Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Pediatr Nephrol ; 39(8): 2351-2353, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38467926

RESUMO

Renal coloboma syndrome (RCS) and dominant optic atrophy are mainly caused by heterozygous mutations in PAX2 and OPA1, respectively. We describe a patient with digenic mutations in PAX2 and OPA1. A female infant was born without perinatal abnormalities. Magnetic resonance imaging at 4 months of age showed bilateral microphthalmia and optic nerve hypoplasia. Appropriate body size was present at 2 years of age, and mental development was favorable. Color fundus photography revealed severe retinal atrophy in both eyes. Electroretinography showed slight responses in the right eye, but no responses in the left eye, suggesting a high risk of blindness. Urinalysis results were normal, creatinine-based estimated glomerular filtration rate was 63.5 mL/min/1.73 m2, and ultrasonography showed bilateral hypoplastic kidneys. Whole exome sequencing revealed de novo frameshift mutations in PAX2 and OPA1. Both variants were classified as pathogenic (PVS1, PS2, PM2) based on the guidelines from the American College of Medical Genetics and Genomics (ACMG). Genetic testing for ocular diseases should be considered for patients with suspected RCS and a high risk of total blindness.


Assuntos
Coloboma , GTP Fosfo-Hidrolases , Fator de Transcrição PAX2 , Refluxo Vesicoureteral , Humanos , Feminino , Fator de Transcrição PAX2/genética , GTP Fosfo-Hidrolases/genética , Coloboma/genética , Coloboma/diagnóstico , Refluxo Vesicoureteral/genética , Refluxo Vesicoureteral/diagnóstico , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/diagnóstico , Anormalidades Urogenitais/genética , Anormalidades Urogenitais/diagnóstico , Anormalidades Urogenitais/complicações , Mutação da Fase de Leitura , Sequenciamento do Exoma , Lactente , Pré-Escolar , Mutação , Insuficiência Renal
2.
Genes (Basel) ; 15(2)2024 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-38397177

RESUMO

Inherited optic neuropathies affect around 1 in 10,000 people in England; in these conditions, vision is lost as retinal ganglion cells lose function or die (usually due to pathological variants in genes concerned with mitochondrial function). Emerging gene therapies for these conditions have emphasised the importance of early and expedient molecular diagnoses, particularly in the paediatric population. Here, we report our real-world clinical experience of such a population, exploring which children presented with the condition, how they were investigated and the time taken for a molecular diagnosis to be reached. A retrospective case-note review of paediatric inherited optic neuropathy patients (0-16 years) in the tertiary neuro-ophthalmology service at Moorfields Eye Hospital between 2016 and 2020 identified 19 patients. Their mean age was 9.3 ± 4.6 (mean ± SD) years at presentation; 68% were male, and 32% were female; and 26% had comorbidities, with diversity of ethnicity. Most patients had undergone genetic testing (95% (n = 18)), of whom 43% (n = 8) received a molecular diagnosis. On average, this took 54.8 ± 19.5 weeks from presentation. A cerebral MRI was performed in 70% (n = 14) and blood testing in 75% (n = 15) of patients as part of their workup. Continual improvement in the investigative pathways for inherited optic neuropathies will be paramount as novel therapeutics become available.


Assuntos
Oftalmologia , Atrofia Óptica Autossômica Dominante , Atrofia Óptica Hereditária de Leber , Doenças do Nervo Óptico , Humanos , Masculino , Feminino , Criança , Pré-Escolar , Adolescente , Atrofia Óptica Hereditária de Leber/genética , Atrofia Óptica Autossômica Dominante/genética , Estudos Retrospectivos , Doenças do Nervo Óptico/diagnóstico , Doenças do Nervo Óptico/genética , Doenças do Nervo Óptico/terapia
3.
Am J Ophthalmol ; 262: 114-124, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38278202

RESUMO

PURPOSE: Heterozygous mutations in the AFG3L2 gene (encoding a mitochondrial protease indirectly reflecting on OPA1 cleavage) and ACO2 gene (encoding the mitochondrial enzyme aconitase) are associated with isolated forms of Dominant Optic Atrophy (DOA). We aimed at describing their neuro-ophthalmological phenotype as compared with classic OPA1-related DOA. DESIGN: Cross-sectional study. METHODS: The following neuro-ophthalmological parameters were collected: logMAR visual acuity (VA), color vision, mean deviation and foveal threshold at visual fields, average and sectorial retinal nerve fiber layer (RNFL), and ganglion cell layer (GCL) thickness on optical coherence tomography. ACO2 and AFG3L2 patients were compared with an age- and sex-matched group of OPA1 patients with a 1:2 ratio. All eyes were analyzed using a clustered Wilcoxon rank sum test with the Rosner-Glynn-Lee method. RESULTS: A total of 44 eyes from 23 ACO2 patients and 26 eyes from 13 AFG3L2 patients were compared with 143 eyes from 72 OPA1 patients. All cases presented with bilateral temporal-predominant optic atrophy with various degree of visual impairment. Comparison between AFG3L2 and OPA1 failed to reveal any significant difference. ACO2 patients compared to both AFG3L2 and OPA1 presented overall higher values of nasal RNFL thickness (P = .029, P = .023), average thickness (P = .012, P = .0007), and sectorial GCL thickness. These results were confirmed also comparing separately affected and subclinical patients. CONCLUSIONS: Clinically, DOA remains a fairly homogeneous entity despite the growing genetic heterogeneity. ACO2 seems to be associated with an overall better preservation of retinal ganglion cells, probably depending on the different pathogenic mechanism involving mtDNA maintenance, as opposed to AFG3L2, which is involved in OPA1 processing and is virtually indistinguishable from classic OPA1-DOA.


Assuntos
GTP Fosfo-Hidrolases , Atrofia Óptica Autossômica Dominante , Células Ganglionares da Retina , Tomografia de Coerência Óptica , Acuidade Visual , Campos Visuais , Humanos , GTP Fosfo-Hidrolases/genética , Masculino , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/fisiopatologia , Atrofia Óptica Autossômica Dominante/diagnóstico , Feminino , Estudos Transversais , Acuidade Visual/fisiologia , Pessoa de Meia-Idade , Adulto , Células Ganglionares da Retina/patologia , Campos Visuais/fisiologia , Fenótipo , Fibras Nervosas/patologia , Estudos de Associação Genética , Adulto Jovem , Idoso , Proteínas Mitocondriais/genética , Proteases Dependentes de ATP/genética , Proteases Dependentes de ATP/metabolismo , Mutação , Adolescente , ATPases Associadas a Diversas Atividades Celulares/genética , Aconitato Hidratase
4.
J Am Assoc Nurse Pract ; 35(1): 2-4, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36602472

RESUMO

ABSTRACT: The multiple functions of mitochondria, including adenosine triphosphate synthesis, are controlled by the coordination of both the mitochondrial DNA (mtDNA) and the nuclear DNA (nDNA) genomes. Mitochondrial disorders manifest because of impairment of energy metabolism. This article focuses on mutations in two nuclear genes and their effect on mitochondrial function. Mutations in the polymerase gamma, or POLG, gene are associated with multisystemic disease processes, including Alpers Syndrome, a severe childhood-onset syndrome. Mutations in the OPA1 gene are associated with autosomal dominant optic atrophy and other neurologic, musculoskeletal, and ophthalmologic symptoms. When assessing for disorders affecting energy metabolism, sequencing of both the mtDNA genome and the nDNA whole exome sequencing is necessary.


Assuntos
Esclerose Cerebral Difusa de Schilder , Doenças Mitocondriais , Atrofia Óptica Autossômica Dominante , Humanos , DNA Mitocondrial/genética , Mitocôndrias/genética , Doenças Mitocondriais/genética , Mutação/genética , Atrofia Óptica Autossômica Dominante/genética , Esclerose Cerebral Difusa de Schilder/genética
5.
Brain ; 146(2): 455-460, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36317462

RESUMO

Hereditary optic neuropathies are caused by the degeneration of retinal ganglion cells whose axons form the optic nerves, with a consistent genetic heterogeneity. As part of our diagnostic activity, we retrospectively evaluated the combination of Leber hereditary optic neuropathy mutations testing with the exon sequencing of 87 nuclear genes on 2186 patients referred for suspected hereditary optic neuropathies. The positive diagnosis rate in individuals referred for Leber hereditary optic neuropathy testing was 18% (199/1126 index cases), with 92% (184/199) carrying one of the three main pathogenic variants of mitochondrial DNA (m.11778G>A, 66.5%; m.3460G>A, 15% and m.14484T>C, 11%). The positive diagnosis rate in individuals referred for autosomal dominant or recessive optic neuropathies was 27% (451/1680 index cases), with 10 genes accounting together for 96% of this cohort. This represents an overall positive diagnostic rate of 30%. The identified top 10 nuclear genes included OPA1, WFS1, ACO2, SPG7, MFN2, AFG3L2, RTN4IP1, TMEM126A, NR2F1 and FDXR. Eleven additional genes, each accounting for less than 1% of cases, were identified in 17 individuals. Our results show that 10 major genes account for more than 96% of the cases diagnosed with our nuclear gene panel.


Assuntos
Atrofia Óptica Autossômica Dominante , Atrofia Óptica Hereditária de Leber , Doenças do Nervo Óptico , Humanos , Atrofia Óptica Hereditária de Leber/genética , Estudos Retrospectivos , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/patologia , Doenças do Nervo Óptico/genética , Mutação/genética , DNA Mitocondrial/genética , ATPases Associadas a Diversas Atividades Celulares/genética , Proteases Dependentes de ATP/genética , Proteínas de Transporte/genética , Proteínas Mitocondriais/genética , Proteínas de Membrana/genética
6.
Eur J Hum Genet ; 30(7): 848-855, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35534703

RESUMO

Autosomal dominant optic atrophy (DOA) is an inherited optic neuropathy that results in progressive, bilateral visual acuity loss and field defects. OPA1 is the causative gene in around 60% of cases of DOA. The majority of patients have a pure ocular phenotype, but 20% have extra-ocular features (DOA +). We report on a patient with DOA + manifesting as bilateral optic atrophy, spastic paraparesis, urinary incontinence and white matter changes in the central nervous system associated with a novel heterozygous splice variant NM_015560.2(OPA1):c.2356-1 G > T. Further characterisation, which was performed using fibroblasts obtained from a skin biopsy, demonstrated that this variant altered mRNA splicing of the OPA1 transcript, specifically a 21 base pair deletion at the start of exon 24, NM_015560.2(OPA1):p.Cys786_Lys792del. The majority of variant transcripts were shown to escape nonsense-mediated decay and modelling of the predicted protein structure suggests that the in-frame 7 amino acid deletion may affect OPA1 oligomerisation. Fibroblasts carrying the c.2356-1 G > T variant demonstrated impaired mitochondrial bioenergetics, membrane potential, increased cell death, and disrupted and fragmented mitochondrial networks in comparison to WT cells. This study suggests that the c.2356-1 G > T OPA1 splice site variant leads to a cryptic splice site activation and may manifest in a dominant-negative manner, which could account for the patient's severe syndromic phenotype.


Assuntos
Atrofia Óptica Autossômica Dominante , Sítios de Splice de RNA , GTP Fosfo-Hidrolases/genética , Humanos , Mitocôndrias/genética , Mitocôndrias/patologia , Mutação , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/patologia
7.
Acta Neuropathol Commun ; 8(1): 93, 2020 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-32600459

RESUMO

Autosomal dominant optic atrophy (ADOA) is a neuro-ophthalmic condition characterized by bilateral degeneration of the optic nerves. Although heterozygous mutations in OPA1 represent the most common genetic cause of ADOA, a significant number of cases remain undiagnosed.Here, we describe a family with a strong ADOA history with most family members spanning three generation having childhood onset of visual symptoms. The proband, in addition to optic atrophy, had neurological symptoms consistent with relapsing remitting multiple sclerosis. Clinical exome analysis detected a novel mutation in the AFG3L2 gene (NM_006796.2:c.1010G > A; p.G337E), which segregated with optic atrophy in family members. AFG3L2 is a metalloprotease of the AAA subfamily which exerts quality control in the inner mitochondrial membrane. Interestingly, the identified mutation localizes close to the AAA domain of AFG3L2, while those localized in the proteolytic domain cause dominant spinocerebellar ataxia type 28 (SCA28) or recessive spastic ataxia with epilepsy (SPAX5). Functional studies in patient fibroblasts demonstrate that the p.G337E AFG3L2 mutation strongly destabilizes the long isoforms of OPA1 via OMA hyper-activation and leads to mitochondrial fragmentation, thus explaining the family phenotype. This study widens the clinical spectrum of neurodegenerative diseases caused by AFG3L2 mutations, which shall be considered as genetic cause of ADOA.


Assuntos
Proteases Dependentes de ATP/genética , ATPases Associadas a Diversas Atividades Celulares/genética , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/metabolismo , Domínio AAA/genética , Adolescente , Criança , Pré-Escolar , Feminino , GTP Fosfo-Hidrolases/metabolismo , Humanos , Masculino , Metaloendopeptidases/metabolismo , Mutação de Sentido Incorreto , Linhagem
8.
Theranostics ; 10(16): 7260-7272, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32641991

RESUMO

Rationale: Mitochondrial disorders preferentially affect tissues with high energy requirements, such as the retina and corneal endothelium, in human eyes. Mesenchymal stem cell (MSC)-based treatment has been demonstrated to be beneficial for ocular degeneration. However, aside from neuroprotective paracrine actions, the mechanisms underlying the beneficial effect of MSCs on retinal and corneal tissues are largely unknown. In this study, we investigated the fate and associated characteristics of mitochondria subjected to intercellular transfer from MSCs to ocular cells. Methods: MSCs were cocultured with corneal endothelial cells (CECs), 661W cells (a photoreceptor cell line) and ARPE-19 cells (a retinal pigment epithelium cell line). Immunofluorescence, fluorescence activated cell sorting and confocal microscopy imaging were employed to investigate the traits of intercellular mitochondrial transfer and the fate of transferred mitochondria. The oxygen consumption rate of recipient cells was measured to investigate the effect of intercellular mitochondrial transfer. Transcriptome analysis was performed to investigate the expression of metabolic genes in recipient cells with donated mitochondria. Results: Mitochondrial transport is a ubiquitous intercellular mechanism between MSCs and various ocular cells, including the corneal endothelium, retinal pigmented epithelium, and photoreceptors. Additionally, our results indicate that the donation process depends on F-actin-based tunneling nanotubes. Rotenone-pretreated cells that received mitochondria from MSCs displayed increased aerobic capacity and upregulation of mitochondrial genes. Furthermore, living imaging determined the ultimate fate of transferred mitochondria through either degradation by lysosomes or exocytosis as extracellular vesicles. Conclusions: For the first time, we determined the characteristics and fate of mitochondria undergoing intercellular transfer from MSCs to various ocular cells through F-actin-based tunneling nanotubes, helping to characterize MSC-based treatment for ocular tissue regeneration.


Assuntos
Comunicação Celular , Metabolismo Energético , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Mitocôndrias/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular , Movimento Celular , Técnicas de Cocultura , Córnea/citologia , Córnea/metabolismo , Córnea/patologia , DNA Mitocondrial/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Distrofia Endotelial de Fuchs/genética , Distrofia Endotelial de Fuchs/patologia , Distrofia Endotelial de Fuchs/terapia , Humanos , Injeções Intraoculares , Células-Tronco Mesenquimais/citologia , Camundongos , Mitocôndrias/genética , Modelos Animais , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/patologia , Atrofia Óptica Autossômica Dominante/terapia , Atrofia Óptica Hereditária de Leber/genética , Atrofia Óptica Hereditária de Leber/patologia , Atrofia Óptica Hereditária de Leber/terapia , Células Fotorreceptoras de Vertebrados/citologia , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patologia , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia
9.
Invest Ophthalmol Vis Sci ; 61(6): 42, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32561926

RESUMO

Purpose: Retinal ganglion cells (RGCs) are susceptible to mitochondrial deficits and also the major cell type affected in patients with mutations in the OPA1 gene in autosomal dominant optic atrophy (ADOA). Here, we characterized mitochondria in RGCs in vitro from a heterozygous B6; C3-Opa1Q285STOP (Opa1+/-) mouse model to investigate mitochondrial changes underlying the pathology in ADOA. Methods: Mouse RGCs were purified from wild-type and Opa1+/- mouse retina by two-step immunopanning. The mitochondria in neurites of RGCs were labeled with MitoTracker Red for structure and motility measurement by time-lapse imaging. Mitochondrial bioenergetics were determined by the real-time measurement of oxygen consumption rate using a Seahorse XFe 96 Extracellular Flux Analyzer. Results: We observed a significant decrease in mitochondrial length in Opa1+/- RGCs with a remarkably higher proportion and density of motile mitochondria along the neurites. We also observed an increased transport velocity with a higher number of contacts between mitochondria in Opa1+/- RGC neurites. The oxygen consumption assays showed a severe impairment in basal respiration, Adenosine triphosphate-linked (ATP-linked) oxygen consumption, as well as reserve respiratory capacity, in RGCs from Opa1+/- mouse retina. Conclusions: Opa1 deficiency leads to significant fragmentation of mitochondrial morphology, activation of mitochondrial motility and impaired respiratory function in RGCs from the B6; C3-Opa1Q285STOP mouse model. This highlights the significant alterations in the intricate interplay between mitochondrial morphology, motility, and energy production in RGCs with Opa1 deficiency long before the onset of clinical symptoms of the pathology.


Assuntos
Metabolismo Energético , GTP Fosfo-Hidrolases/deficiência , Mitocôndrias/metabolismo , Mutação , Atrofia Óptica Autossômica Dominante/genética , Células Ganglionares da Retina/metabolismo , Animais , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , GTP Fosfo-Hidrolases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Atrofia Óptica Autossômica Dominante/metabolismo , Atrofia Óptica Autossômica Dominante/patologia , Células Ganglionares da Retina/patologia
11.
Metab Brain Dis ; 34(4): 1023-1027, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31152339

RESUMO

OPA1 variants most frequently manifest phenotypically with pure autosomal dominant optic atrophy (ADOA) or with ADOA plus. The most frequent abnormalities in ADOA plus in addition to the optic nerve affection include hypoacusis, migraine, myopathy, and neuropathy. Hypertelorism and atrophy of the acoustic nerve have not been reported. The patient is a 48yo Caucasian female with slowly progressive, visual impairment since childhood, bilateral hypoacusis since age 10y, and classical migraine since age 20y. The family history was positive for diabetes (father, mother) and visual impairment (daughter). Clinical examination revealed hypertelorism, visual impairment, hypoacusis, tinnitus, weakness for elbow flexion and finger straddling, and generally reduced tendon reflexes. MRI of the cerebrum was non-informative but hypoplasia of the acoustic nerve bilaterally was described. Visually-evoked potentials revealed markedly prolonged P100-latencies bilaterally. Acoustically-evoked potentials were distorted with poor reproducibility and prolonged latencies. Muscle biopsy revealed reduced activities of complexes I, II, and IV. Genetic work-up revealed the novel variant c.1463G>C in the OPA1 gene. This case provides novel information regarding the genotype of ADOA plus. The novel OPA1 variant c.1463G>C not only manifests with visual impairment, hypoacusis, migraine, and myopathy, but also with hypertelorisms and acoustic nerve atrophy.


Assuntos
Potenciais Evocados Auditivos/fisiologia , GTP Fosfo-Hidrolases/genética , Mutação , Atrofia Óptica Autossômica Dominante/genética , Análise Mutacional de DNA , Feminino , Genótipo , Humanos , Pessoa de Meia-Idade , Linhagem , Fenótipo
12.
Sci Rep ; 8(1): 2468, 2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29410463

RESUMO

Dominant optic atrophy (DOA) is a rare progressive and irreversible blinding disease which is one of the most frequent forms of hereditary optic neuropathy. DOA is mainly caused by dominant mutation in the OPA1 gene encoding a large mitochondrial GTPase with crucial roles in membrane dynamics and cell survival. Hereditary optic neuropathies are commonly characterized by the degeneration of retinal ganglion cells, leading to the optic nerve atrophy and the progressive loss of visual acuity. Up to now, despite increasing advances in the understanding of the pathological mechanisms, DOA remains intractable. Here, we tested the efficiency of gene therapy on a genetically-modified mouse model reproducing DOA vision loss. We performed intravitreal injections of an Adeno-Associated Virus carrying the human OPA1 cDNA under the control of the cytomegalovirus promotor. Our results provide the first evidence that gene therapy is efficient on a mouse model of DOA as the wild-type OPA1 expression is able to alleviate the OPA1-induced retinal ganglion cell degeneration, the hallmark of the disease. These results displayed encouraging effects of gene therapy for Dominant Optic Atrophy, fostering future investigations aiming at clinical trials in patients.


Assuntos
GTP Fosfo-Hidrolases/genética , Terapia Genética/métodos , Mitocôndrias/genética , Atrofia Óptica Autossômica Dominante/terapia , Células Ganglionares da Retina/metabolismo , Baixa Visão/terapia , Animais , Morte Celular , Citomegalovirus/genética , Citomegalovirus/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Modelos Animais de Doenças , Feminino , GTP Fosfo-Hidrolases/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Injeções Intravítreas , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mutação , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/metabolismo , Atrofia Óptica Autossômica Dominante/patologia , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Regiões Promotoras Genéticas , Células Ganglionares da Retina/patologia , Transgenes , Baixa Visão/genética , Baixa Visão/metabolismo , Baixa Visão/patologia
13.
Invest Ophthalmol Vis Sci ; 59(1): 185-195, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29340645

RESUMO

Purpose: Dominant optic atrophy (DOA; MIM [Mendelian Inheritance in Man] 165500), resulting in retinal ganglion cell degeneration, is mainly caused by mutations in the optic atrophy 1 (OPA1) gene, which encodes a dynamin guanosine triphosphate (GTP)ase involved in mitochondrial membrane processing. This work aimed at determining whether plasma from OPA1 pathogenic variant carriers displays a specific metabolic signature. Methods: We applied a nontargeted clinical metabolomics pipeline based on ultra-high-pressure liquid chromatography coupled to high-resolution mass spectrometry (UHPLC-HRMS) allowing the exploration of 500 polar metabolites in plasma. We compared the plasma metabolic profiles of 25 patients with various OPA1 pathogenic variants and phenotypes to those of 20 healthy controls. Statistical analyses were performed using univariate and multivariate (principal component analysis [PCA], orthogonal partial least-squares discriminant analysis [OPLS-DA]) methods and a machine learning approach, the Biosigner algorithm. Results: A robust and relevant predictive model characterizing OPA1 individuals was obtained, based on a complex panel of metabolites with altered concentrations. An impairment of the purine metabolism, including significant differences in xanthine, hypoxanthine, and inosine concentrations, was at the foreground of this signature. In addition, the signature was characterized by differences in urocanate, choline, phosphocholine, glycerate, 1-oleoyl-rac-glycerol, rac-glycerol-1-myristate, aspartate, glutamate, and cystine concentrations. Conclusions: This first metabolic signature reported in the plasma of patient carrying OPA1 pathogenic variants highlights the unexpected involvement of purine metabolism in the pathophysiology of DOA.


Assuntos
GTP Fosfo-Hidrolases/genética , Atrofia Óptica Autossômica Dominante/sangue , Purinas/metabolismo , Adolescente , Adulto , Criança , Cromatografia Líquida de Alta Pressão , Feminino , Genótipo , Humanos , Masculino , Metaboloma , Metabolômica/métodos , Pessoa de Meia-Idade , Atrofia Óptica Autossômica Dominante/genética , Fenótipo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Adulto Jovem
14.
Sci Rep ; 7(1): 5704, 2017 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-28720802

RESUMO

Autosomal dominant optic atrophy (ADOA) is a dominantly inherited optic neuropathy, affecting the specific loss of retinal ganglion cells (RGCs). The majority of affected cases of ADOA are associated with mutations in OPA1 gene. Our previous investigation identified the c.1198C > G (p.P400A) mutation in the OPA1 in a large Han Chinese family with ADOA. In this report, we performed a functional characterization using lymphoblostoid cell lines derived from affected members of this family and control subjects. Mutant cell lines exhibited the aberrant mitochondrial morphology. A ~24.6% decrease in the mitochondrial DNA (mtDNA) copy number was observed in mutant cell lines, as compared with controls. Western blotting analysis revealed the variable reductions (~45.7%) in four mtDNA-encoded polypeptides in mutant cell lines. The impaired mitochondrial translation caused defects in respiratory capacity. Furthermore, defects in mitochondrial ATP synthesis and mitochondrial membrane potential (ΔΨm) were observed in mutant cell lines. These abnormalities resulted in the accumulation of oxidative damage and increasing of apoptosis in the mutant cell lines, as compared with controls. All those alterations may cause the primary degeneration of RGCs and subsequent visual loss. These data provided the direct evidence for c.1198C > G mutation leading to ADOA. Our findings may provide new insights into the understanding of pathophysiology of ADOA.


Assuntos
GTP Fosfo-Hidrolases/genética , Atrofia Óptica Autossômica Dominante/genética , Apoptose , Povo Asiático , Linhagem Celular/virologia , DNA Mitocondrial/análise , GTP Fosfo-Hidrolases/biossíntese , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias , Mutação , Atrofia Óptica Autossômica Dominante/sangue , Espécies Reativas de Oxigênio/metabolismo
15.
Mol Cell Biol ; 37(17)2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28630277

RESUMO

Mitochondria undergo frequent morphological changes to control their function. We show here that T-cell intracellular antigens (TIA1b/TIARb) and Hu antigen R (HuR) have antagonistic roles in mitochondrial function by modulating the expression of mitochondrial shaping proteins. Expression of TIA1b/TIARb alters the mitochondrial dynamic network by enhancing fission and clustering, which is accompanied by a decrease in respiration. In contrast, HuR expression promotes fusion and cristae remodeling and increases respiratory activity. Mechanistically, TIA proteins downregulate the expression of optic atrophy 1 (OPA1) protein via switching of the splicing patterns of OPA1 to facilitate the production of OPA1 variant 5 (OPA1v5). Conversely, HuR enhances the expression of OPA1 mRNA isoforms through increasing steady-state levels and targeting translational efficiency at the 3' untranslated region. Knockdown of TIA1/TIAR or HuR partially reversed the expression profile of OPA1, whereas knockdown of OPA1 or overexpression of OPA1v5 provoked mitochondrial clustering. Middle-term expression of TIA1b/TIARb triggers reactive oxygen species production and mitochondrial DNA damage, which is accompanied by mitophagy, autophagy, and apoptosis. In contrast, HuR expression promotes mitochondrion-dependent cell proliferation. Collectively, these results provide molecular insights into the antagonistic functions of TIA1b/TIARb and HuR in mitochondrial activity dynamics and suggest that their balance might contribute to mitochondrial physiopathology.


Assuntos
Proteína Semelhante a ELAV 1/metabolismo , Expressão Gênica/fisiologia , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/metabolismo , Proteínas de Ligação a Poli(A)/metabolismo , Proliferação de Células , Citoplasma/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Humanos , Mitocôndrias/genética , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , RNA Mensageiro/genética , Antígeno-1 Intracelular de Células T
16.
Hum Mol Genet ; 25(12): 2539-2551, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27260406

RESUMO

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.


Assuntos
GTP Fosfo-Hidrolases/genética , Atrofia Óptica Autossômica Dominante/genética , Degeneração Retiniana/genética , Células Ganglionares da Retina/patologia , Adolescente , Animais , Apoptose/genética , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mitocôndrias/genética , Mitocôndrias/patologia , Proteínas Mutantes/genética , Nervo Óptico/patologia , Pregnenolona/genética , Pregnenolona/metabolismo , Retina/patologia , Degeneração Retiniana/patologia , Caracteres Sexuais
17.
Pharmacol Ther ; 165: 132-52, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27288727

RESUMO

Optic neuropathies are an important cause of blindness worldwide. The study of the most common inherited mitochondrial optic neuropathies, Leber hereditary optic neuropathy (LHON) and autosomal dominant optic atrophy (ADOA) has highlighted a fundamental role for mitochondrial function in the survival of the affected neuron-the retinal ganglion cell. A picture is now emerging that links mitochondrial dysfunction to optic nerve disease and other neurodegenerative processes. Insights gained from the peculiar susceptibility of retinal ganglion cells to mitochondrial dysfunction are likely to inform therapeutic development for glaucoma and other common neurodegenerative diseases of aging. Despite it being a fast-evolving field of research, a lack of access to human ocular tissues and limited animal models of mitochondrial disease have prevented direct retinal ganglion cell experimentation and delayed the development of efficient therapeutic strategies to prevent vision loss. Currently, there are no approved treatments for mitochondrial disease, including optic neuropathies caused by primary or secondary mitochondrial dysfunction. Recent advances in eye research have provided important insights into the molecular mechanisms that mediate pathogenesis, and new therapeutic strategies including gene correction approaches are currently being investigated. Here, we review the general principles of mitochondrial biology relevant to retinal ganglion cell function and provide an overview of the major optic neuropathies with mitochondrial involvement, LHON and ADOA, whilst highlighting the emerging link between mitochondrial dysfunction and glaucoma. The pharmacological strategies currently being trialed to improve mitochondrial dysfunction in these optic neuropathies are discussed in addition to emerging therapeutic approaches to preserve retinal ganglion cell function.


Assuntos
Terapia Genética/métodos , Glaucoma/terapia , Mitocôndrias/transplante , Doenças Mitocondriais/terapia , Atrofia Óptica Autossômica Dominante/terapia , Atrofia Óptica Hereditária de Leber/terapia , Células Ganglionares da Retina/transplante , Transplante de Células-Tronco/métodos , Animais , Restrição Calórica , Metabolismo Energético , Exercício Físico , Glaucoma/genética , Glaucoma/metabolismo , Glaucoma/patologia , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Doenças Mitocondriais/genética , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Dinâmica Mitocondrial , Regeneração Nervosa , Fármacos Neuroprotetores/uso terapêutico , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica Autossômica Dominante/metabolismo , Atrofia Óptica Autossômica Dominante/patologia , Atrofia Óptica Hereditária de Leber/genética , Atrofia Óptica Hereditária de Leber/metabolismo , Atrofia Óptica Hereditária de Leber/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia
18.
Mol Med Rep ; 14(1): 33-40, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27150940

RESUMO

Mutations in the optic atrophy 1 gene (OPA1) are associated with autosomal dominant optic atrophy and 20% of patients demonstrate extra-ocular manifestations. In addition to these autosomal dominant cases, only a few syndromic cases have been reported thus far with compound heterozygous OPA1 mutations, suggestive of either recessive or semi­dominant patterns of inheritance. The majority of these patients were diagnosed with Behr syndrome, characterized by optic atrophy, ataxia and peripheral neuropathy. The present study describes a 10-year-old boy with Behr syndrome presenting with early­onset severe optic atrophy, sensorimotor neuropathy, ataxia and congenital cataracts. He had optic atrophy and was declared legally blind at six years old. Electrophysiological, radiological, and histopathological findings were compatible with axonal sensorimotor polyneuropathy. At birth, he presented with a congenital cataract, which has not been previously described in patients with OPA1 mutations. Whole exome sequencing indicated a pair of novel compound heterozygous mutations: p.L620fs*13 (c.1857­1858delinsT) and p.R905Q (c.G2714A). Neither mutation was observed in controls (n=300), and thus, they were predicted to be pathogenic by multiple in silico analyses. The mutation sites were highly conserved throughout different vertebrate species. The patients parents did not have any ophthalmic or neurologic symptoms and the results of electrophysiological studies were normal, suggestive of an autosomal recessive pattern of inheritance. The present study identified novel compound heterozygous OPA1 mutations in a patient with recessive optic atrophy, sensorimotor neuropathy and congenital cataracts, indicating an expansion of the clinical spectrum of pathologies associated with OPA1 mutations. Thus, OPA1 gene screening is advisable in the workup of patients with recessive optic atrophy, particularly with Behr syndrome and cataracts.


Assuntos
Ataxia/genética , Catarata/genética , GTP Fosfo-Hidrolases/genética , Perda Auditiva/genética , Heterozigoto , Deficiência Intelectual/genética , Mutação , Atrofia Óptica Autossômica Dominante/genética , Atrofia Óptica/congênito , Espasmo/genética , Ataxia/diagnóstico , Biópsia , Catarata/diagnóstico , Criança , Análise Mutacional de DNA , Exoma , Genes Recessivos , Perda Auditiva/diagnóstico , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Deficiência Intelectual/diagnóstico , Perna (Membro)/diagnóstico por imagem , Perna (Membro)/patologia , Imageamento por Ressonância Magnética , Masculino , Atrofia Óptica/diagnóstico , Atrofia Óptica/genética , Atrofia Óptica Autossômica Dominante/diagnóstico , Linhagem , Espasmo/diagnóstico
19.
Stem Cell Res Ther ; 7: 2, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26738566

RESUMO

BACKGROUND: Many retinal degenerative diseases are caused by the loss of retinal ganglion cells (RGCs). Autosomal dominant optic atrophy is the most common hereditary optic atrophy disease and is characterized by central vision loss and degeneration of RGCs. Currently, there is no effective treatment for this group of diseases. However, stem cell therapy holds great potential for replacing lost RGCs of patients. Compared with embryonic stem cells, induced pluripotent stem cells (iPSCs) can be derived from adult somatic cells, and they are associated with fewer ethical concerns and are less prone to immune rejection. In addition, patient-derived iPSCs may provide us with a cellular model for studying the pathogenesis and potential therapeutic agents for optic atrophy. METHODS: In this study, iPSCs were obtained from patients carrying an OPA1 mutation (OPA1 (+/-) -iPSC) that were diagnosed with optic atrophy. These iPSCs were differentiated into putative RGCs, which were subsequently characterized by using RGC-specific expression markers BRN3a and ISLET-1. RESULTS: Mutant OPA1 (+/-) -iPSCs exhibited significantly more apoptosis and were unable to efficiently differentiate into RGCs. However, with the addition of neural induction medium, Noggin, or estrogen, OPA1 (+/-) -iPSC differentiation into RGCs was promoted. CONCLUSIONS: Our results suggest that apoptosis mediated by OPA1 mutations plays an important role in the pathogenesis of optic atrophy, and both noggin and ß-estrogen may represent potential therapeutic agents for OPA1-related optic atrophy.


Assuntos
Células-Tronco Pluripotentes Induzidas/fisiologia , Atrofia Óptica Autossômica Dominante/patologia , Proteínas de Transporte/fisiologia , Diferenciação Celular , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Estradiol/fisiologia , GTP Fosfo-Hidrolases/genética , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Atrofia Óptica Autossômica Dominante/tratamento farmacológico , Atrofia Óptica Autossômica Dominante/genética , Células Ganglionares da Retina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA