Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 58(8): 570-578, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32817297

RESUMEN

BACKGROUND: Inherited retinal disorders are a clinically and genetically heterogeneous group of conditions and a major cause of visual impairment. Common disease subtypes include vitelliform macular dystrophy (VMD) and retinitis pigmentosa (RP). Despite the identification of over 90 genes associated with RP, conventional genetic testing fails to detect a molecular diagnosis in about one third of patients with RP. METHODS: Exome sequencing was carried out for identifying the disease-causing gene in a family with autosomal dominant RP. Gene panel testing and exome sequencing were performed in 596 RP and VMD families to identified additional IMPG1 variants. In vivo analysis in the medaka fish system by knockdown assays was performed to screen IMPG1 possible pathogenic role. RESULTS: Exome sequencing of a family with RP revealed a splice variant in IMPG1. Subsequently, the same variant was identified in individuals from two families with either RP or VMD. A retrospective study of patients with RP or VMD revealed eight additional families with different missense or nonsense variants in IMPG1. In addition, the clinical diagnosis of the IMPG1 retinopathy-associated variant, originally described as benign concentric annular macular dystrophy, was also revised to RP with early macular involvement. Using morpholino-mediated ablation of Impg1 and its paralog Impg2 in medaka fish, we confirmed a phenotype consistent with that observed in the families, including a decreased length of rod and cone photoreceptor outer segments. CONCLUSION: This study discusses a previously unreported association between monoallelic or biallelic IMPG1 variants and RP. Notably, similar observations have been reported for IMPG2.


Asunto(s)
Proteínas de la Matriz Extracelular , Proteínas del Ojo , Genes Recesivos , Predisposición Genética a la Enfermedad , Mutación , Proteoglicanos , Retinitis Pigmentosa , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Exoma/genética , Secuenciación del Exoma/métodos , Proteínas de la Matriz Extracelular/genética , Proteínas del Ojo/genética , Genes Recesivos/genética , Predisposición Genética a la Enfermedad/genética , Patrón de Herencia/genética , Degeneración Macular/genética , Mutación/genética , Linaje , Fenotipo , Proteoglicanos/genética , Retina/patología , Retinitis Pigmentosa/genética , Estudios Retrospectivos
2.
Hum Mol Genet ; 27(15): 2689-2702, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29771326

RESUMEN

CEP290 mutations cause a spectrum of ciliopathies from Leber congenital amaurosis type 10 (LCA10) to embryo-lethal Meckel syndrome (MKS). Using panel-based molecular diagnosis testing for inherited retinal diseases, we identified two individuals with some preserved vision despite biallelism for presumably truncating CEP290 mutations. The first one carried a homozygous 1 base pair deletion in Exon 17, introducing a premature termination codon (PTC) in Exon 18 (c.1666del; p.Ile556Phefs*17). mRNA analysis revealed a basal exon skipping (BES) of Exon 18, providing mutant cells with the ability to escape protein truncation, while disrupting the reading frame in controls. The second individual harbored compound heterozygous nonsense mutations in Exon 8 (c.508A>T, p.Lys170*) and Exon 32 (c.4090G>T, p.Glu1364*), respectively. Some CEP290 lacking Exon 8 were detected in mutant fibroblasts but not in controls whereas some skipping of Exon 32 occurred in both lines, but with higher amplitude in the mutant. Considering that the deletion of either exon maintains the reading frame in either line, skipping in mutant cells likely involves nonsense-associated altered splicing alone (Exon 8), or with BES (Exon 32). Skipping of PTC-containing exons in mutant cells allowed production of CEP290 isoforms with preserved ability to assemble into a high molecular weight complex and to interact efficiently with proteins important for cilia formation and intraflagellar trafficking. In contrast, studying LCA10 and MKS fibroblasts we show moderate to severe cilia alterations, providing support for a correlation between disease severity and the ability of cells to express shortened, yet functional, CEP290 isoforms.


Asunto(s)
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Enfermedades de la Retina/genética , Adolescente , Adulto , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrosoma/metabolismo , Niño , Cilios/fisiología , Codón sin Sentido , Codón de Terminación , Proteínas del Citoesqueleto , Exones , Proteínas del Ojo/metabolismo , Femenino , Fibroblastos/fisiología , Humanos , Masculino , Mutación , Transporte de Proteínas , Empalme del ARN , Enfermedades de la Retina/etiología , Enfermedades de la Retina/patología , Proteínas Supresoras de Tumor/metabolismo
3.
Hum Mutat ; 40(1): 31-35, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30341801

RESUMEN

Choroideremia is a monogenic X-linked recessive chorioretinal disease linked to pathogenic variants in the CHM gene. These variants are commonly base-pair changes, frameshifts, or large deletions. However, a few rare or unusual events comprising large duplications, a retrotransposon insertion, a pseudo-exon activation, and two c-98 promoter substitutions have also been described. Following an exhaustive molecular diagnosis, we identified and characterized three novel atypical disease-causing variants in three unrelated male patients. One is a first-ever reported Alu insertion within CHM and the other two are nucleotide substitutions, c.-90C>G and c.-108A>G, affecting highly conserved promoter positions. RNA analysis combined with western blot and functional assays of patient cells established the pathogenicity of the Alu insertion and the c.-90C>G alteration. Furthermore, luciferase reporter assays suggested a CHM transcription defect associated with the c.-90C>G and c.-108A>G variants. These findings broaden our knowledge of the mutational spectrum and the transcriptional regulation of the CHM gene.


Asunto(s)
Coroideremia/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Elementos Alu/genética , Secuencia de Bases , Exones/genética , Humanos , Regiones Promotoras Genéticas/genética
4.
Hum Mol Genet ; 26(18): 3573-3584, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28911202

RESUMEN

Choroideremia (CHM) is an inherited retinal dystrophy characterised by progressive degeneration of photoreceptors, retinal pigment epithelium (RPE) and underlying choroid. It is caused by loss-of-function mutations in CHM, which has an X-linked inheritance, and is thus an ideal candidate for gene replacement strategies. CHM encodes REP1, which plays a key role in the prenylation of Rab GTPases. We recently showed that an induced pluripotent stem cell (iPSc)-derived RPE model for CHM is fully functional and reproduces the underlying prenylation defect. This criterion can thus be used for testing the pathogenic nature of novel variants. Until recently, missense variants were not associated with CHM. Currently, at least nine such variants have been reported but only two have been shown to be pathogenic. We report here the characterisation of the third pathogenic missense CHM variant, p.Leu457Pro. Clinically, the associated phenotype is indistinguishable from that of loss-of-function mutations. By contrast, this missense variant results in wild type CHM expression levels and detectable levels of mutant protein. The prenylation status of patient-specific fibroblasts and iPSc-derived RPE is within the range observed for loss-of-function mutations, consistent with the clinical phenotype. Lastly, considering the current climate of CHM gene therapy, we assayed whether the presence of mutant REP1 could interfere with a gene replacement strategy by testing the prenylation status of patient-specific iPSc-derived RPE following AAV-mediated gene transfer. Our results show that correction of the functional defect is possible and highlight the predictive value of these models for therapy screening prior to inclusion in clinical trials.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Coroideremia/genética , Coroides/metabolismo , Coroideremia/terapia , Fibroblastos/metabolismo , Genes Ligados a X/genética , Terapia Genética/métodos , Humanos , Células Madre Pluripotentes Inducidas , Mutación , Mutación Missense/genética , Linaje , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Proteínas de Unión al GTP rab/metabolismo
5.
Hum Mol Genet ; 26(22): 4367-4374, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28973654

RESUMEN

In this study, we report a novel duplication causing North Carolina macular dystrophy (NCMD) identified applying whole genome sequencing performed on eight affected members of two presumed unrelated families mapping to the MCDR1 locus. In our families, the NCMD phenotype was associated with a 98.4 kb tandem duplication encompassing the entire CCNC and PRDM13 genes and a common DNase 1 hypersensitivity site. To study the impact of PRDM13 or CCNC dysregulation, we used the Drosophila eye development as a model. Knock-down and overexpression of CycC and CG13296, Drosophila orthologues of CCNC and PRDM13, respectively, were induced separately during eye development. In flies, eye development was not affected, while knocking down either CycC or CG13296 mutant models. Overexpression of CycC also had no effect. Strikingly, overexpression of CG13296 in Drosophila leads to a severe loss of the imaginal eye-antennal disc. This study demonstrated for the first time in an animal model that overexpression of PRDM13 alone causes a severe abnormal retinal development. It is noteworthy that mutations associated with this autosomal dominant foveal developmental disorder are frequently duplications always including an entire copy of PRDM13, or variants in one DNase 1 hypersensitivity site at this locus.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , Ciclina C/genética , N-Metiltransferasa de Histona-Lisina/genética , Adulto , Animales , Mapeo Cromosómico , Cromosomas Humanos Par 6 , Distrofias Hereditarias de la Córnea/metabolismo , Ciclina C/metabolismo , Drosophila melanogaster , Proteínas del Ojo/genética , Femenino , Ligamiento Genético , Haplotipos , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Masculino , Dominios PR-SET , Linaje , Secuenciación Completa del Genoma
6.
Hum Genet ; 138(5): 441-453, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30904946

RESUMEN

Retinitis pigmentosa (RP) is a heterogeneous group of inherited retinal disorders eventually leading to blindness with different ages of onset, progression and severity. Human RP, first characterized by the progressive degeneration of rod photoreceptor cells, shows high genetic heterogeneity with more than 90 genes identified. However, about one-third of patients have no known genetic causes. Interestingly, dogs are also severely affected by similar diseases, called progressive retinal atrophy (PRA). Indeed, RP and PRA have comparable clinical signs, physiopathology and outcomes, similar diagnosis methods and most often, orthologous genes are involved. The many different dog PRAs often segregate in specific breeds. Indeed, undesired alleles have been selected and amplified through drastic selection and excessive use of inbreeding. Out of the 400 breeds, nearly 100 have an inherited form of PRA, which are natural animal models that can be used to investigate the genetics, disease progression and therapies in dogs for the benefit of both dogs and humans. Recent knowledge on the canine genome and access to new genotyping and sequencing technologies now efficiently allows the identification of mutations involved in canine genetic diseases. To date, PRA genes identified in dog breeds correspond to the same genes in humans and represent relevant RP models, and new genes found in dogs represent good candidate for still unknown human RP. We present here a review of the main advantages of the dog models for human RP with the genes already identified and an X-linked PRA in the Border collie as a model for orphan X-linked RPs in human.


Asunto(s)
Enfermedades de los Perros/genética , Degeneración Retiniana/genética , Degeneración Retiniana/veterinaria , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/genética , Animales , Modelos Animales de Enfermedad , Enfermedades de los Perros/patología , Perros , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Degeneración Retiniana/patología , Retinitis Pigmentosa/patología
7.
Am J Hum Genet ; 98(5): 1011-1019, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27063057

RESUMEN

Congenital stationary night blindness (CSNB) is a heterogeneous group of non-progressive inherited retinal disorders with characteristic electroretinogram (ERG) abnormalities. Riggs and Schubert-Bornschein are subtypes of CSNB and demonstrate distinct ERG features. Riggs CSNB demonstrates selective rod photoreceptor dysfunction and occurs due to mutations in genes encoding proteins involved in rod phototransduction cascade; night blindness is the only symptom and eye examination is otherwise normal. Schubert-Bornschein CSNB is a consequence of impaired signal transmission between the photoreceptors and bipolar cells. Schubert-Bornschein CSNB is subdivided into complete CSNB with an ON bipolar signaling defect and incomplete CSNB with both ON and OFF pathway involvement. Both subtypes are associated with variable degrees of night blindness or photophobia, reduced visual acuity, high myopia, and nystagmus. Whole-exome sequencing of a family screened negative for mutations in genes associated with CSNB identified biallelic mutations in the guanine nucleotide-binding protein subunit beta-3 gene (GNB3). Two siblings were compound heterozygous for a deletion (c.170_172delAGA [p.Lys57del]) and a nonsense mutation (c.1017G>A [p.Trp339(∗)]). The maternal aunt was homozygous for the nonsense mutation (c.1017G>A [p.Trp339(∗)]). Mutational analysis of GNB3 in a cohort of 58 subjects with CSNB identified a sporadic case individual with a homozygous GNB3 mutation (c.200C>T [p.Ser67Phe]). GNB3 encodes the ß subunit of G protein heterotrimer (Gαßγ) and is known to modulate ON bipolar cell signaling and cone transducin function in mice. Affected human subjects showed an unusual CSNB phenotype with variable degrees of ON bipolar dysfunction and reduced cone sensitivity. This unique retinal disorder with dual anomaly in visual processing expands our knowledge about retinal signaling.


Asunto(s)
Enfermedades Hereditarias del Ojo/etiología , Genes Recesivos/genética , Enfermedades Genéticas Ligadas al Cromosoma X/etiología , Proteínas de Unión al GTP Heterotriméricas/genética , Mutación/genética , Miopía/etiología , Ceguera Nocturna/etiología , Alelos , Secuencia de Aminoácidos , Animales , Estudios de Casos y Controles , Electrorretinografía , Enfermedades Hereditarias del Ojo/patología , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Genotipo , Proteínas de Unión al GTP Heterotriméricas/química , Homocigoto , Humanos , Masculino , Ratones , Persona de Mediana Edad , Miopía/patología , Ceguera Nocturna/patología , Linaje , Fenotipo , Conformación Proteica , Homología de Secuencia de Aminoácido , Agudeza Visual/genética
8.
Am J Hum Genet ; 99(2): 470-80, 2016 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-27486781

RESUMEN

Inherited retinal dystrophies (iRDs) are a group of genetically and clinically heterogeneous conditions resulting from mutations in over 250 genes. Here, homozygosity mapping and whole-exome sequencing (WES) in a consanguineous family revealed a homozygous missense mutation, c.973C>T (p.His325Tyr), in RCBTB1. In affected individuals, it was found to segregate with retinitis pigmentosa (RP), goiter, primary ovarian insufficiency, and mild intellectual disability. Subsequent analysis of WES data in different cohorts uncovered four additional homozygous missense mutations in five unrelated families in whom iRD segregates with or without syndromic features. Ocular phenotypes ranged from typical RP starting in the second decade to chorioretinal dystrophy with a later age of onset. The five missense mutations affect highly conserved residues either in the sixth repeat of the RCC1 domain or in the BTB1 domain. A founder haplotype was identified for mutation c.919G>A (p.Val307Met), occurring in two families of Mediterranean origin. We showed ubiquitous mRNA expression of RCBTB1 and demonstrated predominant RCBTB1 localization in human inner retina. RCBTB1 was very recently shown to be involved in ubiquitination, more specifically as a CUL3 substrate adaptor. Therefore, the effect on different components of the CUL3 and NFE2L2 (NRF2) pathway was assessed in affected individuals' lymphocytes, revealing decreased mRNA expression of NFE2L2 and several NFE2L2 target genes. In conclusion, our study puts forward mutations in RCBTB1 as a cause of autosomal-recessive non-syndromic and syndromic iRD. Finally, our data support a role for impaired ubiquitination in the pathogenetic mechanism of RCBTB1 mutations.


Asunto(s)
Alelos , Factores de Intercambio de Guanina Nucleótido/genética , Mutación Missense/genética , Distrofias Retinianas/genética , Ubiquitinación/genética , Adolescente , Adulto , Edad de Inicio , Niño , Consanguinidad , Proteínas Cullin/metabolismo , Exoma/genética , Femenino , Efecto Fundador , Genes Recesivos , Haplotipos/genética , Homocigoto , Humanos , Linfocitos/metabolismo , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Linaje , Fenotipo , ARN Mensajero/genética , Retina/metabolismo , Síndrome , Turquía
9.
Ann Surg ; 269(5): 827-835, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30252681

RESUMEN

OBJECTIVE: To compare, in a phase 3, prospective, randomized, multi-center clinical trial functional outcome of reconstruction procedures following total mesorectal excision (TME). SUMMARY BACKGROUND DATA: Intestinal continuity reconstruction following TME is accompanied by postoperative defecation dysfunctions known as "anterior resection syndrome." Commonly used reconstruction techniques are straight colorectal anastomosis (SCA), colon J -pouch (CJP), and side-to-end anastomosis (SEA). Comparison of their functional outcomes in prospective, randomized, multi-center studies, including long-term assessments, is lacking. METHODS: Patients requiring TME for histologically proven rectal tumor, with or without neoadjuvant treatment, age ≥ 18 years, normal sphincter function without history of incontinence, any pretreatment staging or adenoma, expected R0-resection, were randomized for standardized SCA, CJP, or SEA procedures. Primary endpoint was comparison of composite evacuation scores 12 months after TME. Comparison of composite evacuation and incontinence scores at 6, 18 and 24 months after surgery, morbidity, and overall survival represented secondary endpoints. Analysis was based on "per protocol" (PP) population, fully complying with trial requirements, and intention-to treat (ITT) population. RESULTS: Three hundred thirty-six patients from 15 hospitals were randomized. PP population included 257 patients (JCP = 63; SEA = 95; SCA = 99). Composite evacuation scores of PP and ITT populations did not show statistically significant differences among the 3 groups at any time point. Similarly, composite incontinence scores for PP and ITT populations showed no statistically significant difference among the 3 trial arms at any time point. CONCLUSIONS: Within boundaries of investigated procedures, surgeons in charge may continue to perform reconstruction of intestinal continuity following TME at their technical preference.


Asunto(s)
Colon/cirugía , Reservorios Cólicos , Neoplasias del Recto/cirugía , Recto/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Anastomosis Quirúrgica/métodos , Humanos , Persona de Mediana Edad , Estudios Prospectivos , Suiza , Resultado del Tratamiento
10.
Clin Chem ; 65(2): 302-312, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30518662

RESUMEN

BACKGROUND: The early diagnosis of urgent abdominal pain (UAP) is challenging. Most causes of UAP are associated with extensive inflammation. Therefore, we hypothesized that quantifying inflammation using interleukin-6 and/or procalcitonin would provide incremental value in the emergency diagnosis of UAP. METHODS: This was an investigator-initiated prospective, multicenter diagnostic study enrolling patients presenting to the emergency department (ED) with acute abdominal pain. Clinical judgment of the treating physician regarding the presence of UAP was quantified using a visual analog scale after initial clinical and physician-directed laboratory assessment, and again after imaging. Two independent specialists adjudicated the final diagnosis and the classification as UAP (life-threatening, needing urgent surgery and/or hospitalization for acute medical reasons) using all information including histology and follow-up. Interleukin-6 and procalcitonin were measured blinded in a central laboratory. RESULTS: UAP was adjudicated in 376 of 1038 (36%) patients. Diagnostic accuracy for UAP was higher for interleukin-6 [area under the ROC curve (AUC), 0.80; 95% CI, 0.77-0.82] vs procalcitonin (AUC, 0.65; 95% CI, 0.62-0.68) and clinical judgment (AUC, 0.69; 95% CI, 0.65-0.72; both P < 0.001). Combined assessment of interleukin-6 and clinical judgment increased the AUC at presentation to 0.83 (95% CI, 0.80-0.85) and after imaging to 0.87 (95% CI, 0.84-0.89) and improved the correct identification of patients with and without UAP (net improvement in mean predicted probability: presentation, +19%; after imaging, +15%; P < 0.001). Decision curve analysis documented incremental value across the full range of pretest probabilities. A clinical judgment/interleukin-6 algorithm ruled out UAP with a sensitivity of 97% and ruled in UAP with a specificity of 93%. CONCLUSIONS: Interleukin-6 significantly improves the early diagnosis of UAP in the ED.


Asunto(s)
Dolor Abdominal/diagnóstico , Biomarcadores/sangre , Abdomen/diagnóstico por imagen , Adulto , Anciano , Algoritmos , Área Bajo la Curva , Servicio de Urgencia en Hospital , Femenino , Humanos , Interleucina-6/sangre , Juicio , Masculino , Persona de Mediana Edad , Polipéptido alfa Relacionado con Calcitonina/sangre , Estudios Prospectivos , Curva ROC , Tomografía Computarizada por Rayos X
11.
Ann Surg Oncol ; 26(11): 3568-3576, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31228136

RESUMEN

BACKGROUND: Functional outcomes of different reconstruction techniques have an impact on patients' quality of life (QoL), but information on long-term QoL is lacking. We compared QoL among three reconstruction techniques after total mesorectal excision (TME). METHODS: Quality of life was assessed within a randomized, multicenter trial comparing rectal surgery using side-to-end anastomosis (SEA), colon J-pouch (CJP), and straight colorectal anastomosis (SCA) by the Functional Assessment of Cancer Therapy-Colorectal scale (FACT-C) before randomization and every 6 months up to 2 years post-TME. The primary QoL endpoint was the change in the Trial Outcome Index (TOI), including the FACT-C subscales of physical and functional well-being and colorectal cancer symptoms (CSS), from baseline to month 12. Pair-wise comparisons of changes from baseline (presurgery) to each timepoint between the three arms were analyzed by Mann-Whitney tests. RESULTS: For the QoL analysis, 257 of 336 randomized patients were in the per protocol evaluation (SEA = 95; CJP = 63; SCA = 99). Significant differences between the reconstruction techniques were found for selected QoL scales up to 12 months, all in favor of CJP. Patients with SEA or SCA reported a clinically relevant deterioration for TOI and CSS at 6 months, those with SCA for CSS also at 12 months after TME. Patients with CJP remained stable. CONCLUSIONS: Although the three reconstruction techniques differ in their effects on QoL at months 6 and 12, these differences did not persist over the whole observation period of 24 months. Patients with a colon J-pouch may benefit with respect to QoL in the short-term.


Asunto(s)
Anastomosis Quirúrgica/métodos , Reservorios Cólicos/estadística & datos numéricos , Neoplasias Colorrectales/cirugía , Procedimientos de Cirugía Plástica/estadística & datos numéricos , Calidad de Vida , Recto/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/patología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Recto/patología
12.
Hum Mol Genet ; 25(12): 2539-2551, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27260406

RESUMEN

OPA1 mutations are responsible for autosomal dominant optic atrophy (ADOA), a progressive blinding disease characterized by retinal ganglion cell (RGC) degeneration and large phenotypic variations, the underlying mechanisms of which are poorly understood. OPA1 encodes a mitochondrial protein with essential biological functions, its main roles residing in the control of mitochondrial membrane dynamics as a pro-fusion protein and prevention of apoptosis. Considering recent findings showing the importance of the mitochondrial fusion process and the involvement of OPA1 in controlling steroidogenesis, we tested the hypothesis of deregulated steroid production in retina due to a disease-causing OPA1 mutation and its contribution to the visual phenotypic variations. Using the mouse model carrying the human recurrent OPA1 mutation, we disclosed that Opa1 haploinsufficiency leads to very high circulating levels of steroid precursor pregnenolone in females, causing an early-onset vision loss, abolished by ovariectomy. In addition, steroid production in retina is also increased which, in conjunction with high circulating levels, impairs estrogen receptor expression and mitochondrial respiratory complex IV activity, promoting RGC apoptosis in females. We further demonstrate the involvement of Muller glial cells as increased pregnenolone production in female cells is noxious and compromises their role in supporting RGC survival. In parallel, we analyzed ophthalmological data of a multicentre OPA1 patient cohort and found that women undergo more severe visual loss at adolescence and greater progressive thinning of the retinal nerve fibres than males. Thus, we disclosed a gender-dependent effect on ADOA severity, involving for the first time steroids and Müller glial cells, responsible for RGC degeneration.


Asunto(s)
GTP Fosfohidrolasas/genética , Atrofia Óptica Autosómica Dominante/genética , Degeneración Retiniana/genética , Células Ganglionares de la Retina/patología , Adolescente , Animales , Apoptosis/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/genética , Mitocondrias/patología , Proteínas Mutantes/genética , Nervio Óptico/patología , Pregnenolona/genética , Pregnenolona/metabolismo , Retina/patología , Degeneración Retiniana/patología , Caracteres Sexuales
13.
Hum Mol Genet ; 25(5): 916-26, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26744326

RESUMEN

Inherited retinal dystrophies are clinically and genetically heterogeneous with significant number of cases remaining genetically unresolved. We studied a large family from the West Indies islands with a peculiar retinal disease, the Martinique crinkled retinal pigment epitheliopathy that begins around the age of 30 with retinal pigment epithelium (RPE) and Bruch's membrane changes resembling a dry desert land and ends with a retinitis pigmentosa. Whole-exome sequencing identified a heterozygous c.518T>C (p.Leu173Pro) mutation in MAPKAPK3 that segregates with the disease in 14 affected and 28 unaffected siblings from three generations. This unknown variant is predicted to be damaging by bioinformatic predictive tools and the mutated protein to be non-functional by crystal structure analysis. MAPKAPK3 is a serine/threonine protein kinase of the p38 signaling pathway that is activated by a variety of stress stimuli and is implicated in cellular responses and gene regulation. In contrast to other tissues, MAPKAPK3 is highly expressed in the RPE, suggesting a crucial role for retinal physiology. Expression of the mutated allele in HEK cells revealed a mislocalization of the protein in the cytoplasm, leading to cytoskeleton alteration and cytodieresis inhibition. In Mapkapk3-/- mice, Bruch's membrane is irregular with both abnormal thickened and thinned portions. In conclusion, we identified the first pathogenic mutation in MAPKAPK3 associated with a retinal disease. These findings shed new lights on Bruch's membrane/RPE pathophysiology and will open studies of this signaling pathway in diseases with RPE and Bruch's membrane alterations, such as age-related macular degeneration.


Asunto(s)
Lámina Basal de la Coroides/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación , Proteínas Serina-Treonina Quinasas/genética , Distrofias Retinianas/genética , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal/genética , Adulto , Edad de Inicio , Anciano de 80 o más Años , Secuencia de Aminoácidos , Animales , Lámina Basal de la Coroides/patología , Exoma , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Linaje , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Secundaria de Proteína , Distrofias Retinianas/metabolismo , Distrofias Retinianas/patología , Epitelio Pigmentado de la Retina/patología , Alineación de Secuencia , Hermanos
15.
Hum Genet ; 137(2): 111-127, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29305691

RESUMEN

Cerebellar ataxia, areflexia, pes cavus, optic atrophy and sensorineural hearing impairment (CAPOS) is a rare clinically distinct syndrome caused by a single dominant missense mutation, c.2452G>A, p.Glu818Lys, in ATP1A3, encoding the neuron-specific alpha subunit of the Na+/K+-ATPase α3. Allelic mutations cause the neurological diseases rapid dystonia Parkinsonism and alternating hemiplegia of childhood, disorders which do not encompass hearing or visual impairment. We present detailed clinical phenotypic information in 18 genetically confirmed patients from 11 families (10 previously unreported) from Denmark, Sweden, UK and Germany indicating a specific type of hearing impairment-auditory neuropathy (AN). All patients were clinically suspected of CAPOS and had hearing problems. In this retrospective analysis of audiological data, we show for the first time that cochlear outer hair cell activity was preserved as shown by the presence of otoacoustic emissions and cochlear microphonic potentials, but the auditory brainstem responses were grossly abnormal, likely reflecting neural dyssynchrony. Poor speech perception was observed, especially in noise, which was beyond the hearing level obtained in the pure tone audiograms in several of the patients presented here. Molecular modelling and in vitro electrophysiological studies of the specific CAPOS mutation were performed. Heterologous expression studies of α3 with the p.Glu818Lys mutation affects sodium binding to, and release from, the sodium-specific site in the pump, the third ion-binding site. Molecular dynamics simulations confirm that the structure of the C-terminal region is affected. In conclusion, we demonstrate for the first time evidence for auditory neuropathy in CAPOS syndrome, which may reflect impaired propagation of electrical impulses along the spiral ganglion neurons. This has implications for diagnosis and patient management. Auditory neuropathy is difficult to treat with conventional hearing aids, but preliminary improvement in speech perception in some patients suggests that cochlear implantation may be effective in CAPOS patients.


Asunto(s)
Ataxia Cerebelosa/genética , Deformidades Congénitas del Pie/genética , Pérdida Auditiva Central/genética , Pérdida Auditiva Sensorineural/genética , Atrofia Óptica/genética , Reflejo Anormal/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Adolescente , Adulto , Ataxia Cerebelosa/epidemiología , Ataxia Cerebelosa/fisiopatología , Niño , Preescolar , Dinamarca/epidemiología , Femenino , Deformidades Congénitas del Pie/epidemiología , Deformidades Congénitas del Pie/fisiopatología , Alemania/epidemiología , Pérdida Auditiva Central/epidemiología , Pérdida Auditiva Central/fisiopatología , Pérdida Auditiva Sensorineural/epidemiología , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Masculino , Simulación de Dinámica Molecular , Mutación Missense/genética , Atrofia Óptica/epidemiología , Atrofia Óptica/fisiopatología , Fenotipo , Estudios Retrospectivos , ATPasa Intercambiadora de Sodio-Potasio/química , Suecia/epidemiología , Adulto Joven
16.
Am J Hum Genet ; 97(5): 754-60, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26593267

RESUMEN

Autosomal-recessive optic neuropathies are rare blinding conditions related to retinal ganglion cell (RGC) and optic-nerve degeneration, for which only mutations in TMEM126A and ACO2 are known. In four families with early-onset recessive optic neuropathy, we identified mutations in RTN4IP1, which encodes a mitochondrial ubiquinol oxydo-reductase. RTN4IP1 is a partner of RTN4 (also known as NOGO), and its ortholog Rad8 in C. elegans is involved in UV light response. Analysis of fibroblasts from affected individuals with a RTN4IP1 mutation showed loss of the altered protein, a deficit of mitochondrial respiratory complex I and IV activities, and increased susceptibility to UV light. Silencing of RTN4IP1 altered the number and morphogenesis of mouse RGC dendrites in vitro and the eye size, neuro-retinal development, and swimming behavior in zebrafish in vivo. Altogether, these data point to a pathophysiological mechanism responsible for RGC early degeneration and optic neuropathy and linking RTN4IP1 functions to mitochondrial physiology, response to UV light, and dendrite growth during eye maturation.


Asunto(s)
Proteínas Portadoras/genética , Fibroblastos/patología , Mitocondrias/patología , Proteínas Mitocondriales/genética , Mutación/genética , Enfermedades del Nervio Óptico/genética , Enfermedades del Nervio Óptico/patología , Células Ganglionares de la Retina/patología , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Complejo I de Transporte de Electrón , Femenino , Fibroblastos/metabolismo , Estudios de Seguimiento , Genes Recesivos , Humanos , Masculino , Ratones , Mitocondrias/genética , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Degeneración Nerviosa , Linaje , Pronóstico , Células Ganglionares de la Retina/metabolismo , Homología de Secuencia de Aminoácido , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
17.
Brain ; 140(10): 2586-2596, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28969390

RESUMEN

Dominant optic atrophy is a blinding disease due to the degeneration of the retinal ganglion cells, the axons of which form the optic nerves. In most cases, the disease is caused by mutations in OPA1, a gene encoding a mitochondrial large GTPase involved in cristae structure and mitochondrial network fusion. Using exome sequencing, we identified dominant mutations in DNM1L on chromosome 12p11.21 in three large families with isolated optic atrophy, including the two families that defined the OPA5 locus on chromosome 19q12.1-13.1, the existence of which is denied by the present study. Analyses of patient fibroblasts revealed physiological abundance and homo-polymerization of DNM1L, forming aggregates in the cytoplasm and on highly tubulated mitochondrial network, whereas neither structural difference of the peroxisome network, nor alteration of the respiratory machinery was noticed. Fluorescence microscopy of wild-type mouse retina disclosed a strong DNM1L expression in the ganglion cell layer and axons, and comparison between 3-month-old wild-type and Dnm1l+/- mice revealed increased mitochondrial length in retinal ganglion cell soma and axon, but no degeneration. Thus, our results disclose that in addition to OPA1, OPA3, MFN2, AFG3L2 and SPG7, dominant mutations in DNM1L jeopardize the integrity of the optic nerve, suggesting that alterations of the opposing forces governing mitochondrial fusion and fission, similarly affect retinal ganglion cell survival.


Asunto(s)
GTP Fosfohidrolasas/genética , Proteínas Asociadas a Microtúbulos/genética , Dinámicas Mitocondriales/genética , Proteínas Mitocondriales/genética , Mutación/genética , Atrofia Óptica/genética , Adolescente , Adulto , Animales , Células Cultivadas , Niño , Dinaminas , Salud de la Familia , Femenino , Fibroblastos/patología , Fibroblastos/ultraestructura , Humanos , Masculino , Ratones , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Consumo de Oxígeno/genética , Peroxisomas/patología , Retina/patología , Retina/ultraestructura
18.
Hum Mol Genet ; 24(14): 3948-55, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25901006

RESUMEN

Mitochondrial complex I (CI) deficiencies are causing debilitating neurological diseases, among which, the Leber Hereditary Optic Neuropathy and Leigh Syndrome are the most frequent. Here, we describe the first germinal pathogenic mutation in the NDUFA13/GRIM19 gene encoding a CI subunit, in two sisters with early onset hypotonia, dyskinesia and sensorial deficiencies, including a severe optic neuropathy. Biochemical analysis revealed a drastic decrease in CI enzymatic activity in patient muscle biopsies, and reduction of CI-driven respiration in fibroblasts, while the activities of complex II, III and IV were hardly affected. Western blots disclosed that the abundances of NDUFA13 protein, CI holoenzyme and super complexes were drastically reduced in mitochondrial fractions, a situation that was reproduced by silencing NDUFA13 in control cells. Thus, we established here a correlation between the first mutation yet identified in the NDUFA13 gene, which induces CI instability and a severe but slowly evolving clinical presentation affecting the central nervous system.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Discinesias/genética , Complejo I de Transporte de Electrón/deficiencia , Enfermedades Mitocondriales/genética , Hipotonía Muscular/genética , NADH NADPH Oxidorreductasas/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Niño , Preescolar , Complejo I de Transporte de Electrón/genética , Femenino , Estudios de Seguimiento , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Mutación , NADH NADPH Oxidorreductasas/metabolismo , Sistemas de Lectura Abierta , Linaje
19.
Mol Vis ; 23: 198-209, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28442884

RESUMEN

PURPOSE: Sixteen different mutations in the guanylate cyclase activator 1A gene (GUCA1A), have been previously identified to cause autosomal dominant cone dystrophy (adCOD), cone-rod dystrophy (adCORD), macular dystrophy (adMD), and in an isolated patient, retinitis pigmentosa (RP). The purpose of this study is to report on two novel mutations and the patients' clinical features. METHODS: Clinical investigations included visual acuity and visual field testing, fundus examination, high-resolution spectral-domain optical coherence tomography (OCT), fundus autofluorescence imaging, and full-field and multifocal electroretinogram (ERG) recordings. GUCA1A was screened by Sanger sequencing in a cohort of 12 French families with adCOD, adCORD, and adMD. RESULTS: We found two novel GUCA1A mutations-one amino acid deletion, c.302_304delTAG (p.Val101del), and one missense mutation, c.444T>A (p.Asp148Glu)-each of which was found in one family. The p.Asp148Glu mutation affected one of the Ca2+-binding amino acids of the EF4 hand, while the p.Val101del mutation resulted in the in-frame deletion of Valine-101, localized between two Ca2+-binding aspartic acid residues at positions 100 and 102 of the EF3 hand. Both families complained of visual acuity loss worsening with age. However, the p.Asp148Glu mutation was present in one family with adCOD involving abnormal cone function and an absence of macular atrophy, whereas p.Val101del mutation was encountered in another family with adMD without a generalized cone defect. CONCLUSIONS: The two novel mutations described in this study are associated with distinct phenotypes, MD for p.Val101del and COD for p.Asp148Glu, with no intrafamilial phenotypic heterogeneity.


Asunto(s)
Distrofias de Conos y Bastones/genética , Proteínas Activadoras de la Guanilato-Ciclasa/genética , Degeneración Macular/genética , Mutación Missense , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/genética , Eliminación de Secuencia , Adulto , Niño , Distrofias de Conos y Bastones/diagnóstico , Distrofias de Conos y Bastones/fisiopatología , Análisis Mutacional de ADN , Electrorretinografía , Femenino , Genes Dominantes , Humanos , Degeneración Macular/diagnóstico , Degeneración Macular/fisiopatología , Masculino , Persona de Mediana Edad , Imagen Óptica , Linaje , Degeneración Retiniana/diagnóstico , Degeneración Retiniana/fisiopatología , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología , Campos Visuales/fisiología , Adulto Joven
20.
Mol Vis ; 23: 131-139, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28356705

RESUMEN

PURPOSE: To report the clinical and genetic findings of one family with autosomal recessive cone dystrophy (CD) and to identify the causative mutation. METHODS: An institutional study of three family members from two generations. The clinical examination included best-corrected Snellen visual acuity measurement, fundoscopy, the Farnsworth D-15 color vision test, a full-field electroretinogram (ERG) that incorporated the International Society for Clinical Electrophysiology of Vision standards and methodology, fundus autofluorescence (FAF) and infrared (IR), and spectral-domain optical coherence tomography (SD-OCT). Genetic findings were achieved with DNA analysis using whole exome sequencing (WES) and Sanger sequencing. RESULTS: The proband, a 9-year-old boy, presented with a condition that appeared to be congenital and stationary. The clinical presentation initially reflected incomplete congenital stationary night blindness (icCSNB) because of myopia, a decrease in visual acuity, abnormal oscillatory potentials, and reduced amplitudes on the 30 Hz flicker ERG but was atypical because there were no clear electronegative responses. However, no disease-causing mutations in the genes underlying icCSNB were identified. Following WES analysis of family members, a homozygous splice-site mutation in intron 3 of TTLL5 (c.182-3_182-1delinsAA) was found cosegregating within the phenotype in the family. CONCLUSIONS: The distinction between icCSNB and CD phenotypes is not always straightforward in young patients. The patient was quite young, which most likely explains why the progression of the CD was not obvious. WES analysis provided prompt diagnosis for this family; thus, the use of this technique to refine the diagnosis is highlighted in this study.


Asunto(s)
Proteínas Portadoras/genética , Consanguinidad , Mutación/genética , Sitios de Empalme de ARN/genética , Distrofias Retinianas/genética , Niño , Segregación Cromosómica/genética , Familia , Femenino , Homocigoto , Humanos , Masculino , Linaje , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA