Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neurotherapeutics ; 20(4): 1215-1228, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37268847

RESUMEN

Giant axonal neuropathy (GAN) is a disease caused by a deficiency of gigaxonin, a mediator of the degradation of intermediate filament (IF) proteins. A lack of gigaxonin alters the turnover of IF proteins, provoking accumulation and disorganization of neurofilaments (NFs) in neurons, a hallmark of the disease. However, the effects of IF disorganization on neuronal function remain unknown. Here, we report that cultured embryonic dorsal root ganglia (DRG) neurons derived from Gan-/- mice exhibit accumulations of IF proteins and defects in fast axonal transport of organelles. Kymographs generated by time-lapse microscopy revealed substantial reduction of anterograde movements of mitochondria and lysosomes in axons of Gan-/- DRG neurons. Treatment of Gan-/- DRG neurons with Tubastatin A (TubA) increased the levels of acetylated tubulin and it restored the normal axonal transport of these organelles. Furthermore, we tested the effects of TubA in a new mouse model of GAN consisting of Gan-/- mice with overexpression of peripherin (Prph) transgene. Treatment of 12-month-old Gan-/-;TgPer mice with TubA led to a slight amelioration of motor function, especially a significant improvement of gait performance as measured by footprint analyses. Moreover, TubA treatment reduced the abnormal accumulations of Prph and NF proteins in spinal neurons and it boosted the levels of Prph transported into peripheral nerve axons. These results suggest that drug inhibitors of histone deacetylase aiming to enhance axonal transport should be considered as a potential treatment for GAN disease.


Asunto(s)
Proteínas del Citoesqueleto , Neuropatía Axonal Gigante , Ratones , Animales , Proteínas del Citoesqueleto/metabolismo , Transporte Axonal , Proteínas de Filamentos Intermediarios/metabolismo , Axones/metabolismo , Neuropatía Axonal Gigante/metabolismo , Neuropatía Axonal Gigante/terapia , Ganglios Espinales/metabolismo
2.
FASEB J ; 37(5): e22886, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37043392

RESUMEN

Gigaxonin is an adaptor protein for E3 ubiquitin ligase substrates. It is necessary for ubiquitination and degradation of intermediate filament (IF) proteins. Giant axonal neuropathy is a pathological condition caused by mutations in the GAN gene that encodes gigaxonin. This condition is characterized by abnormal accumulation of IFs in both neuronal and non-neuronal cells; however, it is unclear what causes IF aggregation. In this work, we studied the dynamics of IFs using their subunits tagged with a photoconvertible protein mEOS 3.2. We have demonstrated that the loss of gigaxonin dramatically inhibited transport of IFs along microtubules by the microtubule motor kinesin-1. This inhibition was specific for IFs, as other kinesin-1 cargoes, with the exception of mitochondria, were transported normally. Abnormal distribution of IFs in the cytoplasm can be rescued by direct binding of kinesin-1 to IFs, demonstrating that transport inhibition is the primary cause for the abnormal IF distribution. Another effect of gigaxonin loss was a more than 20-fold increase in the amount of soluble vimentin oligomers in the cytosol of gigaxonin knock-out cells. We speculate that these oligomers saturate a yet unidentified adapter that is required for kinesin-1 binding to IFs, which might inhibit IF transport along microtubules causing their abnormal accumulation.


Asunto(s)
Proteínas del Citoesqueleto , Neuropatía Axonal Gigante , Humanos , Proteínas del Citoesqueleto/metabolismo , Filamentos Intermedios/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Neuropatía Axonal Gigante/genética , Neuropatía Axonal Gigante/metabolismo , Neuropatía Axonal Gigante/patología , Microtúbulos/metabolismo
3.
JCI Insight ; 5(1)2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31944090

RESUMEN

Gigaxonin (also known as KLHL16) is an E3 ligase adaptor protein that promotes the ubiquitination and degradation of intermediate filament (IF) proteins. Mutations in human gigaxonin cause the fatal neurodegenerative disease giant axonal neuropathy (GAN), in which IF proteins accumulate and aggregate in axons throughout the nervous system, impairing neuronal function and viability. Despite this pathophysiological significance, the upstream regulation and downstream effects of normal and aberrant gigaxonin function remain incompletely understood. Here, we report that gigaxonin is modified by O-linked ß-N-acetylglucosamine (O-GlcNAc), a prevalent form of intracellular glycosylation, in a nutrient- and growth factor­dependent manner. MS analyses of human gigaxonin revealed 9 candidate sites of O-GlcNAcylation, 2 of which ­ serine 272 and threonine 277 ­ are required for its ability to mediate IF turnover in gigaxonin-deficient human cell models that we created. Taken together, the results suggest that nutrient-responsive gigaxonin O-GlcNAcylation forms a regulatory link between metabolism and IF proteostasis. Our work may have significant implications for understanding the nongenetic modifiers of GAN phenotypes and for the optimization of gene therapy for this disease.


Asunto(s)
Acetilglucosamina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Neuropatía Axonal Gigante/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Antígenos de Neoplasias/metabolismo , Sitios de Unión , Línea Celular , Proteínas del Citoesqueleto/genética , Epigénesis Genética , Terapia Genética , Neuropatía Axonal Gigante/etiología , Neuropatía Axonal Gigante/genética , Neuropatía Axonal Gigante/terapia , Glicosilación , Histona Acetiltransferasas/metabolismo , Humanos , Hialuronoglucosaminidasa/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Modelos Biológicos , Estado Nutricional , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteostasis , Serina/metabolismo , Treonina/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
4.
Orphanet J Rare Dis ; 14(1): 27, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30709364

RESUMEN

Giant axonal neuropathy (GAN; ORPHA: 643; OMIM# 256850) is a rare, hereditary, pediatric neurodegenerative disorder associated with intracellular accumulations of intermediate filaments (IFs). GAN knockout (KO) mouse models mirror the IF dysregulation and widespread nervous system pathology seen in human GAN. Validation of therapeutic efficacy and viral vector delivery systems with these GAN KO models has provided the springboard for the development of a viral vector being delivered intrathecally in an ongoing Phase I gene therapy clinical trial for the treatment of children with GAN ( https://clinicaltrials.gov/ct2/show/NCT02362438 ). During the course of a comprehensive pathologic characterization of the GAN KO mouse, we discovered the very early and unexpected involvement of the ocular lens. Light microscopy revealed the presence of intracytoplasmic inclusion bodies within lens epithelial cells. The inclusion bodies showed strong immunohistochemical positivity for glial fibrillary acidic protein (GFAP). We confirmed that intracytoplasmic inclusion bodies are also present within lens epithelial cells in human GAN. These IF inclusion bodies in lens epithelial cells are unique to GAN. Similar IF inclusion bodies in lens epithelial cells have not been reported previously in experimental animal models or human diseases. Since current paradigms in drug discovery and drug repurposing for IF-associated disorders are often hindered by lack of validated targets, our findings suggest that lens epithelial cells in the GAN KO mouse may provide a potential target, in vivo and in vitro, for evaluating drug efficacy and alternative therapeutic approaches in promoting the clearance of IF inclusions in GAN and other diseases characterized by intracellular IF accumulations.


Asunto(s)
Neuropatía Axonal Gigante/patología , Cristalino/patología , Animales , Proteínas del Citoesqueleto/metabolismo , Neuropatía Axonal Gigante/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Filamentos Intermedios/metabolismo , Cristalino/metabolismo , Ratones , Ratones Noqueados
5.
Mol Biol Cell ; 27(25): 3980-3990, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27798231

RESUMEN

Alexander disease (AxD) is a primary genetic disorder of astrocytes caused by dominant mutations in the gene encoding the intermediate filament (IF) protein GFAP. This disease is characterized by excessive accumulation of GFAP, known as Rosenthal fibers, within astrocytes. Abnormal GFAP aggregation also occurs in giant axon neuropathy (GAN), which is caused by recessive mutations in the gene encoding gigaxonin. Given that one of the functions of gigaxonin is to facilitate proteasomal degradation of several IF proteins, we sought to determine whether gigaxonin is involved in the degradation of GFAP. Using a lentiviral transduction system, we demonstrated that gigaxonin levels influence the degradation of GFAP in primary astrocytes and in cell lines that express this IF protein. Gigaxonin was similarly involved in the degradation of some but not all AxD-associated GFAP mutants. In addition, gigaxonin directly bound to GFAP, and inhibition of proteasome reversed the clearance of GFAP in cells achieved by overexpressing gigaxonin. These studies identify gigaxonin as an important factor that targets GFAP for degradation through the proteasome pathway. Our findings provide a critical foundation for future studies aimed at reducing or reversing pathological accumulation of GFAP as a potential therapeutic strategy for AxD and related diseases.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Enfermedad de Alexander/metabolismo , Astrocitos/metabolismo , Astrocitos/fisiología , Células Cultivadas , Proteínas del Citoesqueleto/genética , Neuropatía Axonal Gigante/genética , Neuropatía Axonal Gigante/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Humanos , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis
6.
PLoS One ; 10(10): e0140157, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26460568

RESUMEN

Patients with giant axonal neuropathy (GAN) show progressive loss of motor and sensory function starting in childhood and typically live for less than 30 years. GAN is caused by autosomal recessive mutations leading to low levels of gigaxonin (GIG), a ubiquitously-expressed BTB/Kelch cytoplasmic protein believed to be an E3 ligase substrate adaptor. GAN pathology is characterized by aggregates of intermediate filaments (IFs) in multiple tissues. To delineate the molecular pathway between GIG deficiency and IF pathology, we undertook a proteomic screen to identify the normal binding partners of GIG. Prominent among them were several classes of IFs, including the neurofilament subunits whose accumulation leads to the axonal swellings for which GAN is named. We showed these interactions were dependent on the Kelch domain of GIG. Furthermore, we identified the E3 ligase MYCBP2 and the heat shock proteins HSP90AA1/AB1 as interactors with the BTB domain that may result in the ubiquitination and subsequent degradation of intermediate filaments. Our open-ended proteomic screen provides support to GIG's role as an adaptor protein, linking IF proteins through its Kelch domain to the ubiquitin pathway proteins via its BTB domain, and points to future approaches for reversing the phenotype in human patients.


Asunto(s)
Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Neuropatía Axonal Gigante/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Células HEK293 , Humanos , Unión Proteica , Mapas de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteómica , Relación Estructura-Actividad
7.
Acta Neuropathol Commun ; 2: 47, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24758703

RESUMEN

BACKGROUND: The BTB-KELCH protein Gigaxonin plays key roles in sustaining neuron survival and cytoskeleton architecture. Indeed, recessive mutations in the Gigaxonin-encoding gene cause Giant Axonal Neuropathy (GAN), a severe neurodegenerative disorder characterized by a wide disorganization of the Intermediate Filament network. Growing evidences suggest that GAN is a continuum with the peripheral neuropathy Charcot-Marie-Tooth diseases type 2 (CMT2). Sharing similar sensory-motor alterations and aggregation of Neurofilaments, few reports have revealed that GAN and some CMT2 forms can be misdiagnosed on clinical and histopathological examination. The goal of this study is to propose a new differential diagnostic test for GAN/CMT2. Moreover, we aim at identifying the mechanisms causing the loss-of-function of Gigaxonin, which has been proposed to bind CUL3 and substrates as part of an E3 ligase complex. RESULTS: We establish that determining Gigaxonin level constitutes a very valuable diagnostic test in discriminating new GAN cases from clinically related inherited neuropathies. Indeed, in a set of seven new families presenting a neuropathy resembling GAN/CMT2, only five exhibiting a reduced Gigaxonin abundance have been subsequently genetically linked to GAN. Generating the homology modeling of Gigaxonin, we suggest that disease mutations would lead to a range of defects in Gigaxonin stability, impairing its homodimerization, BTB or KELCH domain folding, or CUL3 and substrate binding. We further demonstrate that regardless of the mutations or the severity of the disease, Gigaxonin abundance is severely reduced in all GAN patients due to both mRNA and protein instability mechanisms. CONCLUSIONS: In this study, we developed a new penetrant and specific test to diagnose GAN among a set of individuals exhibiting CMT2 of unknown etiology to suggest that the prevalence of GAN is probably under-evaluated among peripheral neuropathies. We propose to use this new test in concert with the clinical examination and prior to the systematic screening of GAN mutations that has shown strong limitations for large deletions. Combining the generation of the structural modeling of Gigaxonin to an analysis of Gigaxonin transcripts and proteins in patients, we provide the first evidences of the instability of this E3 ligase adaptor in disease.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Neuropatía Axonal Gigante/genética , Neuropatía Axonal Gigante/metabolismo , Mutación/genética , Adulto , Animales , Células COS , Niño , Chlorocebus aethiops , Análisis Mutacional de ADN , Conjuntos de Datos como Asunto , Femenino , Regulación de la Expresión Génica/genética , Neuropatía Axonal Gigante/patología , Humanos , Masculino , Modelos Moleculares , Fenotipo , Transfección , Adulto Joven
8.
Hum Gene Ther ; 24(2): 209-19, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23316953

RESUMEN

Giant axonal neuropathy (GAN) is caused by loss of function of the gigaxonin protein. On a cellular level GAN is characterized by intermediate filament (IF) aggregation, leading to a progressive and fatal peripheral neuropathy in humans. This study sought to determine if re-introduction of the GAN gene into GAN-deficient cells and mice would restore proper cytoskeleton IF homeostasis. Treatment of primary skin fibroblast cultures from three different GAN patients with an adeno-associated virus type 2 (AAV2) vector containing a normal human GAN transgene significantly reduced the number of cells displaying vimentin IF aggregates. A proteomic analysis of these treated cells was also performed, wherein the abundance of 32 of 780 identified proteins significantly changed in response to gigaxonin gene transfer. While 29 of these responding proteins have not been directly described in association with gigaxonin, three were previously identified as being disregulated in GAN and were now shifted toward normal levels. To assess the potential application of this approach in vivo and eventually in humans, GAN mice received an intracisternal injection of an AAV9/GAN vector to globally deliver the GAN gene to the brainstem and spinal cord. The treated mice showed a nearly complete clearance of peripherin IF accumulations at 3 weeks post-injection. These studies demonstrate that gigaxonin gene transfer can reverse the cellular IF aggregate pathology associated with GAN.


Asunto(s)
Proteínas del Citoesqueleto/administración & dosificación , Citoesqueleto/metabolismo , Fibroblastos/patología , Neuropatía Axonal Gigante/terapia , Animales , Células Cultivadas , Proteínas del Citoesqueleto/genética , Citoesqueleto/patología , Dependovirus/genética , Dependovirus/metabolismo , Fibroblastos/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Neuropatía Axonal Gigante/metabolismo , Neuropatía Axonal Gigante/patología , Células HEK293 , Homeostasis , Humanos , Ratones , Ratones Noqueados , Mutación Missense , Cultivo Primario de Células , Proteoma/análisis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transfección , Vimentina/metabolismo
9.
Muscle Nerve ; 46(2): 246-56, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22806374

RESUMEN

INTRODUCTION: Giant axonal neuropathy (GAN) is a progressive hereditary disease that affects the peripheral and central nervous systems. It is characterized morphologically by aggregates of intermediate filaments in different tissues. Mutations have been reported in the gene that codes for gigaxonin. Nevertheless, the underlying molecular mechanism remains obscure. METHODS: Cell lines from 4 GAN patients and 4 controls were analyzed by iTRAQ. RESULTS: Among the dysregulated proteins were ribosomal protein L29, ribosomal protein L37, galectin-1, glia-derived nexin, and aminopeptidase N. Also, nuclear proteins linked to formin-binding proteins were found to be dysregulated. Although the major role of gigaxonin is reported to be degradation of cytoskeleton-associated proteins, the amount of 76 structural cytoskeletal proteins was unaltered. CONCLUSIONS: Several of the dysregulated proteins play a role in cytoskeletal reorganization. Based on these findings, we speculate that disturbed cytoskeletal regulation is responsible for formation of aggregates of intermediate filaments.


Asunto(s)
Axones/metabolismo , Neuropatía Axonal Gigante/metabolismo , Antígenos CD13/metabolismo , Femenino , Fibroblastos/metabolismo , Galectina 1/metabolismo , Humanos , Masculino , Proteómica , Proteínas Ribosómicas/metabolismo , Serpina E2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...