


Assuntos
Retinose Pigmentar , Animais , Cílios/genética , Cílios/metabolismo , Camundongos , Ligação Proteica , Domínios Proteicos , Retina/metabolismo , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo
3.
Stem Cell Res ; 60: 102689, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35121194

RESUMO

Leber congenital amaurosis (LCA) can be caused by mutations in more than 20 different genes. One of these, RPE65, encodes a protein essential for the visual cycle that is expressed in retinal pigment epithelium cells. In this work, we describe the generation and characterization of the human iPSC line SCTCi16-A. This hiPSC line was generated from peripheral blood mononuclear cells (PBMCs) from a patient affected with LCA caused by the homozygous c.11+5G>A variant in the RPE65 gene. Reprograming was conducted using episomal vectors containing OCT3/4, SOX2, KLF4, L-MYC, and LIN28.


Assuntos
Células-Tronco Pluripotentes Induzidas , Amaurose Congênita de Leber , Linhagem Celular , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/metabolismo , Leucócitos Mononucleares/metabolismo , Mutação , cis-trans-Isomerases/genética
4.
Oncotarget ; 11(5): 573, 2020 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-32082491

RESUMO

[This corrects the article DOI: 10.18632/oncotarget.26873.].

5.
Eur J Hum Genet ; 26(12): 1819-1823, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30181612

RESUMO

Familial exudative vitreoretinopathy (FEVR) is an inherited retinal disorder, which is primarily characterized by abnormal development of retinal vasculature. In this study, we reported a subject presenting the clinical features of FEVR as well as microcephaly. Screening of the KIF11 gene in this patient revealed a novel heterozygous protein-truncating variant (c.2717del, p.(L906*), NM_004523.3). Segregation analysis in the unaffected parents using Sanger sequencing suggested the variant to be present in a mosaic state in the unaffected mother. KIF11 exon 19 which harbors the variant was amplified from the proband and her father, as well as three different tissues of the mother, followed by amplicon-based deep sequencing. This analysis revealed that the variant is present in different tissues of the mother at various rates, i.e. in blood (16.9%), saliva (20.7%), or skin biopsy-derived fibroblast cells (6.6%). These data demonstrate the importance of deep sequencing in unaffected parents upon detection of a genetic defect in isolated cases to detect possible mosaicisms, enabling a  more reliable recurrence risk assessment and thereby improve genetic counseling.


Assuntos
Oftalmopatias Hereditárias/genética , Cinesinas/genética , Microcefalia/genética , Mosaicismo , Doenças Retinianas/genética , Adolescente , Células Cultivadas , Oftalmopatias Hereditárias/patologia , Vitreorretinopatias Exsudativas Familiares , Feminino , Fibroblastos/metabolismo , Heterozigoto , Humanos , Cinesinas/metabolismo , Microcefalia/patologia , Mutação , Linhagem , Doenças Retinianas/patologia
6.
Invest Ophthalmol Vis Sci ; 59(11): 4384-4391, 2018 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-30193310

RESUMO

Purpose: To describe the phenotypic spectrum of retinal disease caused by the c.2991+1655A>G mutation in CEP290 and to compare disease severity between homozygous and compound heterozygous patients. Methods: Medical records were reviewed for best-corrected visual acuity (BCVA), age of onset, fundoscopy descriptions. Foveal outer nuclear layer (ONL) and ellipsoid zone (EZ) presence was assessed using spectral-domain optical coherence tomography (SD-OCT). Differences between compound heterozygous and homozygous patients were analyzed based on visual performance and visual development. Results: A total of 66 patients were included. The majority of patients had either light perception or no light perception. In the remaining group of 14 patients, median BCVA was 20/195 Snellen (0.99 LogMAR; range 0.12-1.90) for the right eye, and 20/148 Snellen (0.87 LogMAR; range 0.22-1.90) for the left. Homozygous patients tended to be more likely to develop light perception compared to more severely affected compound heterozygous patients (P = 0.080) and are more likely to improve from no light perception to light perception (P = 0.022) before the age of 6 years. OCT data were available in 12 patients, 11 of whom had retained foveal ONL and EZ integrity up to 48 years (median 23 years) of age. Conclusions: Homozygous patients seem less severely affected compared to their compound-heterozygous peers. Improvement of visual function may occur in the early years of life, suggesting a time window for therapeutic intervention up to the approximate age of 17 years. This period may be extended by an intact foveal ONL and EZ on OCT.


Assuntos
Antígenos de Neoplasias/genética , Íntrons/genética , Amaurose Congênita de Leber/diagnóstico , Amaurose Congênita de Leber/genética , Mutação , Proteínas de Neoplasias/genética , Adolescente , Adulto , Proteínas de Ciclo Celular , Criança , Pré-Escolar , Proteínas do Citoesqueleto , Eletrorretinografia , Feminino , Seguimentos , Amplificação de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Amaurose Congênita de Leber/fisiopatologia , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Retina/diagnóstico por imagem , Retina/fisiopatologia , Estudos Retrospectivos , Tomografia de Coerência Óptica , Acuidade Visual/fisiologia , Adulto Jovem
7.
Int J Mol Sci ; 19(3)2018 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-29518907

RESUMO

Leber congenital amaurosis (LCA) is a rare inherited retinal disorder affecting approximately 1:50,000 people worldwide. So far, mutations in 25 genes have been associated with LCA, with CEP290 (encoding the Centrosomal protein of 290 kDa) being the most frequently mutated gene. The most recurrent LCA-causing CEP290 mutation, c.2991+1655A>G, causes the insertion of a pseudoexon into a variable proportion of CEP290 transcripts. We previously demonstrated that antisense oligonucleotides (AONs) have a high therapeutic potential for patients homozygously harbouring this mutation, although to date, it is unclear whether rescuing one single allele is enough to restore CEP290 function. Here, we assessed the AON efficacy at RNA, protein and cellular levels in samples that are compound heterozygous for this mutation, together with a protein-truncating mutation in CEP290. We demonstrate that AONs can efficiently restore splicing and increase protein levels. However, due to a high variability in ciliation among the patient-derived cell lines, the efficacy of the AONs was more difficult to assess at the cellular level. This observation points towards the importance of the severity of the second allele and possibly other genetic variants present in each individual. Overall, AONs seem to be a promising tool to treat CEP290-associated LCA, not only in homozygous but also in compound heterozygous carriers of the c.2991+1655A>G variant.


Assuntos
Alelos , Antígenos de Neoplasias/genética , Heterozigoto , Amaurose Congênita de Leber/genética , Mutação , Proteínas de Neoplasias/genética , Oligonucleotídeos Antissenso , Splicing de RNA , Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular , Proteínas do Citoesqueleto , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Amaurose Congênita de Leber/metabolismo , Masculino , Proteínas de Neoplasias/metabolismo
8.
Am J Hum Genet ; 100(6): 960-968, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28575650

RESUMO

Familial exudative vitreoretinopathy (FEVR) is an inherited blinding disorder characterized by the abnormal development of the retinal vasculature. The majority of mutations identified in FEVR are found within four genes that encode the receptor complex (FZD4, LRP5, and TSPAN12) and ligand (NDP) of a molecular pathway that controls angiogenesis, the Norrin-ß-catenin signaling pathway. However, half of all FEVR-affected case subjects do not harbor mutations in these genes, indicating that further mutated genes remain to be identified. Here we report the identification of mutations in CTNNB1, the gene encoding ß-catenin, as a cause of FEVR. We describe heterozygous mutations (c.2142_2157dup [p.His720∗] and c.2128C>T [p.Arg710Cys]) in two dominant FEVR-affected families and a de novo mutation (c.1434_1435insC [p.Glu479Argfs∗18]) in a simplex case subject. Previous studies have reported heterozygous de novo CTNNB1 mutations as a cause of syndromic intellectual disability (ID) and autism spectrum disorder, and somatic mutations are linked to many cancers. However, in this study we show that Mendelian inherited CTNNB1 mutations can cause non-syndromic FEVR and that FEVR can be a part of the syndromic ID phenotype, further establishing the role that ß-catenin signaling plays in the development of the retinal vasculature.


Assuntos
Doenças Retinianas/genética , Transdução de Sinais , beta Catenina/metabolismo , Sequência de Bases , Oftalmopatias Hereditárias , Vitreorretinopatias Exsudativas Familiares , Feminino , Heterozigoto , Humanos , Luciferases/metabolismo , Masculino , Modelos Biológicos , Proteínas Mutantes/metabolismo , Mutação/genética , Linhagem , Fenótipo , Transcrição Gênica
9.
Hum Mol Genet ; 25(12): 2552-2563, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27106101

RESUMO

Leber congenital amaurosis (LCA) is a severe disorder resulting in visual impairment usually starting in the first year of life. The most frequent genetic cause of LCA is an intronic mutation in CEP290 (c.2991 + 1655A > G) that creates a cryptic splice donor site resulting in the insertion of a pseudoexon (exon X) into CEP290 mRNA. Previously, we showed that naked antisense oligonucleotides (AONs) effectively restored normal CEP290 splicing in patient-derived lymphoblastoid cells. We here explore the therapeutic potential of naked and adeno-associated virus (AAV)-packaged AONs in vitro and in vivo In both cases, AON delivery fully restored CEP290 pre-mRNA splicing, significantly increased CEP290 protein levels and rescued a ciliary phenotype present in patient-derived fibroblast cells. Moreover, administration of naked and AAV-packaged AONs to the retina of a humanized mutant Cep290 mouse model, carrying the intronic mutation, showed a statistically significant reduction of exon X-containing Cep290 transcripts, without compromising the retinal structure. Together, our data highlight the tremendous therapeutic prospective of AONs for the treatment of not only CEP290-associated LCA but potentially many other subtypes of retinal dystrophy caused by splicing mutations.


Assuntos
Antígenos de Neoplasias/genética , Cegueira/terapia , Terapia Genética , Amaurose Congênita de Leber/terapia , Proteínas de Neoplasias/genética , Oligonucleotídeos Antissenso/administração & dosagem , Animais , Antígenos de Neoplasias/uso terapêutico , Cegueira/genética , Cegueira/patologia , Proteínas de Ciclo Celular , Proteínas do Citoesqueleto , Dependovirus/genética , Modelos Animais de Doenças , Éxons/genética , Humanos , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/patologia , Camundongos , Mutação , Proteínas de Neoplasias/uso terapêutico , Oligonucleotídeos Antissenso/genética , Fenótipo , Sítios de Splice de RNA/genética , Splicing de RNA/genética , Retina/efeitos dos fármacos , Retina/patologia
10.
Adv Exp Med Biol ; 854: 517-24, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26427454

RESUMO

Inherited retinal dystrophies (IRDs) are an extremely heterogeneous group of genetic diseases for which currently no effective treatment strategies exist. Over the last decade, significant progress has been made utilizing gene augmentation therapy for a few genetic subtypes of IRD, although several technical challenges so far prevent a broad clinical application of this approach for other forms of IRD. Many of the mutations leading to these retinal diseases affect pre-mRNA splicing of the mutated genes . Antisense oligonucleotide (AON)-mediated splice modulation appears to be a powerful approach to correct the consequences of such mutations at the pre-mRNA level , as demonstrated by promising results in clinical trials for several inherited disorders like Duchenne muscular dystrophy, hypercholesterolemia and various types of cancer. In this mini-review, we summarize ongoing pre-clinical research on AON-based therapy for a few genetic subtypes of IRD , speculate on other potential therapeutic targets, and discuss the opportunities and challenges that lie ahead to translate splice modulation therapy for retinal disorders to the clinic.


Assuntos
Oligonucleotídeos Antissenso/genética , Distrofias Retinianas/genética , Distrofias Retinianas/terapia , Reparo Gênico Alvo-Dirigido/métodos , Animais , Modelos Animais de Doenças , Predisposição Genética para Doença/genética , Humanos , Mutação , Precursores de RNA/genética , Splicing de RNA/genética
11.
Eur J Hum Genet ; 24(7): 1071-9, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26626312

RESUMO

Leber congenital amaurosis (LCA) represents the most severe form of inherited retinal dystrophies with an onset during the first year of life. Currently, 21 genes are known to be associated with LCA and recurrent mutations have been observed in AIPL1, CEP290, CRB1 and GUCY2D. In addition, sequence analysis of LRAT and RPE65 may be important in view of treatments that are emerging for patients carrying variants in these genes. Screening of the aforementioned variants and genes was performed in 64 Danish LCA probands. Upon the identification of heterozygous variants, Sanger sequencing was performed of the relevant genes to identify the second allele. In combination with prior arrayed primer extension analysis, this led to the identification of two variants in 42 of 86 cases (49%). Remarkably, biallelic RPE65 variants were identified in 16% of the cases, and one novel variant, p.(D110G), was found in seven RPE65 alleles. We also collected all previously published RPE65 variants, identified in 914 alleles of 539 patients with LCA or early-onset retinitis pigmentosa, and deposited them in the RPE65 Leiden Open Variation Database (LOVD). The in silico pathogenicity assessment of the missense and noncanonical splice site variants, as well as an analysis of their frequency in ~60 000 control individuals, rendered 864 of the alleles to affect function or probably affect function. This comprehensive database can now be used to select patients eligible for gene augmentation or retinoid supplementation therapies.


Assuntos
Amaurose Congênita de Leber/genética , Taxa de Mutação , cis-trans-Isomerases/genética , Adulto , Criança , Dinamarca , Feminino , Heterozigoto , Humanos , Lactente , Amaurose Congênita de Leber/patologia , Masculino , Mutação de Sentido Incorreto , Linhagem , Splicing de RNA
12.
PLoS One ; 10(3): e0119806, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25775262

RESUMO

BACKGROUND: Homozygosity mapping has facilitated the identification of the genetic causes underlying inherited diseases, particularly in consanguineous families with multiple affected individuals. This knowledge has also resulted in a mutation dataset that can be used in a cost and time effective manner to screen frequent population-specific genetic variations associated with diseases such as inherited retinal disease (IRD). METHODS: We genetically screened 13 families from a cohort of 81 Pakistani IRD families diagnosed with Leber congenital amaurosis (LCA), retinitis pigmentosa (RP), congenital stationary night blindness (CSNB), or cone dystrophy (CD). We employed genome-wide single nucleotide polymorphism (SNP) array analysis to identify homozygous regions shared by affected individuals and performed Sanger sequencing of IRD-associated genes located in the sizeable homozygous regions. In addition, based on population specific mutation data we performed targeted Sanger sequencing (TSS) of frequent variants in AIPL1, CEP290, CRB1, GUCY2D, LCA5, RPGRIP1 and TULP1, in probands from 28 LCA families. RESULTS: Homozygosity mapping and Sanger sequencing of IRD-associated genes revealed the underlying mutations in 10 families. TSS revealed causative variants in three families. In these 13 families four novel mutations were identified in CNGA1, CNGB1, GUCY2D, and RPGRIP1. CONCLUSIONS: Homozygosity mapping and TSS revealed the underlying genetic cause in 13 IRD families, which is useful for genetic counseling as well as therapeutic interventions that are likely to become available in the near future.


Assuntos
Mutação , Doenças Retinianas/genética , Consanguinidade , Análise Mutacional de DNA , Feminino , Genótipo , Homozigoto , Humanos , Amaurose Congênita de Leber/epidemiologia , Amaurose Congênita de Leber/genética , Masculino , Paquistão/epidemiologia , Linhagem , Polimorfismo de Nucleotídeo Único , Retina/metabolismo , Retina/patologia , Doenças Retinianas/epidemiologia , Retinose Pigmentar/epidemiologia , Retinose Pigmentar/genética
13.
Int J Mol Sci ; 16(3): 5285-98, 2015 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-25761237

RESUMO

A mutation in intron 26 of CEP290 (c.2991+1655A>G) is the most common genetic cause of Leber congenital amaurosis (LCA), a severe type of inherited retinal degeneration. This mutation creates a cryptic splice donor site, resulting in the insertion of an aberrant exon (exon X) into ~50% of all CEP290 transcripts. A humanized mouse model with this mutation did not recapitulate the aberrant CEP290 splicing observed in LCA patients, suggesting differential recognition of cryptic splice sites between species. To further assess this phenomenon, we generated two CEP290 minigene constructs, with and without the intronic mutation, and transfected these in cell lines of various species. RT-PCR analysis revealed that exon X is well recognized by the splicing machinery in human and non-human primate cell lines. Intriguingly, this recognition decreases in cell lines derived from species such as dog and rodents, and it is completely absent in Drosophila. In addition, other cryptic splicing events corresponding to sequences in intron 26 of CEP290 were observed to varying degrees in the different cell lines. Together, these results highlight the complexity of splice site recognition among different species, and show that care is warranted when generating animal models to mimic splice site mutations in vivo.


Assuntos
Amaurose Congênita de Leber/genética , Proteínas Nucleares/genética , Animais , Antígenos de Neoplasias , Sequência de Bases , Proteínas de Ciclo Celular , Linhagem Celular , Proteínas do Citoesqueleto , Cães , Drosophila/genética , Éxons , Células HEK293 , Humanos , Íntrons , Amaurose Congênita de Leber/patologia , Camundongos , Mutação , Proteínas Nucleares/metabolismo , Splicing de RNA , Especificidade da Espécie
14.
Mol Vis ; 20: 753-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24940029

RESUMO

PURPOSE: The gene encoding nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) was recently found to be mutated in a subset of patients with Leber congenital amaurosis (LCA) with macular atrophy. The aim of this study was to determine the occurrence and frequency of NMNAT1 mutations and associated phenotypes in different types of inherited retinal dystrophies. METHODS: DNA samples of 161 patients with LCA without genetic diagnosis were analyzed for variants in NMNAT1 using Sanger sequencing. Variants in exon 5 of NMNAT1, which harbors the majority of the previously identified mutations, were screened in 532 additional patients with retinal dystrophies. This cohort encompassed 108 persons with isolated or autosomal recessive cone-rod dystrophy (CRD), 271 with isolated or autosomal recessive retinitis pigmentosa (RP), and 49 with autosomal dominant RP, as well as 104 persons with LCA in whom the causative mutation was previously identified. RESULTS: Compound heterozygous alterations were found in six patients with LCA and in one person with early-onset RP. All except one carried the common p.E257K variant on one allele. Macular atrophy was absent in one patient, who carried this variant in combination with a truncating mutation on the other allele. The p.E257K alteration was also found in a heterozygous state in five individuals with LCA and one with RP while no mutation was detected on the other allele. Two individuals with LCA carried other NMNAT1 variants in a heterozygous state, whereas no NMNAT1 variants in exon 5 were identified in individuals with CRD. The p.E257K variant was found to be enriched in a heterozygous state in individuals with LCA (0.94%) compared to Caucasian controls (0.18%), although the difference was statistically insignificant (p=0.12). CONCLUSIONS: Although macular atrophy can occur in LCA and CRD, no NMNAT1 mutations were found in the latter cohort. NMNAT1 variants were also not found in a large group of patients with sporadic or autosomal recessive RP. The enrichment of p.E257K in a heterozygous state in patients with LCA versus controls suggests that this allele could act as a modifier in other genetic subtypes of LCA.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Heterozigoto , Amaurose Congênita de Leber/enzimologia , Amaurose Congênita de Leber/genética , Nicotinamida-Nucleotídeo Adenililtransferase/genética , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação/genética , Adulto Jovem
15.
JAMA Ophthalmol ; 132(8): 1002-4, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24830548

RESUMO

IMPORTANCE: The NMNAT1 gene was recently found to be mutated in a subset of patients with Leber congenital amaurosis and macular atrophy. The most prevalent NMNAT1 variant was p.Glu257Lys, which was observed in 38 of 106 alleles (35.8%). On the basis of functional assays, it was deemed a severe variant. OBSERVATIONS: The p.Glu257Lys variant was 80-fold less frequent in a homozygous state in patients with Leber congenital amaurosis than predicted based on its heterozygosity frequency in the European American population. Moreover, we identified this variant in a homozygous state in a patient with no ocular abnormalities. CONCLUSIONS AND RELEVANCE: On the basis of these results, the p.Glu257Lys variant is considered not fully penetrant. Homozygotes of the p.Glu257Lys variant in most persons are therefore not associated with ocular disease. Consequently, genetic counselors should exercise great caution in the interpretation of this variant.


Assuntos
Amaurose Congênita de Leber/genética , Mutação , Nicotinamida-Nucleotídeo Adenililtransferase/genética , Humanos
16.
J Med Genet ; 51(3): 143-51, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24401286

RESUMO

Many proteins depend on post-translational prenylation for a correct subcellular localisation and membrane anchoring. This involves the covalent attachment of farnesyl or geranylgeranyl residues to cysteines residing in consensus motifs at the C-terminal parts of proteins. Retinal photoreceptor cells are highly compartmentalised and membranous structures, and therefore it can be expected that the proper function of many retinal proteins depends on prenylation, which has been proven for several proteins that are absent or defective in different inherited retinal diseases (IRDs). These include proteins involved in the phototransduction cascade, such as GRK1, the phosphodiesterase 6 subunits and the transducin γ subunit, or proteins involved in transport processes, such as RAB28 and retinitis pigmentosa GTPase regulator (RPGR). In addition, there is another class of general prenylation defects due to mutations in proteins such as AIPL1, PDE6D and rab escort protein-1 (REP-1), which can act as chaperones for subsets of prenylated retinal proteins that are associated with IRDs. REP-1 also is a key accessory protein of geranylgeranyltransferase II, an enzyme involved in the geranylgeranylation of almost all members of a large family of Rab GTPases. Finally, mutations in the mevalonate kinase (MVK) gene, which were known to be principally associated with mevalonic aciduria, were recently associated with non-syndromic retinitis pigmentosa. We hypothesise that MVK deficiency results in a depletion of prenyl moieties that affects the prenylation of many proteins synthesised specifically in the retina, including Rabs. In this review, we discuss the entire spectrum of prenylation defects underlying progressive degeneration of photoreceptors, the retinal pigment epithelium and the choroid.


Assuntos
Prenilação de Proteína , Doenças Retinianas , Animais , Humanos , Camundongos
17.
PLoS One ; 8(11): e79369, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24223178

RESUMO

Leber congenital amaurosis (LCA) is the most severe form of retinal dystrophy with an onset in the first year of life. The most frequent genetic cause of LCA, accounting for up to 15% of all LCA cases in Europe and North-America, is a mutation (c.2991+1655AG) in intron 26 of CEP290. This mutation generates a cryptic splice donor site resulting in the insertion of an aberrant exon (exon X) containing a premature stop codon to CEP290 mRNA. In order to study the pathophysiology of the intronic CEP290 mutation, we generated two humanized knock-in mouse models each carrying ~6.3 kb of the human CEP290 gene, either with or without the intronic mutation. Transcriptional characterization of these mouse models revealed an unexpected splice pattern of CEP290 mRNA, especially in the retina. In both models, a new cryptic exon (coined exon Y) was identified in ~5 to 12% of all Cep290 transcripts. This exon Y was expressed in all murine tissues analyzed but not detected in human retina or fibroblasts of LCA patients. In addition, exon x that is characteristic of LCA in humans, was expressed at only very low levels in the retina of the LCA mouse model. Western blot and immunohistochemical analyses did not reveal any differences between the two transgenic models and wild-type mice. Together, our results show clear differences in the recognition of splice sites between mice and humans, and emphasize that care is warranted when generating animal models for human genetic diseases caused by splice mutations.


Assuntos
Antígenos de Neoplasias/genética , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Amaurose Congênita de Leber/genética , Proteínas de Neoplasias/genética , Splicing de RNA , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Proteínas do Citoesqueleto , Éxons/genética , Feminino , Humanos , Camundongos , Gravidez , Retina/metabolismo , Especificidade da Espécie , Transcrição Gênica/genética
18.
Mol Vis ; 17: 3013-24, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22128245

RESUMO

PURPOSE: Retinitis pigmentosa (RP) is a clinically and genetically heterogeneous retinal disorder. Despite tremendous knowledge about the genes involved in RP, little is known about the genetic causes of RP in Indonesia. Here, we aim to identify the molecular genetic causes underlying RP in a small cohort of Indonesian patients, using genome-wide homozygosity mapping. METHODS: DNA samples from affected and healthy individuals from 14 Indonesian families segregating autosomal recessive, X-linked, or isolated RP were collected. Homozygosity mapping was conducted using Illumina 6k or Affymetrix 5.0 single nucleotide polymorphism (SNP) arrays. Known autosomal recessive RP (arRP) genes residing in homozygous regions and X-linked RP genes were sequenced for mutations. RESULTS: In ten out of the 14 families, homozygous regions were identified that contained genes known to be involved in the pathogenesis of RP. Sequence analysis of these genes revealed seven novel homozygous mutations in ATP-binding cassette, sub-family A, member 4 (ABCA4), crumbs homolog 1 (CRB1), eyes shut homolog (Drosophila) (EYS), c-mer proto-oncogene tyrosine kinase (MERTK), nuclear receptor subfamily 2, group E, member 3 (NR2E3) and phosphodiesterase 6A, cGMP-specific, rod, alpha (PDE6A), all segregating in the respective families. No mutations were identified in the X-linked genes retinitis pigmentosa GTPase regulator (RPGR) and retinitis pigmentosa 2 (X-linked recessive; RP2). CONCLUSIONS: Homozygosity mapping is a powerful tool to identify the genetic defects underlying RP in the Indonesian population. Compared to studies involving patients from other populations, the same genes appear to be implicated in the etiology of recessive RP in Indonesia, although all mutations that were discovered are novel and as such may be unique for this population.


Assuntos
Povo Asiático , Mapeamento Cromossômico/métodos , Proteínas do Olho/genética , Genes Recessivos , Genoma Humano , Homozigoto , Retinose Pigmentar/genética , Adolescente , Adulto , Idoso , Criança , Estudos de Coortes , Análise Mutacional de DNA , Eletrorretinografia , Feminino , Genes Ligados ao Cromossomo X , Testes Genéticos , Estudo de Associação Genômica Ampla , Humanos , Indonésia/epidemiologia , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação , Linhagem , Polimorfismo de Nucleotídeo Único , Proto-Oncogene Mas , Retinose Pigmentar/etnologia , Retinose Pigmentar/patologia
19.
Invest Ophthalmol Vis Sci ; 52(7): 4774-9, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21421862

RESUMO

PURPOSE: To investigate COL9A1 in two families suggestive of autosomal recessive Stickler syndrome and to delineate the associated phenotype. METHODS: The probands of two consanguineous autosomal recessive Stickler families were evaluated for homozygosity using SNP microarray in one and haplotype analysis in the other. Subsequently, the entire COL9A1 open reading frame was analyzed by DNA sequencing in all members of the respective families. Several family members were investigated for dysmorphic features as well as ophthalmic, audiologic, and radiologic abnormalities. RESULTS: A novel homozygous COL9A1 mutation (p.R507X) was identified in two affected Turkish sisters, and the previously published mutation (p.R295X) was found in a Moroccan boy. Ophthalmic assessment revealed myopia, cataracts, distinct vitreous changes, progressive chorioretinal degeneration, and exudative and rhegmatogenous retinal detachments. All three had sensorineural hearing loss and epiphyseal dysplasia. Intervertebral disc bulging was observed in one patient and in two heterozygous carriers of the p.R507X mutation. CONCLUSIONS: A second, novel mutation was identified in COL9A1, causing autosomal recessive Stickler syndrome together with the previously described nucleotide change in two separate families. Although the overall phenotype was comparable to autosomal dominant Stickler, vitreous changes that may enable recognition of patients who are likely to carry mutations in COL9A1 were identified, and exudative retinal detachment was observed as a new finding in Stickler syndrome.


Assuntos
Colágeno Tipo IX/genética , DNA/genética , Predisposição Genética para Doença , Mutação , Adulto , Idoso , Artrite/diagnóstico , Artrite/genética , Artrite/fisiopatologia , Doenças do Tecido Conjuntivo/diagnóstico , Doenças do Tecido Conjuntivo/genética , Doenças do Tecido Conjuntivo/fisiopatologia , Análise Mutacional de DNA , Feminino , Perda Auditiva Neurossensorial/diagnóstico , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Prognóstico , Descolamento Retiniano/diagnóstico , Descolamento Retiniano/genética , Descolamento Retiniano/fisiopatologia , Tomografia de Coerência Óptica , Acuidade Visual
20.
Invest Ophthalmol Vis Sci ; 52(2): 834-9, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-20881296

RESUMO

PURPOSE: Leber congenital amaurosis (LCA) is genetically heterogeneous, with 15 genes identified thus far, accounting for ∼70% of LCA patients. The aim of the present study was to identify new genetic causes of LCA. METHODS: Homozygosity mapping in >150 LCA patients of worldwide origin was performed with high-density SNP microarrays to identify new disease-causing genes. RESULTS: In three isolated LCA patients, the authors identified large homozygous regions on chromosome 3 encompassing the IQCB1 gene, which has been associated with Senior-Loken syndrome (SLSN), characterized by nephronophthisis and retinal degeneration. Mutation analysis of IQCB1 in these three patients and a subsequent cohort of 222 additional LCA patients identified frameshift and nonsense mutations in 11 patients diagnosed with LCA. On re-inspection of the patient's disease status, seven were found to have developed SLSN, but four maintained the diagnosis of LCA as the kidney function remained normal. CONCLUSIONS: Results show that the onset of renal failure in patients with IQCB1 mutations is highly variable, and that mutations are also found in LCA patients without nephronophthisis, rendering IQCB1 a new gene for LCA. However, these patients are at high risk for developing renal failure, which in early stages is often not recognized and can cause sudden death from fluid and electrolyte imbalance. It is therefore recommended that all LCA patients be screened for IQCB1 mutations, to follow them more closely for kidney disease.


Assuntos
Proteínas de Ligação a Calmodulina/genética , Códon sem Sentido , Mutação da Fase de Leitura , Amaurose Congênita de Leber/genética , Adolescente , Adulto , Criança , Pré-Escolar , Cromossomos Humanos Par 3/genética , Ciliopatias , Análise Mutacional de DNA , Feminino , Genótipo , Humanos , Doenças Renais Císticas/genética , Amaurose Congênita de Leber/diagnóstico , Masculino , Pessoa de Meia-Idade , Atrofias Ópticas Hereditárias/genética , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA