Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Pathol ; 191(8): 1454-1473, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34022179

RESUMEN

Age-related macular degeneration (AMD) is a progressive eye disease and the most common cause of blindness among the elderly. AMD is characterized by early atrophy of the choriocapillaris and retinal pigment epithelium (RPE). Although AMD is a multifactorial disease with many environmental and genetic risk factors, a hallmark of the disease is the origination of extracellular deposits, or drusen, between the RPE and Bruch membrane. Human retinal G-protein-coupled receptor (RGR) gene generates an exon-skipping splice variant of RGR-opsin (RGR-d; NP_001012740) that is a persistent component of small and large drusen. Herein, the findings show that abnormal RGR proteins, including RGR-d, are pathogenic in an animal retina with degeneration of the choriocapillaris, RPE, and photoreceptors. A frameshift truncating mutation resulted in severe retinal degeneration with a continuous band of basal deposits along the Bruch membrane. RGR-d produced less severe disease with choriocapillaris and RPE atrophy, including focal accumulation of abnormal RGR-d protein at the basal boundary of the RPE. Degeneration of the choriocapillaris was marked by a decrease in endothelial CD31 protein and choriocapillaris breakdown at the ultrastructural level. Fundus lesions with patchy depigmentation were characteristic of old RGR-d mice. RGR-d was mislocalized in cultured cells and caused a strong cell growth defect. These results uphold the notion of a potential hidden link between AMD and a high-frequency RGR allele.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Degeneración Macular/genética , Degeneración Macular/patología , Receptores Acoplados a Proteínas G/genética , Animales , Atrofia/patología , Coroides/metabolismo , Coroides/patología , Proteínas del Ojo/metabolismo , Humanos , Ratones , Receptores Acoplados a Proteínas G/metabolismo , Retina/metabolismo , Retina/patología
2.
Hum Mol Genet ; 26(R2): R139-R150, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28977448

RESUMEN

The optic nerve and the cells that give origin to its 1.2 million axons, the retinal ganglion cells (RGCs), are particularly vulnerable to neurodegeneration related to mitochondrial dysfunction. Optic neuropathies may range from non-syndromic genetic entities, to rare syndromic multisystem diseases with optic atrophy such as mitochondrial encephalomyopathies, to age-related neurodegenerative diseases such as Alzheimer's and Parkinson's disease where optic nerve involvement has, until recently, been a relatively overlooked feature. New tools are available to thoroughly investigate optic nerve function, allowing unparalleled access to this part of the central nervous system. Understanding the molecular pathophysiology of RGC neurodegeneration and optic atrophy, is key to broadly understanding the pathogenesis of neurodegenerative disorders, for monitoring their progression in describing the natural history, and ultimately as outcome measures to evaluate therapies. In this review, the different layers, from molecular to anatomical, that may contribute to RGC neurodegeneration and optic atrophy are tackled in an integrated way, considering all relevant players. These include RGC dendrites, cell bodies and axons, the unmyelinated retinal nerve fiber layer and the myelinated post-laminar axons, as well as olygodendrocytes and astrocytes, looked for unconventional functions. Dysfunctional mitochondrial dynamics, transport, homeostatic control of mitobiogenesis and mitophagic removal, as well as specific propensity to apoptosis may target differently cell types and anatomical settings. Ultimately, we can envisage new investigative approaches and therapeutic options that will speed the early diagnosis of neurodegenerative diseases and their cure.


Asunto(s)
Enfermedades del Nervio Óptico/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Animales , Apoptosis , Axones/metabolismo , ADN Mitocondrial/genética , Humanos , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Degeneración Nerviosa/genética , Enfermedades Neurodegenerativas/fisiopatología , Atrofia Óptica/fisiopatología , Atrofia Óptica Autosómica Dominante/genética , Atrofia Óptica Hereditaria de Leber/genética , Nervio Óptico/metabolismo , Nervio Óptico/patología , Retina/metabolismo
3.
Ann Neurol ; 79(1): 90-109, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26505992

RESUMEN

OBJECTIVE: Melanopsin retinal ganglion cells (mRGCs) are photoreceptors driving circadian photoentrainment, and circadian dysfunction characterizes Alzheimer disease (AD). We investigated mRGCs in AD, hypothesizing that they contribute to circadian dysfunction. METHODS: We assessed retinal nerve fiber layer (RNFL) thickness by optical coherence tomography (OCT) in 21 mild-moderate AD patients, and in a subgroup of 16 we evaluated rest-activity circadian rhythm by actigraphy. We studied postmortem mRGCs by immunohistochemistry in retinas, and axons in optic nerve cross-sections of 14 neuropathologically confirmed AD patients. We coimmunostained for retinal amyloid ß (Aß) deposition and melanopsin to locate mRGCs. All AD cohorts were compared with age-matched controls. RESULTS: We demonstrated an age-related optic neuropathy in AD by OCT, with a significant reduction of RNFL thickness (p = 0.038), more evident in the superior quadrant (p = 0.006). Axonal loss was confirmed in postmortem AD optic nerves. Abnormal circadian function characterized only a subgroup of AD patients. Sleep efficiency was significantly reduced in AD patients (p = 0.001). We also found a significant loss of mRGCs in postmortem AD retinal specimens (p = 0.003) across all ages and abnormal mRGC dendritic morphology and size (p = 0.003). In flat-mounted AD retinas, Aß accumulation was remarkably evident inside and around mRGCs. INTERPRETATION: We show variable degrees of rest-activity circadian dysfunction in AD patients. We also demonstrate age-related loss of optic nerve axons and specifically mRGC loss and pathology in postmortem AD retinal specimens, associated with Aß deposition. These results all support the concept that mRGC degeneration is a contributor to circadian rhythm dysfunction in AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Axones/patología , Trastornos Cronobiológicos , Nervio Óptico/patología , Células Ganglionares de la Retina , Actigrafía , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Trastornos Cronobiológicos/etiología , Trastornos Cronobiológicos/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Opsinas de Bastones/metabolismo , Tomografía de Coherencia Óptica
4.
Brain ; 137(Pt 2): 335-53, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24369379

RESUMEN

Leber's hereditary optic neuropathy is a maternally inherited blinding disease caused as a result of homoplasmic point mutations in complex I subunit genes of mitochondrial DNA. It is characterized by incomplete penetrance, as only some mutation carriers become affected. Thus, the mitochondrial DNA mutation is necessary but not sufficient to cause optic neuropathy. Environmental triggers and genetic modifying factors have been considered to explain its variable penetrance. We measured the mitochondrial DNA copy number and mitochondrial mass indicators in blood cells from affected and carrier individuals, screening three large pedigrees and 39 independently collected smaller families with Leber's hereditary optic neuropathy, as well as muscle biopsies and cells isolated by laser capturing from post-mortem specimens of retina and optic nerves, the latter being the disease targets. We show that unaffected mutation carriers have a significantly higher mitochondrial DNA copy number and mitochondrial mass compared with their affected relatives and control individuals. Comparative studies of fibroblasts from affected, carriers and controls, under different paradigms of metabolic demand, show that carriers display the highest capacity for activating mitochondrial biogenesis. Therefore we postulate that the increased mitochondrial biogenesis in carriers may overcome some of the pathogenic effect of mitochondrial DNA mutations. Screening of a few selected genetic variants in candidate genes involved in mitochondrial biogenesis failed to reveal any significant association. Our study provides a valuable mechanism to explain variability of penetrance in Leber's hereditary optic neuropathy and clues for high throughput genetic screening to identify the nuclear modifying gene(s), opening an avenue to develop predictive genetic tests on disease risk and therapeutic strategies.


Asunto(s)
ADN Mitocondrial/genética , Recambio Mitocondrial/genética , Atrofia Óptica Hereditaria de Leber/diagnóstico , Atrofia Óptica Hereditaria de Leber/genética , Penetrancia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje , Adulto Joven
5.
Proc Natl Acad Sci U S A ; 109(49): 20065-70, 2012 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-23129651

RESUMEN

An animal model of Leber hereditary optic neuropathy (LHON) was produced by introducing the human optic atrophy mtDNA ND6 P25L mutation into the mouse. Mice with this mutation exhibited reduction in retinal function by elecroretinogram (ERG), age-related decline in central smaller caliber optic nerve fibers with sparing of larger peripheral fibers, neuronal accumulation of abnormal mitochondria, axonal swelling, and demyelination. Mitochondrial analysis revealed partial complex I and respiration defects and increased reactive oxygen species (ROS) production, whereas synaptosome analysis revealed decreased complex I activity and increased ROS but no diminution of ATP production. Thus, LHON pathophysiology may result from oxidative stress.


Asunto(s)
ADN Mitocondrial/genética , Modelos Animales de Enfermedad , NADH Deshidrogenasa/genética , Atrofia Óptica Hereditaria de Leber/genética , Atrofia Óptica Hereditaria de Leber/fisiopatología , Estrés Oxidativo/fisiología , Retina/patología , Adenosina Trifosfato/metabolismo , Factores de Edad , Animales , Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/patología , Electrorretinografía , Humanos , Immunoblotting , Ratones , Mutación Missense/genética , Atrofia Óptica Hereditaria de Leber/complicaciones , Nervio Óptico/patología , Especies Reactivas de Oxígeno/metabolismo , Sinaptosomas/metabolismo
6.
Cell Rep Med ; 5(2): 101383, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38272025

RESUMEN

Idebenone, the only approved treatment for Leber hereditary optic neuropathy (LHON), promotes recovery of visual function in up to 50% of patients, but we can neither predict nor understand the non-responders. Idebenone is reduced by the cytosolic NAD(P)H oxidoreductase I (NQO1) and directly shuttles electrons to respiratory complex III, bypassing complex I affected in LHON. We show here that two polymorphic variants drastically reduce NQO1 protein levels when homozygous or compound heterozygous. This hampers idebenone reduction. In its oxidized form, idebenone inhibits complex I, decreasing respiratory function in cells. By retrospectively analyzing a large cohort of idebenone-treated LHON patients, classified by their response to therapy, we show that patients with homozygous or compound heterozygous NQO1 variants have the poorest therapy response, particularly if carrying the m.3460G>A/MT-ND1 LHON mutation. These results suggest consideration of patient NQO1 genotype and mitochondrial DNA mutation in the context of idebenone therapy.


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/metabolismo , Antioxidantes/uso terapéutico , Antioxidantes/farmacología , Estudios Retrospectivos , Ubiquinona/farmacología , Ubiquinona/uso terapéutico , Ubiquinona/metabolismo , Complejo I de Transporte de Electrón/genética , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo
7.
Brain ; 134(Pt 1): 220-34, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20943885

RESUMEN

Leber's hereditary optic neuropathy, the most frequent mitochondrial disease due to mitochondrial DNA point mutations in complex I, is characterized by the selective degeneration of retinal ganglion cells, leading to optic atrophy and loss of central vision prevalently in young males. The current study investigated the reasons for the higher prevalence of Leber's hereditary optic neuropathy in males, exploring the potential compensatory effects of oestrogens on mutant cell metabolism. Control and Leber's hereditary optic neuropathy osteosarcoma-derived cybrids (11778/ND4, 3460/ND1 and 14484/ND6) were grown in glucose or glucose-free, galactose-supplemented medium. After having shown the nuclear and mitochondrial localization of oestrogen receptors in cybrids, experiments were carried out by adding 100 nM of 17ß-oestradiol. In a set of experiments, cells were pre-incubated with the oestrogen receptor antagonist ICI 182780. Leber's hereditary optic neuropathy cybrids in galactose medium presented overproduction of reactive oxygen species, which led to decrease in mitochondrial membrane potential, increased apoptotic rate, loss of cell viability and hyper-fragmented mitochondrial morphology compared with control cybrids. Treatment with 17ß-oestradiol significantly rescued these pathological features and led to the activation of the antioxidant enzyme superoxide dismutase 2. In addition, 17ß-oestradiol induced a general activation of mitochondrial biogenesis and a small although significant improvement in energetic competence. All these effects were oestrogen receptor mediated. Finally, we showed that the oestrogen receptor ß localizes to the mitochondrial network of human retinal ganglion cells. Our results strongly support a metabolic basis for the unexplained male prevalence in Leber's hereditary optic neuropathy and hold promises for a therapeutic use for oestrogen-like molecules.


Asunto(s)
Estradiol/farmacología , Mitocondrias/efectos de los fármacos , Atrofia Óptica Hereditaria de Leber/fisiopatología , Células Ganglionares de la Retina/efectos de los fármacos , Análisis de Varianza , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Western Blotting , Línea Celular , ADN Mitocondrial/metabolismo , Estradiol/metabolismo , Estrógenos/metabolismo , Estrógenos/farmacología , Humanos , Inmunohistoquímica , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/fisiología , Atrofia Óptica Hereditaria de Leber/metabolismo , Atrofia Óptica Hereditaria de Leber/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/metabolismo
9.
Cell Rep ; 40(3): 111124, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35858578

RESUMEN

Leber's hereditary optic neuropathy (LHON), a disease associated with a mitochondrial DNA mutation, is characterized by blindness due to degeneration of retinal ganglion cells (RGCs) and their axons, which form the optic nerve. We show that a sustained pathological autophagy and compartment-specific mitophagy activity affects LHON patient-derived cells and cybrids, as well as induced pluripotent-stem-cell-derived neurons. This is variably counterbalanced by compensatory mitobiogenesis. The aberrant quality control disrupts mitochondrial homeostasis as reflected by defective bioenergetics and excessive reactive oxygen species production, a stress phenotype that ultimately challenges cell viability by increasing the rate of apoptosis. We counteract this pathological mechanism by using autophagy regulators (clozapine and chloroquine) and redox modulators (idebenone), as well as genetically activating mitochondrial biogenesis (PGC1-α overexpression). This study substantially advances our understanding of LHON pathophysiology, providing an integrated paradigm for pathogenesis of mitochondrial diseases and druggable targets for therapy.


Asunto(s)
Atrofia Óptica Hereditaria de Leber , ADN Mitocondrial/genética , Homeostasis , Humanos , Mitocondrias/genética , Mitofagia/genética , Mutación , Atrofia Óptica Hereditaria de Leber/genética , Atrofia Óptica Hereditaria de Leber/patología
10.
Brain ; 133(Pt 8): 2426-38, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20659957

RESUMEN

Mitochondrial optic neuropathies, that is, Leber hereditary optic neuropathy and dominant optic atrophy, selectively affect retinal ganglion cells, causing visual loss with relatively preserved pupillary light reflex. The mammalian eye contains a light detection system based on a subset of retinal ganglion cells containing the photopigment melanopsin. These cells give origin to the retinohypothalamic tract and support the non-image-forming visual functions of the eye, which include the photoentrainment of circadian rhythms, light-induced suppression of melatonin secretion and pupillary light reflex. We studied the integrity of the retinohypothalamic tract in five patients with Leber hereditary optic neuropathy, in four with dominant optic atrophy and in nine controls by testing the light-induced suppression of nocturnal melatonin secretion. This response was maintained in optic neuropathy subjects as in controls, indicating that the retinohypothalamic tract is sufficiently preserved to drive light information detected by melanopsin retinal ganglion cells. We then investigated the histology of post-mortem eyes from two patients with Leber hereditary optic neuropathy and one case with dominant optic atrophy, compared with three age-matched controls. On these retinas, melanopsin retinal ganglion cells were characterized by immunohistochemistry and their number and distribution evaluated by a new protocol. In control retinas, we show that melanopsin retinal ganglion cells are lost with age and are more represented in the parafoveal region. In patients, we demonstrate a relative sparing of these cells compared with the massive loss of total retinal ganglion cells, even in the most affected areas of the retina. Our results demonstrate that melanopsin retinal ganglion cells resist neurodegeneration due to mitochondrial dysfunction and maintain non-image-forming functions of the eye in these visually impaired patients. We also show that in normal human retinas, these cells are more concentrated around the fovea and are lost with ageing. The current results provide a plausible explanation for the preservation of pupillary light reaction despite profound visual loss in patients with mitochondrial optic neuropathy, revealing the robustness of melanopsin retinal ganglion cells to a metabolic insult and opening the question of mechanisms that might protect these cells.


Asunto(s)
Degeneración Nerviosa/fisiopatología , Atrofia Óptica Autosómica Dominante/fisiopatología , Atrofia Óptica Hereditaria de Leber/fisiopatología , Células Ganglionares de la Retina/fisiología , Opsinas de Bastones/metabolismo , Vías Visuales/fisiopatología , Adulto , Anciano de 80 o más Años , Envejecimiento/patología , Envejecimiento/fisiología , Estudios de Casos y Controles , Femenino , Humanos , Hipotálamo/patología , Hipotálamo/fisiopatología , Masculino , Persona de Mediana Edad , Enfermedades Mitocondriales/patología , Enfermedades Mitocondriales/fisiopatología , Degeneración Nerviosa/patología , Atrofia Óptica Autosómica Dominante/patología , Atrofia Óptica Hereditaria de Leber/patología , Retina/patología , Retina/fisiopatología , Células Ganglionares de la Retina/patología , Vías Visuales/patología
11.
J Neuroophthalmol ; 31(1): 6-11, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21139512

RESUMEN

BACKGROUND: Leber hereditary optic neuropathy (LHON) is a mitochondrial DNA (mtDNA) genetic disorder characterized by profound bilateral loss of central vision due to selective loss of retinal ganglion cells. Most patients with LHON do not have complaints related to the peripheral nervous system. We investigated possible qualitative and quantitative histological changes in the peripheral nerve of a patient with LHON as compared to normal controls. METHODS: Brachial plexus specimens were obtained at necropsy from a patient with LHON carrying the 3460/ND1 mtDNA mutation and age-matched controls without known history of neurological disease. The nerves were evaluated by light microscope coupled to a digital camera-based morphometric analysis and electron microscopy. RESULTS: Extensive axonal degeneration of the large heavily myelinated fibers was found in the brachial plexus from the patient with LHON. In LHON nerve fascicles, we counted over 10 times as many degenerated profiles as found in the control nerve fascicles. CONCLUSIONS: Microscopic examination of the brachial plexus in the patient with LHON clearly demonstrated a significant pattern of neurodegeneration. Our study suggests that peripheral neuropathy may be a subclinical feature associated with LHON.


Asunto(s)
Neuropatías del Plexo Braquial/genética , Neuropatías del Plexo Braquial/patología , Atrofia Óptica Hereditaria de Leber/complicaciones , Atrofia Óptica Hereditaria de Leber/genética , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/patología , Anciano , Neuropatías del Plexo Braquial/fisiopatología , Progresión de la Enfermedad , Femenino , Humanos , Atrofia Óptica Hereditaria de Leber/fisiopatología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Degeneración Walleriana/genética , Degeneración Walleriana/patología , Degeneración Walleriana/fisiopatología
12.
J Neuroophthalmol ; 31(2): 139-46, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21593627

RESUMEN

BACKGROUND: Alzheimer disease (AD) is associated with optic nerve degeneration, yet the underlying pathophysiology of this disease and the optic nerve disorder remain poorly understood. Low-density lipoprotein receptor-related protein (LRP) is implicated in the pathogenesis of AD by mediating the transport of amyloid-ß (Aß) out of the brain into the systemic circulation. As a key player in the reaction to central nervous system injury, astrocytes associate with LRP in AD. This study investigates the role of LRP and astrocytes in the pathogenesis of AD optic neuropathy. METHODS: To investigate the role of LRP and astrocytes in the pathogenesis of AD optic neuropathy, we conducted immunohistochemical studies on postmortem optic nerves in AD patients (n = 11) and age-matched controls (n = 10) to examine the presence of LRP. Quantitative analyses using imaging software were used to document the extent of LRP in neural tissues. Axonal integrity was assessed by performing immunohistochemistry on the subjects' optic nerves with an antibody to neurofilament (NF) protein. Double-immunofluorescence labeling was performed to investigate whether LRP colocalized with astrocytes, expressing glial fibrillary acidic protein. RESULTS: LRP expression was decreased in AD optic nerves compared to that in controls (P < 0.001). LRP immunoreactivity was observed in the microvasculature and perivascularly in close proximity to the astrocytic processes. Colocalization of LRP in the astrocytes of optic nerves was also demonstrated. The presence of optic neuropathy was confirmed in the AD optic nerves by demonstrating greatly reduced immunostaining for NF protein as compared to controls. CONCLUSIONS: The reduction of LRP in the AD degenerative optic nerves supports the hypothesis that LRP may play a role in the pathophysiology of AD optic neuropathy.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Proteínas Relacionadas con Receptor de LDL/metabolismo , Enfermedades del Nervio Óptico/etiología , Enfermedades del Nervio Óptico/metabolismo , Anciano , Anciano de 80 o más Años , Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Autopsia/métodos , Estudios de Casos y Controles , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neurofilamentos/metabolismo , Enfermedades del Nervio Óptico/patología
13.
Biochim Biophys Acta ; 1787(5): 518-28, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19268652

RESUMEN

Since the early days of mitochondrial medicine, it has been clear that optic atrophy is a very common and sometimes the singular pathological feature in mitochondrial disorders. The first point mutation of mitochondrial DNA (mtDNA) associated with the maternally inherited blinding disorder, Leber's hereditary optic neuropathy (LHON), was recognized in 1988. In 2000, the other blinding disorder, dominant optic atrophy (DOA) Kjer type, was found associated with mutations in the nuclear gene OPA1 that encodes a mitochondrial protein. Besides these two non-syndromic optic neuropathies, optic atrophy is a prominent feature in many other neurodegenerative diseases that are now recognized as due to primary mitochondrial dysfunction. We will consider mtDNA based syndromes such as LHON/dystonia/Mitochondrial Encephalomyopahty Lactic Acidosis Stroke-like (MELAS)/Leigh overlapping syndrome, or nuclear based diseases such as Friedreich ataxia (mutations in FXN gene), deafness-dystonia-optic atrophy (Mohr-Tranebjerg) syndrome (mutations in TIMM8A), complicated hereditary spastic paraplegia (mutations in SPG7), DOA "plus" syndromes (mutations in OPA1), Charcot-Marie-Tooth type 2A (CMT2A) with optic atrophy or hereditary motor and sensory neuropathy type VI (HMSN VI) (mutations in MFN2), and Costeff syndrome and DOA with cataract (mutations in OPA3). Thus, genetic errors in both nuclear and mitochondrial genomes often lead to retinal ganglion cell death, a specific target for mitochondrial mediated neurodegeneration. Many mechanisms have been studied and proposed as the bases for the pathogenesis of mitochondrial optic neuropathies including bioenergetic failure, oxidative stress, glutamate toxicity, abnormal mitochondrial dynamics and axonal transport, and susceptibility to apoptosis.


Asunto(s)
ADN Mitocondrial/genética , Enfermedades Mitocondriales/genética , Degeneración Nerviosa/genética , Atrofia Óptica Hereditaria de Leber/genética , Células Ganglionares de la Retina/patología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Ciclo Celular/genética , Núcleo Celular/genética , ADN/genética , Dinaminas , Ataxia de Friedreich/genética , GTP Fosfohidrolasas/genética , Humanos , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Mitocondriales/genética , Proteínas Nucleares/genética , Atrofia Óptica Hereditaria de Leber/patología , Síndromes Orofaciodigitales/genética
15.
Nat Commun ; 11(1): 4029, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32788597

RESUMEN

In autosomal dominant optic atrophy (ADOA), caused by mutations in the mitochondrial cristae biogenesis and fusion protein optic atrophy 1 (Opa1), retinal ganglion cell (RGC) dysfunction and visual loss occur by unknown mechanisms. Here, we show a role for autophagy in ADOA pathogenesis. In RGCs expressing mutated Opa1, active 5' AMP-activated protein kinase (AMPK) and its autophagy effector ULK1 accumulate at axonal hillocks. This AMPK activation triggers localized hillock autophagosome accumulation and mitophagy, ultimately resulting in reduced axonal mitochondrial content that is restored by genetic inhibition of AMPK and autophagy. In C. elegans, deletion of AMPK or of key autophagy and mitophagy genes normalizes the axonal mitochondrial content that is reduced upon mitochondrial dysfunction. In conditional, RGC specific Opa1-deficient mice, depletion of the essential autophagy gene Atg7 normalizes the excess autophagy and corrects the visual defects caused by Opa1 ablation. Thus, our data identify AMPK and autophagy as targetable components of ADOA pathogenesis.


Asunto(s)
Autofagia , Atrofia Óptica Autosómica Dominante/complicaciones , Trastornos de la Visión/complicaciones , Adenilato Quinasa/metabolismo , Animales , Autofagia/genética , Axones/patología , Caenorhabditis elegans/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , GTP Fosfohidrolasas/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitofagia , Mutación/genética , Fosforilación , Células Ganglionares de la Retina/patología
16.
Eye (Lond) ; 34(9): 1624-1630, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-31804625

RESUMEN

BACKGROUND/OBJECTIVES: Choroidal thinning has been suggested in Leber's hereditary optic neuropathy (LHON). No study has been conducted of the choroid in relation to the retinal ganglion cell-inner plexiform layer (RGC-IPL). We sought to measure choroidal thickness in chronic LHON and to correlate thickness changes with the RGC-IPL. SUBJECTS/METHODS: Chronic LHON, 11778 mitochondrial DNA (mtDNA) mutation, patients (26 eyes; mean age: 35.1 ± 16.1 years) were prospectively recruited at Doheny Eye Center, University of California Los Angeles from March 2016 to July 2017. Age-matched healthy controls (27 eyes; mean age: 32.4 ± 11.1 years) were enroled for comparison. Swept-source optical coherence tomography (SS-OCT) imaging was performed in chronic LHON patients and compared with age-matched healthy controls. RESULTS: The macular choroid was significantly thinner in chronic LHON (250.5 ± 62.2 µm) compared with controls (313.9 ± 60.2 µm; p < 0.0001). The peripapillary choroid was also significantly thinner in chronic LHON (135.7 ± 51.4 µm) compared with controls (183.0 ± 61.8 µm, p < 0.001). Choroidal thickness strongly correlated with retinal nerve fibre layer (RNFL) thickness in both the macular (R2 = 0.72; 95% CI, 0.57-0.84) and peripapillary regions (R2 = 0.53; 95% CI, 0.31-0.70). Choroidal thickness was also significantly correlated with macular RGC-IPL thickness (R2 = 0.51; 95% CI, 0.26-0.73). CONCLUSIONS: Choroidal thinning in chronic LHON correlated strongly with both RNFL and RGC-IPL thicknesses. These findings may suggest a pathophysiological mechanism involving vascular pathology of the choroid in relation to the retinal ganglion cell complex in LHON.


Asunto(s)
Atrofia Óptica Hereditaria de Leber , Células Ganglionares de la Retina , Adolescente , Adulto , Coroides , Humanos , Persona de Mediana Edad , Fibras Nerviosas , Estudios Prospectivos , Tomografía de Coherencia Óptica , Adulto Joven
18.
PLoS One ; 15(5): e0232785, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32469871

RESUMEN

BACKGROUND: Alzheimer's disease (AD) pathology precedes symptoms and its detection can identify at-risk individuals who may benefit from early treatment. Since the retinal nerve fiber layer (RNFL) is depleted in established AD, we tested whether its thickness can predict whether cognitively healthy (CH) individuals have a normal or pathological cerebrospinal fluid (CSF) Aß42 (A) and tau (T) ratio. METHODS: As part of an ongoing longitudinal study, we enrolled CH individuals, excluding those with cognitive impairment and significant ocular pathology. We classified the CH group into two sub-groups, normal (CH-NAT, n = 16) or pathological (CH-PAT, n = 27), using a logistic regression model from the CSF AT ratio that identified >85% of patients with a clinically probable AD diagnosis. Spectral-domain optical coherence tomography (OCT) was acquired for RNFL, ganglion cell-inner plexiform layer (GC-IPL), and macular thickness. Group differences were tested using mixed model repeated measures and a classification model derived using multiple logistic regression. RESULTS: Mean age (± standard deviation) in the CH-PAT group (n = 27; 75.2 ± 8.4 years) was similar (p = 0.50) to the CH-NAT group (n = 16; 74.1 ± 7.9 years). Mean RNFL (standard error) was thinner in the CH-PAT group by 9.8 (2.7) µm; p < 0.001. RNFL thickness classified CH-NAT vs. CH-PAT with 87% sensitivity and 56.3% specificity. CONCLUSIONS: Our retinal data predict which individuals have CSF biomarkers of AD pathology before cognitive deficits are detectable with 87% sensitivity. Such results from easy-to-acquire, objective and non-invasive measurements of the RNFL merit further study of OCT technology to monitor or screen for early AD pathology.


Asunto(s)
Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Disfunción Cognitiva/genética , Proteínas tau/genética , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/líquido cefalorraquídeo , Amiloidosis/líquido cefalorraquídeo , Amiloidosis/diagnóstico por imagen , Amiloidosis/genética , Amiloidosis/patología , Biomarcadores/líquido cefalorraquídeo , Disfunción Cognitiva/líquido cefalorraquídeo , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fibras Nerviosas/metabolismo , Fibras Nerviosas/patología , Disco Óptico/diagnóstico por imagen , Disco Óptico/metabolismo , Disco Óptico/patología , Retina/diagnóstico por imagen , Retina/metabolismo , Retina/patología , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Tomografía de Coherencia Óptica , Proteínas tau/líquido cefalorraquídeo
19.
Acta Neuropathol ; 118(3): 381-9, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19277685

RESUMEN

Although Alzheimer's disease (AD) has been shown to be associated with a true primary optic neuropathy, the underlying pathophysiology of this disease and in particular the optic nerve disorder is still poorly understood. The receptor for advanced glycation end products (RAGE) has been implicated in the pathogenesis of AD by mediating the transport of plasma amyloid-beta into the brain. Once ligated, RAGE can play a role in signal transduction, leading to amplification and perpetuation of inflammatory processes. As a key player in the reaction to CNS injury, astrocytes have been shown to associate with RAGE in a number of diseases, including AD. To investigate the role of RAGE and astrocytes in the pathogenesis of AD optic neuropathy, we conducted immunohistochemical studies to examine the presence of RAGE in donor eyes from patients with AD (n = 10) and controls (n = 3). Both qualitative observation and quantitative analyses using imaging software were used to document the extent of RAGE in the neural tissues. The intensity and extent of RAGE expression was more prominent in AD nerves compared to controls (P < 0.05). The RAGE immunoreactivity was observed in the microvasculature and in close proximity to astrocytic processes. While RAGE immunoreactivity increased with age, the increase was more precipitous in the AD group compared to the controls. The up-regulation of RAGE in the AD optic nerves indicates that RAGE may play a role in the pathophysiology of AD optic neuropathy.


Asunto(s)
Enfermedad de Alzheimer/patología , Astrocitos/metabolismo , Enfermedades del Nervio Óptico/patología , Nervio Óptico/patología , Receptores Inmunológicos/metabolismo , Factores de Edad , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/complicaciones , Femenino , Humanos , Inmunohistoquímica , Masculino , Microvasos/metabolismo , Microvasos/patología , Persona de Mediana Edad , Nervio Óptico/metabolismo , Enfermedades del Nervio Óptico/etiología , Receptor para Productos Finales de Glicación Avanzada , Regulación hacia Arriba
20.
Alzheimers Dement (Amst) ; 11: 775-783, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31737776

RESUMEN

INTRODUCTION: Previous in vivo optical coherence tomography studies have proposed the retinal choroid as a potential oculovascular biomarker for Alzheimer's disease (AD). However, the clinical use of the choroid as a purported surrogate marker remains poorly understood. We pursued a histopathological approach to assess choroidal thickness and vascular morphology in severe disease. METHODS: Human postmortem tissues from 8 patients with AD (mean age: 80.1 ± 12.7 years) and from 11 age-matched controls (mean age: 78.4 ± 16.57 years) were analyzed. Thickness, area, and vascularity of the retinal choroid and its sublayers were measured from the nasal and temporal quadrants of the superior retina. RESULTS: Nasally, the choroid was thinner in the patients with AD than in the controls (22% thickness reduction; P < .001), but to our surprise, the choroid was thicker in the patients with AD than in the controls (~60% increase; P < .03) within the macula, temporally. The choroidal area was also significantly greater in the patients with AD than in the controls (~60% increase; P < .03). Choroidal thickening in AD was strongly correlated with the stromal vessel number (R2 = 0.96, P < .001). DISCUSSION: We found significant differences in the retinal choroid by layer and by region, nasally and temporally with respect to the optic nerve. Intriguingly, the choroid was markedly thicker in the central macular region and was strongly associated with vessel number in the stromal vascular layer. These quantified histological findings in severe disease expand our understanding of vascular pathology in AD and suggest vascularity as a potential biomarker supplementary to thickness when evaluating the retinal choroid in AD.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA