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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(27): 15684-15693, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32571921

RESUMEN

Mitochondria are known to play an essential role in photoreceptor function and survival that enables normal vision. Within photoreceptors, mitochondria are elongated and extend most of the inner-segment length, where they supply energy for protein synthesis and the phototransduction machinery in the outer segment, as well as acting as a calcium store. Here, we examined the arrangement of the mitochondria within the inner segment in detail using three-dimensional (3D) electron microscopy techniques and show they are tethered to the plasma membrane in a highly specialized arrangement. Remarkably, mitochondria and their cristae openings align with those of neighboring inner segments. The pathway by which photoreceptors meet their high energy demands is not fully understood. We propose this to be a mechanism to share metabolites and assist in maintaining homeostasis across the photoreceptor cell layer. In the extracellular space between photoreceptors, Müller glial processes were identified. Due to the often close proximity to the inner-segment mitochondria, they may, too, play a role in the inner-segment mitochondrial arrangement as well as metabolite shuttling. OPA1 is an important factor in mitochondrial homeostasis, including cristae remodeling; therefore, we examined the photoreceptors of a heterozygous Opa1 knockout mouse model. The cristae structure in the Opa1+/- photoreceptors was not greatly affected, but the mitochondria were enlarged and had reduced alignment to neighboring inner-segment mitochondria. This indicates the importance of key regulators in maintaining this specialized photoreceptor mitochondrial arrangement.


Asunto(s)
GTP Fosfohidrolasas/genética , Mitocondrias/genética , Membranas Mitocondriales/ultraestructura , Visión Ocular/genética , Animales , Membrana Celular/genética , Membrana Celular/ultraestructura , Células Ependimogliales/metabolismo , Células Ependimogliales/ultraestructura , Humanos , Ratones , Microscopía Electrónica , Mitocondrias/ultraestructura , Membranas Mitocondriales/metabolismo , Células Fotorreceptoras/ultraestructura , Visión Ocular/fisiología
2.
J Inherit Metab Dis ; 45(4): 796-803, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35543492

RESUMEN

Primary mitochondrial disorders encompass a wide range of clinical presentations and a spectrum of severity. They currently lack effective disease-modifying therapies and have a high mortality and morbidity rate. It is therefore essential to know that competitively funded research designed by academics meets the core needs of people with mitochondrial disorders and their clinicians. Priority setting partnerships are an established collaborative methodology that brings patients, carers and families, charity representatives and clinicians together to try to establish the most pressing and unanswered research priorities for a particular disease. We developed a web-based questionnaire, requesting all patients affected by primary mitochondrial disease, their carers and clinicians to pose their research questions. This yielded 709 questions from 147 participants. These were grouped into overarching themes including basic biology, causation, health services, clinical management, social impacts, prognosis, prevention, symptoms, treatment and psychological impact. Following the removal of "answered questions", the process resulted in a list of 42 discrete, answerable questions. This was further refined by web-based ranking by the community to 24 questions. These were debated at a face-to-face workshop attended by a diverse range of patients, carers, charity representatives and clinicians to create a definitive "Top 10 of unanswered research questions for primary mitochondrial disorders". These Top 10 questions related to understanding biological processes, including triggers of disease onset, mechanisms underlying progression and reasons for differential symptoms between individuals with identical genetic mutations; new treatments; biomarker discovery; psychological support and optimal management of stroke-like episodes and fatigue.


Asunto(s)
Investigación Biomédica , Enfermedades Mitocondriales , Cuidadores , Prioridades en Salud , Humanos , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/terapia , Encuestas y Cuestionarios
3.
Int J Mol Sci ; 22(11)2021 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-34072974

RESUMEN

This study investigates whether reduced optic atrophy 1 (Opa1) level promotes apoptosis and retinal vascular lesions associated with diabetic retinopathy (DR). Four groups of mice: wild type (WT) control mice, streptozotocin (STZ)-induced diabetic mice, Opa1+/- mice, and diabetic Opa1+/- mice were used in this study. 16 weeks after diabetes onset, retinas were assessed for Opa1 and Bax levels by Western blot analysis, and retinal networks were examined for acellular capillaries (AC) and pericyte loss (PL). Apoptotic cells were detected in retinal capillaries using TUNEL assay, and caspase-3 activity was assessed using fluorometric analysis. Opa1 expression was significantly downregulated in retinas of diabetic and Opa1+/- mice compared with those of WT mice. Inducing diabetes further decreased Opa1 expression in retinas of Opa1+/- mice. Increased cytochrome c release concomitant with increased level of pro-apoptotic Bax and elevated caspase-3 activity were observed in retinas of diabetic and Opa1+/- mice; the number of TUNEL-positive cells and AC/PL was also significantly increased. An additional decrease in the Opa1 level in retinas of diabetic Opa1+/- mice exacerbated the development of apoptotic cells and AC/PL compared with those of diabetic mice. Diabetes-induced Opa1 downregulation contributes, at least in part, to the development of retinal vascular lesions characteristic of DR.


Asunto(s)
Capilares , Diabetes Mellitus Experimental/metabolismo , Retinopatía Diabética/metabolismo , GTP Fosfohidrolasas/fisiología , Vasos Retinianos , Animales , Apoptosis , Capilares/metabolismo , Capilares/patología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Retinopatía Diabética/etiología , Retinopatía Diabética/patología , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/patología
4.
Int J Mol Sci ; 22(16)2021 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-34445085

RESUMEN

Retinal ganglion cells (RGCs) undergo dendritic pruning in a variety of neurodegenerative diseases, including glaucoma and autosomal dominant optic atrophy (ADOA). Axotomising RGCs by severing the optic nerve generates an acute model of RGC dendropathy, which can be utilized to assess the therapeutic potential of treatments for RGC degeneration. Photobiomodulation (PBM) with red light provided neuroprotection to RGCs when administered ex vivo to wild-type retinal explants. In the current study, we used aged (13-15-month-old) wild-type and heterozygous B6;C3-Opa1Q285STOP (Opa1+/-) mice, a model of ADOA exhibiting RGC dendropathy. These mice were pre-treated with 4 J/cm2 of 670 nm light for five consecutive days before the eyes were enucleated and the retinas flat-mounted into explant cultures for 0-, 8- or 16-h ex vivo. RGCs were imaged by confocal microscopy, and their dendritic architecture was quantified by Sholl analysis. In vivo 670 nm light pretreatment inhibited the RGC dendropathy observed in untreated wild-type retinas over 16 h ex vivo and inhibited dendropathy in ON-center RGCs in wild-type but not Opa1+/- retinas. Immunohistochemistry revealed that aged Opa1+/- RGCs exhibited increased nitrosative damage alongside significantly lower activation of NF-κB and upregulation of DJ-1. PBM restored NF-κB activation in Opa1+/- RGCs and enhanced DJ-1 expression in both genotypes, indicating a potential molecular mechanism priming the retina to resist future oxidative insult. These data support the potential of PBM as a treatment for diseases involving RGC degeneration.


Asunto(s)
Atrofia Óptica Autosómica Dominante/terapia , Fototerapia , Proteína Desglicasa DJ-1/análisis , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/efectos de la radiación , Animales , Modelos Animales de Enfermedad , Luz , Ratones , Neuroprotección/efectos de la radiación , Atrofia Óptica Autosómica Dominante/patología , Degeneración Retiniana , Células Ganglionares de la Retina/citología , Regulación hacia Arriba/efectos de la radiación
5.
Ann Neurol ; 86(3): 368-383, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31298765

RESUMEN

OBJECTIVE: Autosomal dominant optic atrophy (ADOA) starts in early childhood with loss of visual acuity and color vision deficits. OPA1 mutations are responsible for the majority of cases, but in a portion of patients with a clinical diagnosis of ADOA, the cause remains unknown. This study aimed to identify novel ADOA-associated genes and explore their causality. METHODS: Linkage analysis and sequencing were performed in multigeneration families and unrelated patients to identify disease-causing variants. Functional consequences were investigated in silico and confirmed experimentally using the zebrafish model. RESULTS: We defined a new ADOA locus on 7q33-q35 and identified 3 different missense variants in SSBP1 (NM_001256510.1; c.113G>A [p.(Arg38Gln)], c.320G>A [p.(Arg107Gln)] and c.422G>A [p.(Ser141Asn)]) in affected individuals from 2 families and 2 singletons with ADOA and variable retinal degeneration. The mutated arginine residues are part of a basic patch that is essential for single-strand DNA binding. The loss of a positive charge at these positions is very likely to lower the affinity of SSBP1 for single-strand DNA. Antisense-mediated knockdown of endogenous ssbp1 messenger RNA (mRNA) in zebrafish resulted in compromised differentiation of retinal ganglion cells. A similar effect was achieved when mutated mRNAs were administered. These findings point toward an essential role of ssbp1 in retinal development and the dominant-negative nature of the identified human variants, which is consistent with the segregation pattern observed in 2 multigeneration families studied. INTERPRETATION: SSBP1 is an essential protein for mitochondrial DNA replication and maintenance. Our data have established pathogenic variants in SSBP1 as a cause of ADOA and variable retinal degeneration. ANN NEUROL 2019;86:368-383.


Asunto(s)
Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad/genética , Proteínas Mitocondriales/genética , Atrofia Óptica Autosómica Dominante/genética , Animales , Diferenciación Celular/genética , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Ligamiento Genético/genética , Humanos , Masculino , Ratones , Mutación Missense , Atrofia Óptica Autosómica Dominante/patología , Linaje , ARN Mensajero/genética , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Pez Cebra/genética
6.
J Neuroophthalmol ; 40(4): 558-565, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32991388

RESUMEN

BACKGROUND: Leber hereditary optic neuropathy (LHON) leads to bilateral central vision loss. In a clinical trial setting, idebenone has been shown to be safe and to provide a trend toward improved visual acuity, but long-term evidence of effectiveness in real-world clinical practice is sparse. METHODS: Open-label, multicenter, retrospective, noncontrolled analysis of long-term visual acuity and safety in 111 LHON patients treated with idebenone (900 mg/day) in an expanded access program. Eligible patients had a confirmed mitochondrial DNA mutation and had experienced the onset of symptoms (most recent eye) within 1 year before enrollment. Data on visual acuity and adverse events were collected as per normal clinical practice. Efficacy was assessed as the proportion of patients with either a clinically relevant recovery (CRR) or a clinically relevant stabilization (CRS) of visual acuity. In the case of CRR, time to and magnitude of recovery over the course of time were also assessed. RESULTS: At time of analysis, 87 patients had provided longitudinal efficacy data. Average treatment duration was 25.6 months. CRR was observed in 46.0% of patients. Analysis of treatment effect by duration showed that the proportion of patients with recovery and the magnitude of recovery increased with treatment duration. Average gain in best-corrected visual acuity for responders was 0.72 logarithm of the minimal angle of resolution (logMAR), equivalent to more than 7 lines on the Early Treatment Diabetic Retinopathy Study (ETDRS) chart. Furthermore, 50% of patients who had a visual acuity below 1.0 logMAR in at least one eye at initiation of treatment successfully maintained their vision below this threshold by last observation. Idebenone was well tolerated, with most adverse events classified as minor. CONCLUSIONS: These data demonstrate the benefit of idebenone treatment in recovering lost vision and maintaining good residual vision in a real-world setting. Together, these findings indicate that idebenone treatment should be initiated early and be maintained more than 24 months to maximize efficacy. Safety results were consistent with the known safety profile of idebenone.


Asunto(s)
Atrofia Óptica Hereditaria de Leber/tratamiento farmacológico , Ubiquinona/análogos & derivados , Agudeza Visual , Adolescente , Adulto , Anciano , Antioxidantes/uso terapéutico , Niño , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Atrofia Óptica Hereditaria de Leber/fisiopatología , Estudios Retrospectivos , Factores de Tiempo , Resultado del Tratamiento , Ubiquinona/uso terapéutico , Adulto Joven
7.
Hum Mol Genet ; 25(12): 2404-2416, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27106103

RESUMEN

Mitochondrial dysfunction connects metabolic disturbance with numerous pathologies, but the significance of mitochondrial activity in bone remains unclear. We have, therefore, characterized the skeletal phenotype in the Opa3L122P mouse model for Costeff syndrome, in which a missense mutation of the mitochondrial membrane protein, Opa3, impairs mitochondrial activity resulting in visual and metabolic dysfunction. Although widely expressed in the developing normal mouse head, Opa3 expression was restricted after E14.5 to the retina, brain, teeth and mandibular bone. Opa3 was also expressed in adult tibiae, including at the trabecular surfaces and in cortical osteocytes, epiphyseal chondrocytes, marrow adipocytes and mesenchymal stem cell rosettes. Opa3L122P mice displayed craniofacial abnormalities, including undergrowth of the lower mandible, accompanied in some individuals by cranial asymmetry and incisor malocclusion. Opa3L122P mice showed an 8-fold elevation in tibial marrow adiposity, due largely to increased adipogenesis. In addition, femoral length and cortical diameter and wall thickness were reduced, the weakening of the calcified tissue and the geometric component of strength reducing overall cortical strength in Opa3L122P mice by 65%. In lumbar vertebrae reduced vertebral body area and wall thickness were accompanied by a proportionate reduction in marrow adiposity. Although the total biomechanical strength of lumbar vertebrae was reduced by 35%, the strength of the calcified tissue (σmax) was proportionate to a 38% increase in trabecular number. Thus, mitochondrial function is important for the development and maintenance of skeletal integrity, impaired bone growth and strength, particularly in limb bones, representing a significant new feature of the Costeff syndrome phenotype.


Asunto(s)
Desarrollo Óseo/genética , Corea/genética , Errores Innatos del Metabolismo/genética , Mitocondrias/genética , Atrofia Óptica/genética , Proteínas/genética , Paraplejía Espástica Hereditaria/genética , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/fisiopatología , Corea/fisiopatología , Modelos Animales de Enfermedad , Cabeza/crecimiento & desarrollo , Cabeza/fisiopatología , Humanos , Mandíbula/crecimiento & desarrollo , Mandíbula/fisiopatología , Errores Innatos del Metabolismo/fisiopatología , Ratones , Mitocondrias/patología , Mutación Missense , Atrofia Óptica/fisiopatología , Retina/crecimiento & desarrollo , Retina/fisiopatología , Esqueleto/crecimiento & desarrollo , Esqueleto/fisiopatología , Paraplejía Espástica Hereditaria/fisiopatología , Diente/crecimiento & desarrollo , Diente/fisiopatología
9.
J Neuroophthalmol ; 37(4): 371-381, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28991104

RESUMEN

Leber hereditary optic neuropathy (LHON) is currently estimated as the most frequent mitochondrial disease (1 in 27,000-45,000). Its molecular pathogenesis and natural history is now fairly well understood. LHON also is the first mitochondrial disease for which a treatment has been approved (idebenone-Raxone, Santhera Pharmaceuticals) by the European Medicine Agency, under exceptional circumstances because of the rarity and severity of the disease. However, what remains unclear includes the optimal target population, timing, dose, and frequency of administration of idebenone in LHON due to lack of accepted definitions, criteria, and general guidelines for the clinical management of LHON. To address these issues, a consensus conference with a panel of experts from Europe and North America was held in Milan, Italy, in 2016. The intent was to provide expert consensus statements for the clinical and therapeutic management of LHON based on the currently available evidence. We report the conclusions of this conference, providing the guidelines for clinical and therapeutic management of LHON.


Asunto(s)
Consenso , Manejo de la Enfermedad , Oftalmología , Atrofia Óptica Hereditaria de Leber/tratamiento farmacológico , Sociedades Médicas , Ubiquinona/análogos & derivados , Antioxidantes/uso terapéutico , Congresos como Asunto , Humanos , Cooperación Internacional , Ubiquinona/uso terapéutico
10.
Acta Neuropathol ; 132(6): 789-806, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27696015

RESUMEN

Mitochondrial optic neuropathies constitute an important cause of chronic visual morbidity and registrable blindness in both the paediatric and adult population. It is a genetically heterogeneous group of disorders caused by both mitochondrial DNA (mtDNA) mutations and a growing list of nuclear genetic defects that invariably affect a critical component of the mitochondrial machinery. The two classical paradigms are Leber hereditary optic neuropathy (LHON), which is a primary mtDNA disorder, and autosomal dominant optic atrophy (DOA) secondary to pathogenic mutations within the nuclear gene OPA1 that encodes for a mitochondrial inner membrane protein. The defining neuropathological feature is the preferential loss of retinal ganglion cells (RGCs) within the inner retina but, rather strikingly, the smaller calibre RGCs that constitute the papillomacular bundle are particularly vulnerable, whereas melanopsin-containing RGCs are relatively spared. Although the majority of patients with LHON and DOA will present with isolated optic nerve involvement, some individuals will also develop additional neurological complications pointing towards a greater vulnerability of the central nervous system (CNS) in susceptible mutation carriers. These so-called "plus" phenotypes are mechanistically important as they put the loss of RGCs within the broader perspective of neuronal loss and mitochondrial dysfunction, highlighting common pathways that could be modulated to halt progressive neurodegeneration in other related CNS disorders. The management of patients with mitochondrial optic neuropathies still remains largely supportive, but the development of effective disease-modifying treatments is now within tantalising reach helped by major advances in drug discovery and delivery, and targeted genetic manipulation.


Asunto(s)
ADN Mitocondrial/genética , Atrofia Óptica Autosómica Dominante/genética , Atrofia Óptica Hereditaria de Leber/genética , Animales , Humanos
11.
Neurogenetics ; 16(1): 69-75, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25159689

RESUMEN

Leber hereditary optic neuropathy and autosomal dominant optic atrophy are the two most common inherited optic neuropathies. The latter has been associated with mutations in the OPA1 and OPA3 genes. To date, only six families with OPA3-associated dominant optic atrophy have been reported. In order to identify additional families, we performed Sanger sequencing of the OPA3 gene in 75 unrelated optic neuropathy patients. Affected individuals from two families were found to harbour the c.313C > G, p.(Gln105Glu) change in heterozygous state; this genetic defect has been previously reported in four dominant optic atrophy families. Intra- and interfamilial variability in age of onset and presenting symptoms was observed. Although dominant OPA3 mutations are typically associated with optic atrophy and cataracts, the former can be observed in isolation; we report a case with no lens opacities at age 38. Conversely, it is important to consider OPA3-related disease in individuals with bilateral infantile-onset cataracts and to assess optic nerve health in those whose vision fail to improve following lens surgery. The papillomacular bundle is primarily affected and vision is typically worse than 20/40. Notably, we describe one subject who retained normal acuities into the fifth decade of life. The condition can be associated with extraocular clinical features: two affected individuals in the present study had sensorineural hearing loss. The clinical heterogeneity observed in the individuals reported here (all having the same genetic defect in OPA3) suggests that the molecular pathology of the disorder is likely to be complex.


Asunto(s)
Mutación , Atrofia Óptica Autosómica Dominante/diagnóstico , Atrofia Óptica Autosómica Dominante/genética , Proteínas/genética , Adulto , Anciano , Análisis Mutacional de ADN , Femenino , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Disco Óptico/patología , Linaje , Agudeza Visual/genética , Adulto Joven
12.
Hum Mol Genet ; 21(22): 4836-44, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22869679

RESUMEN

The interrelationship between brown adipose tissue (BAT) and white adipose tissue (WAT) is emerging as an important factor in obesity, but the effect of impairing non-shivering thermogenesis in BAT on lipid storage in WAT remains unclear. To address this, we have characterized the metabolic phenotype of a mouse model for Costeff syndrome, in which a point mutation in the mitochondrial membrane protein Opa3 impairs mitochondrial activity. Opa3(L122P) mice displayed an 80% reduction in insulin-like growth factor 1, postnatal growth retardation and hepatic steatosis. A 90% reduction in uncoupling protein 1 (UCP1) expression in interscapular BAT was accompanied by a marked reduction in surface body temperature, with a 2.5-fold elevation in interscapular BAT mass and lipid storage. The sequestration of circulating lipid into BAT resulted in profound reductions in epididymal and retroperitoneal WAT mass, without affecting subcutaneous WAT. The histological appearance and intense mitochondrial staining in intra-abdominal WAT suggest significant 'browning', but with UCP1 expression in WAT of Opa3(L122P) mice only 62% of that in wild-type littermates, any precursor differentiation does not appear to result in thermogenically active beige adipocytes. Thus, we have identified Opa3 as a novel regulator of lipid metabolism, coupling lipid uptake with lipid processing in liver and with thermogenesis in BAT. These findings indicate that skeletal and metabolic impairment in Costeff syndrome may be more significant than previously thought and that uncoupling lipid uptake from lipid metabolism in BAT may represent a novel approach to controlling WAT mass in obesity.


Asunto(s)
Grasa Abdominal/metabolismo , Adiposidad/genética , Síndrome de Costello/genética , Síndrome de Costello/metabolismo , Mitocondrias/metabolismo , Proteínas/genética , Termogénesis/genética , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/patología , Animales , Síndrome de Costello/sangre , Modelos Animales de Enfermedad , Femenino , Genotipo , Metabolismo de los Lípidos , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Proteínas/metabolismo
13.
Cell Rep Med ; 5(3): 101437, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38428428

RESUMEN

Leber hereditary optic neuropathy (LHON) is a mitochondrial disease leading to rapid and severe bilateral vision loss. Idebenone has been shown to be effective in stabilizing and restoring vision in patients treated within 1 year of onset of vision loss. The open-label, international, multicenter, natural history-controlled LEROS study (ClinicalTrials.gov NCT02774005) assesses the efficacy and safety of idebenone treatment (900 mg/day) in patients with LHON up to 5 years after symptom onset (N = 199) and over a treatment period of 24 months, compared to an external natural history control cohort (N = 372), matched by time since symptom onset. LEROS meets its primary endpoint and confirms the long-term efficacy of idebenone in the subacute/dynamic and chronic phases; the treatment effect varies depending on disease phase and the causative mtDNA mutation. The findings of the LEROS study will help guide the clinical management of patients with LHON.


Asunto(s)
Atrofia Óptica Hereditaria de Leber , Ubiquinona/análogos & derivados , Humanos , Atrofia Óptica Hereditaria de Leber/tratamiento farmacológico , Atrofia Óptica Hereditaria de Leber/genética , Atrofia Óptica Hereditaria de Leber/diagnóstico , Antioxidantes/uso terapéutico , Ubiquinona/uso terapéutico , Ubiquinona/genética , Mutación
14.
Brain ; 135(Pt 2): 493-505, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22300878

RESUMEN

Retinal ganglion cell dendritic pruning has been reported in association with a 50% reduction in Opa1 transcript and protein in retinal and neural tissue, which manifests as visual dysfunction in the heterozygous mutant mouse, B6;C3-Opa1(Q285STOP). Here we report a marked reduction in retinal ganglion cell synaptic connectivity in the absence of soma loss and explore the mechanism and relationship between mitochondrial integrity and synaptic connectivity. We observed decreased levels of postsynaptic density protein 95 in Opa1(+/-) mutant mice consistent with synaptic loss in the inner plexiform layer. Glutamatergic but not γ-aminobutyric acid-ergic synaptic sites were reduced in Opa1(+/-) mice. We observed increased synaptic vesicle number in bipolar cell terminal arbours assessed by immunohistochemistry, electron microscopy and western blot analysis. These changes occur without significant loss of mitochondrial membrane potential in retina and optic nerve. Analysis of biolistically transfected retinal ganglion cells shows the retraction of mitochondria towards the soma, and mitochondrial fragmentation, preceding dendritic loss. These processes cast light on the intimate relationship between normal mitochondrial fusion and fission balances, as influenced by the OPA1 protein, in neural cell connectivity in the mammalian retina.


Asunto(s)
Complejo Mediador/metabolismo , Mitocondrias/metabolismo , Red Nerviosa/metabolismo , Atrofia Óptica Autosómica Dominante/metabolismo , Células Ganglionares de la Retina/metabolismo , Sinapsis/metabolismo , Animales , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Ácido Glutámico/metabolismo , Guanilato-Quinasas/genética , Guanilato-Quinasas/metabolismo , Complejo Mediador/genética , Potencial de la Membrana Mitocondrial/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Atrofia Óptica Autosómica Dominante/genética , Retina/metabolismo , Ácido gamma-Aminobutírico/metabolismo
15.
Exp Eye Res ; 93(5): 771-7, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21803037

RESUMEN

Autosomal dominant optic atrophy (ADOA) is a slowly progressive optic neuropathy caused by mutations in the OPA1 gene. OPA1 is ubiquitously expressed and plays a key role in mitochondrial fusion. Heterozygous Opa1 mutant mice (B6; C3-Opa1(Q285STOP)), have previously been reported to develop visual defects and optic nerve changes. In this study, in vivo visual electrophysiological testing (ERGs and VEPs) was performed on 11-13 month old B6; C3-Opa1(Q285STOP) mice (n = 5) and age/sex matched wildtype littermate controls. Full intensity series were recorded in response to brief (4 ms) single flash stimuli delivered in a Ganzfeld dome under dark- and light-adapted conditions. The major ERG components (a-wave and b-wave) showed no detectable difference from wildtype in the amplitude or implicit time of dark-adapted ERGs across the full intensity range tested. This was also true for the components of the dark-adapted VEP. However, the light-adapted ERG responses revealed a significant reduction in the photopic negative response (PhNR) amplitude in Opa1(+/-) animals relative to wildtypes at the brighter intensities tested. Elements of the light-adapted VEP were also abnormal in mutant mice. Overall Opa1(+/-) mice display functional deficits in electrophysiology that are consistent with ganglion cell dysfunction. These deficits may correlate with a reduction in the dendritic arborisation of retinal ganglion cells, which has been previously reported to occur at a similar age in the same mutant mouse line (Williams et al., 2010). The functional phenotype we have described in this mouse model may be useful in the robust and accurate assessment of potential treatments for ADOA.


Asunto(s)
Modelos Animales de Enfermedad , Potenciales Evocados Visuales/fisiología , Atrofia Óptica Autosómica Dominante/fisiopatología , Retina/fisiopatología , Trastornos de la Visión/fisiopatología , Animales , Adaptación a la Oscuridad , Electrorretinografía , GTP Fosfohidrolasas/genética , Ratones , Ratones Endogámicos C57BL , Atrofia Óptica Autosómica Dominante/genética , Estimulación Luminosa , Células Ganglionares de la Retina/patología
16.
Brain ; 133(10): 2942-51, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20817698

RESUMEN

The heterozygous mutation B6;C3-Opa1(Q285STOP), which models autosomal dominant optic atrophy, leads to a 50% reduction in Opa1 transcript and protein in the mouse retina and neural tissues and is associated with visual dysfunction and structural changes in the murine retina and optic nerve. In this article we use this model to quantify and evaluate the dendritic morphology of retinal ganglion cells. Retinal ganglion cells in Opa1(+/-) mutant mice (n=16) and accompanying age- and sex-matched controls (n=11) (age ranges of <10, 10-15 and >20 months) were labelled DiOlistically with carbocyanine dyes to quantify changes in dendritic tree architecture as a function of age. We observed localized dendritic reduction to sublamina b of the inner plexiform layer without retinal ganglion cell loss, showing dendritic pruning of on- but not off-centre retinal ganglion cells, and this effect was exacerbated with age. The mean dendritic field area was reduced in on-centre retinal ganglion cells of 10- to 15-month-old mice (-24.24%; C(V) =0.68; P<0.05) and >20-month-old mice (-43.22%; C(V) =0.75; P<0.05) compared with age-matched wild-type controls. Similar changes were seen in average total dendritic length in on-centre retinal ganglion cells of 10- to 15-month-old mice (-31.66%; C(V) =0.67; P<0.05) and >20-month-old mice (-49.55%; C(V) =0.63; P<0.05). Sholl analysis showed a marked difference in the dendritic arborization of on-centre retinal ganglion cells in the 10- to 15-month-old group (area under the curve -21.67%; P>0.05) and of the >20-month-old group (area under the curve -42.12%; P<0.05) compared with the control group. There was no detectable change in dendritic morphology in <10-month-old Opa1(+/-) mutant mice compared with wild-type (P>0.05). No significant changes (P>0.05) were seen in off-centre retinal ganglion cells. Finally, there was also no significant change (P>0.05) in the retinal ganglion cell count across all age groups. In conclusion, we show dendritic pruning in on-centre retinal ganglion cells of the Opa1(+/-) mouse model of autosomal dominant optic atrophy from as early as 10 months of age. These results highlight the importance of normal mitochondrial fusion balance, as influenced by the OPA1 protein in maintaining the dendritic morphology of retinal ganglion cells. Dendritic pruning precedes the onset of clinical visual loss and structural changes in the optic nerve in the absence of significant cell loss.


Asunto(s)
Dendritas/patología , GTP Fosfohidrolasas/metabolismo , Atrofia Óptica Autosómica Dominante/metabolismo , Células Ganglionares de la Retina/patología , Animales , Atrofia/metabolismo , Atrofia/patología , Recuento de Células , Dendritas/metabolismo , GTP Fosfohidrolasas/genética , Ratones , Ratones Transgénicos , Atrofia Óptica Autosómica Dominante/genética , Atrofia Óptica Autosómica Dominante/patología , Células Ganglionares de la Retina/metabolismo
17.
Front Neurol ; 12: 641259, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33927681

RESUMEN

Autosomal Dominant Optic Atrophy (ADOA) is an ophthalmological condition associated primarily with mutations in the OPA1 gene. It has variable onset, sometimes juvenile, but in other patients, the disease does not manifest until adult middle age despite the presence of a pathological mutation. Thus, individuals carrying mutations are considered healthy before the onset of clinical symptoms. Our research, nonetheless, indicates that on the cellular level pathology is evident from birth and mutant cells are different from controls. We argue that the adaptation and early recruitment of cytoprotective responses allows normal development and functioning but leads to an exhaustion of cellular reserves, leading to premature cellular aging, especially in neurons and skeletal muscle cells. The appearance of clinical symptoms, thus, indicates the overwhelming of natural cellular defenses and break-down of native protective mechanisms.

18.
Ther Adv Rare Dis ; 2: 26330040211029037, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-37181108

RESUMEN

Mitochondrial optic neuropathies are a group of optic nerve atrophies exemplified by the two commonest conditions in this group, autosomal dominant optic atrophy (ADOA) and Leber's hereditary optic neuropathy (LHON). Their clinical features comprise reduced visual acuity, colour vision deficits, centro-caecal scotomas and optic disc pallor with thinning of the retinal nerve fibre layer. The primary aetiology is genetic, with underlying nuclear or mitochondrial gene mutations. The primary pathology is owing to retinal ganglion cell dysfunction and degeneration. There is currently only one approved treatment and no curative therapy is available. In this review we summarise the genetic and clinical features of ADOA and LHON and then examine what new avenues there may be for therapeutic intervention. The therapeutic strategies to manage LHON and ADOA can be split into four categories: prevention, compensation, replacement and repair. Prevention is technically an option by modifying risk factors such as smoking cessation, or by utilising pre-implantation genetic diagnosis, although this is unlikely to be applied in mitochondrial optic neuropathies due to the non-life threatening and variable nature of these conditions. Compensation involves pharmacological interventions that ameliorate the mitochondrial dysfunction at a cellular and tissue level. Replacement and repair are exciting new emerging areas. Clinical trials, both published and underway, in this area are likely to reveal future potential benefits, since new therapies are desperately needed. Plain language summary: Optic nerve damage leading to loss of vision can be caused by a variety of insults. One group of conditions leading to optic nerve damage is caused by defects in genes that are essential for cells to make energy in small organelles called mitochondria. These conditions are known as mitochondrial optic neuropathies and two predominant examples are called autosomal dominant optic atrophy and Leber's hereditary optic neuropathy. Both conditions are caused by problems with the energy powerhouse of cells: mitochondria. The cells that are most vulnerable to this mitochondrial malfunction are called retinal ganglion cells, otherwise collectively known as the optic nerve, and they take the electrical impulse from the retina in the eye to the brain. The malfunction leads to death of some of the optic nerve cells, the degree of vision loss being linked to the number of those cells which are impacted in this way. Patients will lose visual acuity and colour vision and develop a central blind spot in their field of vision. There is currently no cure and very few treatment options. New treatments are desperately needed for patients affected by these devastating diseases. New treatments can potentially arise in four ways: prevention, compensation, replacement and repair of the defects. Here we explore how present and possible future treatments might provide hope for those suffering from these conditions.

19.
Redox Biol ; 43: 101988, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33932867

RESUMEN

Nicotinamide adenine dinucleotide (NAD) is a REDOX cofactor and metabolite essential for neuronal survival. Glaucoma is a common neurodegenerative disease in which neuronal levels of NAD decline. We assess the effects of nicotinamide (a precursor to NAD) on retinal ganglion cells (the affected neuron in glaucoma) in normal physiological conditions and across a range of glaucoma relevant insults including mitochondrial stress and axon degenerative insults. We demonstrate retinal ganglion cell somal, axonal, and dendritic neuroprotection by nicotinamide in rodent models which represent isolated ocular hypertensive, axon degenerative, and mitochondrial degenerative insults. We performed metabolomics enriched for small molecular weight metabolites for the retina, optic nerve, and superior colliculus which demonstrates that ocular hypertension induces widespread metabolic disruption, including consistent changes to α-ketoglutaric acid, creatine/creatinine, homocysteine, and glycerophosphocholine. This metabolic disruption is prevented by nicotinamide. Nicotinamide provides further neuroprotective effects by increasing oxidative phosphorylation, buffering and preventing metabolic stress, and increasing mitochondrial size and motility whilst simultaneously dampening action potential firing frequency. These data support continued determination of the utility of long-term nicotinamide treatment as a neuroprotective therapy for human glaucoma.


Asunto(s)
Glaucoma , Enfermedades Neurodegenerativas , Animales , Modelos Animales de Enfermedad , Humanos , Neuroprotección , Niacinamida , Células Ganglionares de la Retina
20.
Invest Ophthalmol Vis Sci ; 61(6): 42, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32561926

RESUMEN

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.


Asunto(s)
Metabolismo Energético , GTP Fosfohidrolasas/deficiencia , Mitocondrias/metabolismo , Mutación , Atrofia Óptica Autosómica Dominante/genética , Células Ganglionares de la Retina/metabolismo , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , GTP Fosfohidrolasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Atrofia Óptica Autosómica Dominante/metabolismo , Atrofia Óptica Autosómica Dominante/patología , Células Ganglionares de la Retina/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA