Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
RNA ; 28(3): 303-319, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34893560

RESUMEN

Spinal muscular atrophy (SMA) is a devastating neurodegenerative disease caused by reduced amounts of the ubiquitously expressed Survival of Motor Neuron (SMN) protein. In agreement with its crucial role in the biogenesis of spliceosomal snRNPs, SMN-deficiency is correlated to numerous splicing alterations in patient cells and various tissues of SMA mouse models. Among the snRNPs whose assembly is impacted by SMN-deficiency, those involved in the minor spliceosome are particularly affected. Importantly, splicing of several, but not all U12-dependent introns has been shown to be affected in different SMA models. Here, we have investigated the molecular determinants of this differential splicing in spinal cords from SMA mice. We show that the branchpoint sequence (BPS) is a key element controlling splicing efficiency of minor introns. Unexpectedly, splicing of several minor introns with suboptimal BPS is not affected in SMA mice. Using in vitro splicing experiments and oligonucleotides targeting minor or major snRNAs, we show for the first time that splicing of these introns involves both the minor and major machineries. Our results strongly suggest that splicing of a subset of minor introns is not affected in SMA mice because components of the major spliceosome compensate for the loss of minor splicing activity.


Asunto(s)
Atrofia Muscular Espinal/genética , Empalme del ARN , Empalmosomas/metabolismo , Animales , Células HeLa , Humanos , Intrones , Ratones , Atrofia Muscular Espinal/metabolismo , Sitios de Empalme de ARN , Ribonucleoproteínas Nucleares Pequeñas/metabolismo
2.
Mol Ther ; 28(8): 1887-1901, 2020 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-32470325

RESUMEN

Spinal muscular atrophy (SMA) is a neuromuscular disease mainly caused by mutations or deletions in the survival of motor neuron 1 (SMN1) gene and characterized by the degeneration of motor neurons and progressive muscle weakness. A viable therapeutic approach for SMA patients is a gene replacement strategy that restores functional SMN expression using adeno-associated virus serotype 9 (AAV9) vectors. Currently, systemic or intra-cerebrospinal fluid (CSF) delivery of AAV9-SMN is being explored in clinical trials. In this study, we show that the postnatal delivery of an AAV9 that expresses SMN under the control of the neuron-specific promoter synapsin selectively targets neurons without inducing re-expression in the peripheral organs of SMA mice. However, this approach is less efficient in restoring the survival and neuromuscular functions of SMA mice than the systemic or intra-CSF delivery of an AAV9 in which SMN is placed under the control of a ubiquitous promoter. This study suggests that further efforts are needed to understand the extent to which SMN is required in neurons and peripheral organs for a successful therapeutic effect.


Asunto(s)
Dependovirus/genética , Vectores Genéticos/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/virología , Atrofia Muscular Espinal/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Animales , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Locomoción , Ratones , Atrofia Muscular Espinal/tratamiento farmacológico , Fenotipo , Pronóstico , Regiones Promotoras Genéticas , Médula Espinal/metabolismo , Médula Espinal/patología , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Transducción Genética , Resultado del Tratamiento
3.
Hum Mol Genet ; 27(6): 954-968, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29325092

RESUMEN

Sandhoff disease (SD) is a rare inherited disorder caused by a deficiency of ß-hexosaminidase activity which is fatal because no effective treatment is available. A mouse model of Hexb deficiency reproduces the key pathognomonic features of SD patients with severe ubiquitous lysosomal dysfunction, GM2 accumulation, neuroinflammation and neurodegeneration, culminating in death at 4 months. Here, we show that a single intravenous neonatal administration of a self-complementary adeno-associated virus 9 vector (scAAV9) expressing the Hexb cDNA in SD mice is safe and sufficient to prevent disease development. Importantly, we demonstrate for the first time that this treatment results in a normal lifespan (over 700 days) and normalizes motor function assessed by a battery of behavioral tests, with scAAV9-treated SD mice being indistinguishable from wild-type littermates. Biochemical analyses in multiple tissues showed a significant increase in hexosaminidase A activity, which reached 10-15% of normal levels. AAV9 treatment was sufficient to prevent GM2 and GA2 storage almost completely in the cerebrum (less so in the cerebellum), as well as thalamic reactive gliosis and thalamocortical neuron loss in treated Hexb-/- mice. In summary, this study demonstrated a widespread protective effect throughout the entire CNS after a single intravenous administration of the scAAV9-Hexb vector to neonatal SD mice.


Asunto(s)
Hexosaminidasa B/farmacología , Enfermedad de Sandhoff/tratamiento farmacológico , Enfermedad de Sandhoff/patología , Administración Intravenosa , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Femenino , Gangliósido G(M2)/metabolismo , Gangliósidos/metabolismo , Hexosaminidasa B/genética , Hexosaminidasa B/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad de Sandhoff/metabolismo
4.
Nucleic Acids Res ; 45(9): 5399-5413, 2017 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-28115638

RESUMEN

Selenoprotein synthesis requires the co-translational recoding of a UGASec codon. This process involves an RNA structural element, called Selenocysteine Insertion Sequence (SECIS) and the SECIS binding protein 2 (SBP2). Several selenoprotein mRNAs undergo unusual cap hypermethylation by the trimethylguanosine synthase 1 (Tgs1), which is recruited by the ubiquitous Survival of MotoNeurons (SMN) protein. SMN, the protein involved in spinal muscular atrophy, is part of a chaperone complex that collaborates with the methylosome for RNP assembly. Here, we analyze the role of individual SMN and methylosome components in selenoprotein mRNP assembly and translation. We show that SBP2 interacts directly with four proteins of the SMN complex and the methylosome core proteins. Nevertheless, SBP2 is not a methylation substrate of the methylosome. We found that both SMN and methylosome complexes are required for efficient translation of the selenoprotein GPx1 in vivo. We establish that the steady-state level of several selenoprotein mRNAs, major regulators of oxidative stress damage in neurons, is specifically reduced in the spinal cord of SMN-deficient mice and that cap hypermethylation of GPx1 mRNA is affected. Altogether we identified a new function of the SMN complex and the methylosome in selenoprotein mRNP assembly and expression.


Asunto(s)
Biosíntesis de Proteínas , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/metabolismo , Proteínas del Complejo SMN/metabolismo , Selenoproteínas/metabolismo , Glutatión Peroxidasa , Células HEK293 , Células HeLa , Humanos , Metilación , Modelos Biológicos , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patología , Unión Proteica , Médula Espinal/metabolismo , Glutatión Peroxidasa GPX1
6.
Mol Ther ; 25(9): 2038-2052, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28663100

RESUMEN

One of the most promising therapeutic approaches for familial amyotrophic lateral sclerosis linked to superoxide dismutase 1 (SOD1) is the suppression of toxic mutant SOD1 in the affected tissues. Here, we report an innovative molecular strategy for inducing substantial, widespread, and sustained reduction of mutant human SOD1 (hSOD1) levels throughout the body of SOD1G93A mice, leading to therapeutic effects in animals. Adeno-associated virus serotype rh10 vectors (AAV10) were used to mediate exon skipping of the hSOD1 pre-mRNA by expression of exon-2-targeted antisense sequences embedded in a modified U7 small-nuclear RNA (AAV10-U7-hSOD). Skipping of hSOD1 exon 2 led to the generation of a premature termination codon, inducing production of a deleted transcript that was subsequently degraded by the activation of nonsense-mediated decay. Combined intravenous and intracerebroventricular delivery of AAV10-U7-hSOD increased the survival of SOD1G93A mice injected either at birth or at 50 days of age (by 92% and 58%, respectively) and prevented weight loss and the decline of neuromuscular function. This study reports the effectiveness of an exon-skipping approach in SOD1-ALS mice, supporting the translation of this technology to the treatment of this as yet incurable disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Superóxido Dismutasa-1/genética , Edad de Inicio , Esclerosis Amiotrófica Lateral/mortalidad , Esclerosis Amiotrófica Lateral/fisiopatología , Esclerosis Amiotrófica Lateral/terapia , Animales , Modelos Animales de Enfermedad , Exones , Orden Génico , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Humanos , Ratones , Ratones Transgénicos , Actividad Motora/genética , Oligonucleótidos Antisentido , Sitios de Empalme de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recuperación de la Función , Superóxido Dismutasa-1/metabolismo , Tasa de Supervivencia , Transducción Genética
7.
Neurobiol Dis ; 99: 1-11, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27974239

RESUMEN

Rett syndrome (RTT) is a severe X-linked neurodevelopmental disorder that is primarily caused by mutations in the methyl CpG binding protein 2 gene (MECP2). RTT is the second most prevalent cause of intellectual disability in girls and there is currently no cure for the disease. The finding that the deficits caused by the loss of Mecp2 are reversible in the mouse has bolstered interest in gene therapy as a cure for RTT. In order to assess the feasibility of gene therapy in a RTT mouse model, and in keeping with translational goals, we investigated the efficacy of a self-complementary AAV9 vector expressing a codon-optimized version of Mecp2 (AAV9-MCO) delivered via a systemic approach in early symptomatic Mecp2-deficient (KO) mice. Our results show that AAV9-MCO administered at a dose of 2×1011 viral genome (vg)/mouse was able to significantly increase survival and weight gain, and delay the occurrence of behavioral deficits. Apneas, which are one of the core RTT breathing deficits, were significantly decreased to WT levels in Mecp2 KO mice after AAV9-MCO administration. Semi-quantitative analysis showed that AAV9-MCO administration in Mecp2 KO mice resulted in 10 to 20% Mecp2 immunopositive cells compared to WT animals, with the highest Mecp2 expression found in midbrain regions known to regulate cardio-respiratory functions. In addition, we also found a cell autonomous increase in tyrosine hydroxylase levels in the A1C1 and A2C2 catecholaminergic Mecp2+ neurons in treated Mecp2 KO mice, which may partly explain the beneficial effect of AAV9-MCO administration on apneas occurrence.


Asunto(s)
Terapia Genética , Proteína 2 de Unión a Metil-CpG/administración & dosificación , Síndrome de Rett/terapia , Aminas , Animales , Apnea/metabolismo , Apnea/patología , Apnea/prevención & control , Codón , Ácidos Ciclohexanocarboxílicos , Dependovirus , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Gabapentina , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Mesencéfalo/metabolismo , Mesencéfalo/patología , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Respiración , Síndrome de Rett/metabolismo , Síndrome de Rett/patología , Análisis de Supervivencia , Tirosina 3-Monooxigenasa/metabolismo , Aumento de Peso , Ácido gamma-Aminobutírico
8.
Acta Neuropathol ; 132(2): 257-276, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27138984

RESUMEN

Endosomal-autophagic-lysosomal (EAL) dysfunction is an early and prominent neuropathological feature of Alzheimers's disease, yet the exact molecular mechanisms contributing to this pathology remain undefined. By combined biochemical, immunohistochemical and ultrastructural approaches, we demonstrate a link between EAL pathology and the intraneuronal accumulation of the ß-secretase-derived ßAPP fragment (C99) in two in vivo models, 3xTgAD mice and adeno-associated viral-mediated C99-infected mice. We present a pathological loop in which the accumulation of C99 is both the effect and causality of impaired lysosomal-autophagic function. The deleterious effect of C99 was found to be linked to its aggregation within EAL-vesicle membranes leading to disrupted lysosomal proteolysis and autophagic impairment. This effect was Aß independent and was even exacerbated when γ-secretase was pharmacologically inhibited. No effect was observed in inhibitor-treated wild-type animals suggesting that lysosomal dysfunction was indeed directly linked to C99 accumulation. In some brain areas, strong C99 expression also led to inflammatory responses and synaptic dysfunction. Taken together, this work demonstrates a toxic effect of C99 which could underlie some of the early-stage anatomical hallmarks of Alzheimer's disease pathology. Our work also proposes molecular mechanisms likely explaining some of the unfavorable side-effects associated with γ-secretase inhibitor-directed therapies.


Asunto(s)
Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/patología , Neuronas/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Autofagia/fisiología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Endosomas/metabolismo , Lisosomas/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/patología
9.
Expert Opin Emerg Drugs ; 20(3): 353-6, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25920617

RESUMEN

Spinal muscular atrophy (SMA), one of the most frequent and devastating genetic disorders causing neuromuscular degeneration, has reached the forefront of clinical translation. The quite unique genetic situation of SMA patients, who lack functional SMN1 but carry the misspliced SMN2 copy gene, creates the possibility of correcting SMN2 splicing by antisense oligonucleotides or drugs. Both strategies showed impressive results in pre-clinical trials and are now in Phase II-III clinical trials. SMN gene therapy approaches using AAV9-SMN vectors are also highly promising and have entered a Phase I clinical trial. However, careful analysis of SMA animal models and patients has revealed some limitations that need to be taken very seriously, including: i) a limited time-window for successful therapy delivery, making neonatal screening of SMA mandatory; ii) multi-organ impairment, requiring systemic delivery of therapies; and iii) a potential need for combined therapies that both increase SMN levels and target pathways that preserve/rescue motor neuron function over the lifespan. Meeting these challenges will likely be crucial to cure SMA, instead of only ameliorating symptoms, particularly in its most severe form. This review discusses therapies currently in clinical trials, the hopes for SMA therapy, and the potential limitations of these new approaches.


Asunto(s)
Terapia Genética/métodos , Atrofia Muscular Espinal/terapia , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos , Humanos , Recién Nacido , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/fisiopatología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/genética
10.
Nucleic Acids Res ; 41(2): 1255-72, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23221635

RESUMEN

Spinal muscular atrophy is a severe motor neuron disease caused by reduced levels of the ubiquitous Survival of MotoNeurons (SMN) protein. SMN is part of a complex that is essential for spliceosomal UsnRNP biogenesis. Signal recognition particle (SRP) is a ribonucleoprotein particle crucial for co-translational targeting of secretory and membrane proteins to the endoplasmic reticulum. SRP biogenesis is a nucleo-cytoplasmic multistep process in which the protein components, except SRP54, assemble with 7S RNA in the nucleolus. Then, SRP54 is incorporated after export of the pre-particle into the cytoplasm. The assembly factors necessary for SRP biogenesis remain to be identified. Here, we show that 7S RNA binds to purified SMN complexes in vitro and that SMN complexes associate with SRP in cellular extracts. We identified the RNA determinants required. Moreover, we report a specific reduction of 7S RNA levels in the spinal cord of SMN-deficient mice, and in a Schizosaccharomyces pombe strain carrying a temperature-degron allele of SMN. Additionally, microinjected antibodies directed against SMN or Gemin2 interfere with the association of SRP54 with 7S RNA in Xenopus laevis oocytes. Our data show that reduced levels of the SMN protein lead to defect in SRP steady-state level and describe the SMN complex as the first identified cellular factor required for SRP biogenesis.


Asunto(s)
ARN Citoplasmático Pequeño/metabolismo , Proteínas del Complejo SMN/metabolismo , Partícula de Reconocimiento de Señal/metabolismo , Alelos , Animales , Anticuerpos/farmacología , Secuencia de Bases , Citoplasma/metabolismo , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Atrofia Muscular Espinal/metabolismo , Mutación , ARN Citoplasmático Pequeño/química , ARN Nuclear Pequeño/metabolismo , Proteínas del Complejo SMN/antagonistas & inhibidores , Proteínas del Complejo SMN/inmunología , Schizosaccharomyces/genética , Partícula de Reconocimiento de Señal/química , Médula Espinal/metabolismo , Xenopus laevis
11.
Acta Neuropathol ; 128(5): 705-22, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24859968

RESUMEN

There is still no treatment for polyglutamine disorders, but clearance of mutant proteins might represent a potential therapeutic strategy. Autophagy, the major pathway for organelle and protein turnover, has been implicated in these diseases. To determine whether the autophagy/lysosome system contributes to the pathogenesis of spinocerebellar ataxia type 7 (SCA7), caused by expansion of a polyglutamine tract in the ataxin-7 protein, we looked for biochemical, histological and transcriptomic abnormalities in components of the autophagy/lysosome pathway in a knock-in mouse model of the disease, postmortem brain and peripheral blood mononuclear cells (PBMC) from patients. In the mouse model, mutant ataxin-7 accumulated in inclusions immunoreactive for the autophagy-associated proteins mTOR, beclin-1, p62 and ubiquitin. Atypical accumulations of the autophagosome/lysosome markers LC3, LAMP-1, LAMP2 and cathepsin-D were also found in the cerebellum of the SCA7 knock-in mice. In patients, abnormal accumulations of autophagy markers were detected in the cerebellum and cerebral cortex of patients, but not in the striatum that is spared in SCA7, suggesting that autophagy might be impaired by the selective accumulation of mutant ataxin-7. In vitro studies demonstrated that the autophagic flux was impaired in cells overexpressing full-length mutant ataxin-7. Interestingly, the expression of the early autophagy-associated gene ATG12 was increased in PBMC from SCA7 patients in correlation with disease severity. These results provide evidence that the autophagy/lysosome pathway is impaired in neurons undergoing degeneration in SCA7. Autophagy/lysosome-associated molecules might, therefore, be useful markers for monitoring the effects of potential therapeutic approaches using modulators of autophagy in SCA7 and other autophagy/lysosome-associated neurodegenerative disorders.


Asunto(s)
Autofagia/fisiología , Encéfalo/patología , Lisosomas/metabolismo , Lisosomas/patología , Proteínas del Tejido Nervioso/metabolismo , Ataxias Espinocerebelosas/patología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Ataxina-7 , Beclina-1 , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Estudios de Casos y Controles , Línea Celular Transformada , Femenino , Regulación de la Expresión Génica/genética , Humanos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Lisosomas/ultraestructura , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/ultraestructura , Proteínas de Unión a Fosfato , Transducción de Señal/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ataxias Espinocerebelosas/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Repeticiones de Trinucleótidos/genética
12.
Brain ; 136(Pt 6): 1732-45, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23518714

RESUMEN

We showed previously, in a cell model of spinocerebellar ataxia 7, that interferon beta induces the expression of PML protein and the formation of PML protein nuclear bodies that degrade mutant ataxin 7, suggesting that the cytokine, used to treat multiple sclerosis, might have therapeutic value in spinocerebellar ataxia 7. We now show that interferon beta also induces PML-dependent clearance of ataxin 7 in a preclinical model, SCA7(266Q/5Q) knock-in mice, and improves motor function. Interestingly, the presence of mutant ataxin 7 in the mice induces itself the expression of endogenous interferon beta and its receptor. Immunohistological studies in brains from two patients with spinocerebellar ataxia 7 confirmed that these modifications are also caused by the disease in humans. Interferon beta, administered intraperitoneally three times a week in the knock-in mice, was internalized with its receptor in Purkinje and other cells and translocated to the nucleus. The treatment induced PML protein expression and the formation of PML protein nuclear bodies and decreased mutant ataxin 7 in neuronal intranuclear inclusions, the hallmark of the disease. No reactive gliosis or other signs of toxicity were observed in the brain or internal organs. The performance of the SCA7(266Q/5Q) knock-in mice was significantly improved on two behavioural tests sensitive to cerebellar function: the Locotronic® Test of locomotor function and the Beam Walking Test of balance, motor coordination and fine movements, which are affected in patients with spinocerebellar ataxia 7. In addition to motor dysfunction, SCA7(266Q/5Q) mice present abnormalities in the retina as in patients: ataxin 7-positive neuronal intranuclear inclusions that were reduced by interferon beta treatment. Finally, since neuronal death does not occur in the cerebellum of SCA7(266Q/5Q) mice, we showed in primary cell cultures expressing mutant ataxin 7 that interferon beta treatment improves Purkinje cell survival.


Asunto(s)
Interferón beta/uso terapéutico , Actividad Motora/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/fisiopatología , Adulto , Anciano , Animales , Ataxina-7 , Células Cultivadas , Niño , Técnicas de Sustitución del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Wistar , Ataxias Espinocerebelosas/tratamiento farmacológico
13.
Mol Ther ; 21(2): 282-90, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23295949

RESUMEN

We have recently demonstrated the remarkable efficiency of self-complementary (sc) AAV9 vectors for central nervous system (CNS) gene transfer following intravenous delivery in mice and larger animals. Here, we investigated whether gene delivery to motor neurons (MNs) could also be achieved via intramuscular (i.m.) scAAV9 injection and subsequent retrograde transport along the MNs axons. Unexpectedly, we found that a single injection of scAAV9 into the adult mouse gastrocnemius (GA) mediated widespread MN transduction along the whole spinal cord, without limitation to the MNs connected to the injected muscle. Spinal cord astrocytes and peripheral organs were also transduced, indicating vector spread from the injected muscle to both the CNS and the periphery through release into the blood circulation. Moreover, we showed that i.m. injection of scAAV9 vectors expressing "survival of motor neuron" (Smn) in spinal muscular atrophy (SMA) mice mediated high survival motor neuron (SMN) expression levels at both the CNS and the periphery, and increased the median lifespan from 12 days to 163 days. These findings represent to date the longest extent in survival obtained in SMA mice following i.m. viral vector gene delivery, and might generate a renewed interest in the use of i.m. adeno-associated viruses (AAV) delivery for the development of gene therapy strategies for MN diseases.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Atrofia Muscular Espinal/terapia , Médula Espinal/patología , Animales , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Vectores Genéticos , Inyecciones Intramusculares , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/patología , Médula Espinal/metabolismo , Transgenes
14.
Hum Mol Genet ; 20(4): 681-93, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21118896

RESUMEN

Spinal muscular atrophy (SMA) is the most common genetic disease leading to infant mortality. This neuromuscular disorder is caused by the loss or mutation of the telomeric copy of the 'survival of motor neuron' (Smn) gene, termed SMN1. Loss of SMN1 leads to reduced SMN protein levels, inducing degeneration of motor neurons (MN) and progressive muscle weakness and atrophy. To date, SMA remains incurable due to the lack of a method to deliver therapeutically active molecules to the spinal cord. Gene therapy, consisting of reintroducing SMN1 in MNs, is an attractive approach for SMA. Here we used postnatal day 1 systemic injection of self-complementary adeno-associated virus (scAAV9) vectors carrying a codon-optimized SMN1 sequence and a chimeric intron placed downstream of the strong phosphoglycerate kinase (PGK) promoter (SMNopti) to overexpress the human SMN protein in a mouse model of severe SMA. Survival analysis showed that this treatment rescued 100% of the mice, increasing life expectancy from 27 to over 340 days (median survival of 199 days) in mice that normally survive about 13 days. The systemic scAAV9 therapy mediated complete correction of motor function, prevented MN death and rescued the weight loss phenotype close to normal. This study reports the most efficient rescue of SMA mice to date after a single intravenous injection of an optimized SMN-encoding scAAV9, highlighting the considerable potential of this method for the treatment of human SMA.


Asunto(s)
Dependovirus/genética , Terapia Genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Humanos , Ratones , Ratones Noqueados , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/patología , Fenotipo , Médula Espinal/metabolismo , Médula Espinal/patología , Análisis de Supervivencia , Resultado del Tratamiento
15.
Ann Neurol ; 71(4): 509-19, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22522442

RESUMEN

OBJECTIVE: Distal hereditary motor neuropathies (dHMN) form a clinically and genetically heterogeneous group of disorders, characterized by muscle weakness and atrophy predominating at the distal part of the limbs, due to the progressive degeneration of motor neurons in the spinal cord. We report here a novel rare variant of dHMN with autosomal recessive inheritance in a large Jewish family originating from Morocco. The disease is characterized by a predominance of paralysis at the lower limbs and an early adulthood onset. We performed a genetic study in this family to identify and characterized the causing mutation. METHODS: Homozygosity mapping strategy and sequencing of the candidate genes were performed. Expression studies were made on patient fibroblasts. Functional experiments were performed on a cellular model of motor neuron disease. RESULTS: We mapped the disease to the 2q34-q36.1 chromosomal region and identified a homozygous splice mutation in the gene HSJ1 (DNAJB2) decreasing the expression of the 2 main isoforms HSJ1a and HSJ1b. Overexpression of both HSJ1a and HSJ1b reduced inclusion formation induced by the mutated SOD1-A4V in a neuronal cellular model. INTERPRETATION: HSJ1 is a neuronal enriched member of the HSP40/DNAJ co-chaperone family. Previous studies have shown that HSP40 proteins play a crucial role in protein aggregation and neurodegeneration in several neuronal types, in animal models and human diseases. Interestingly, this mutation causing a loss-of-function of HSJ1 is linked to a pure lower motor neuron disease, strongly suggesting that HSJ1 also plays an important and specific role in motor neurons.


Asunto(s)
Proteínas del Choque Térmico HSP40/genética , Chaperonas Moleculares/genética , Enfermedad de la Neurona Motora/genética , Enfermedad de la Neurona Motora/fisiopatología , Mutación , Adolescente , Adulto , Edad de Inicio , Secuencia de Bases , Mapeo Cromosómico , Electromiografía , Femenino , Humanos , Judíos/genética , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Marruecos , Linaje , Adulto Joven
16.
Mol Ther Methods Clin Dev ; 20: 1-17, 2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33335943

RESUMEN

Fabry disease is a rare X-linked disorder affecting α-galactosidase A, a rate-limiting enzyme in lysosomal catabolism of glycosphingolipids. Current treatments present important limitations, such as low half-life and limited distribution, which gene therapy can overcome. The aim of this work was to test a novel adeno-associated viral vector, serotype 9 (AAV9), ubiquitously expressing human α-galactosidase A to treat Fabry disease (scAAV9-PGK-GLA). The vector was preliminary tested in newborns of a Fabry disease mouse model. 5 months after treatment, α-galactosidase A activity was detectable in the analyzed tissues, including the central nervous system. Moreover, we tested the vector in adult animals of both sexes at two doses and disease stages (presymptomatic and symptomatic) by single intravenous injection. We found that the exogenous α-galactosidase A was active in peripheral tissues as well as the central nervous system and prevented glycosphingolipid accumulation in treated animals up to 5 months following injection. Antibodies against α-galactosidase A were produced in 9 out of 32 treated animals, although enzyme activity in tissues was not significantly affected. These results demonstrate that scAAV9-PGK-GLA can drive widespread and sustained expression of α-galactosidase A, cross the blood brain barrier after systemic delivery, and reduce pathological signs of the Fabry disease mouse model.

17.
Mol Ther ; 17(7): 1187-96, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19367261

RESUMEN

Therapeutic gene delivery to the whole spinal cord is a major challenge for the treatment of motor neuron (MN) diseases. Systemic administration of viral gene vectors would provide an optimal means for the long-term delivery of therapeutic molecules from blood to the spinal cord but this approach is hindered by the presence of the blood-brain barrier (BBB). Here, we describe the first successful study of MN transduction in adult animals following intravenous (i.v.) delivery of self-complementary (sc) AAV9 vectors (up to 28% in mice). Intravenous MN transduction was achieved in adults without pharmacological disruption of the BBB and transgene expression lasted at least 5 months. Importantly, this finding was successfully translated to large animals, with the demonstration of an efficient systemic scAAV9 gene delivery to the neonate and adult cat spinal cord. This new and noninvasive procedure raises the hope of whole spinal cord correction of MN diseases and may lead to the development of new gene therapy protocols in patients.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/genética , Transducción Genética/métodos , Animales , Animales Recién Nacidos , Gatos , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos/administración & dosificación , Inmunohistoquímica , Bombas de Infusión , Ratones , Ratones Endogámicos C57BL , Enfermedad de la Neurona Motora/terapia , Embarazo , Médula Espinal/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Med Sci (Paris) ; 36(2): 137-140, 2020 Feb.
Artículo en Francés | MEDLINE | ID: mdl-32129749

RESUMEN

Spinal muscular atrophy (SMA) is the most common genetic disease leading to infant mortality. This neuro-muscular disorder is caused by the loss or mutation of the telomeric copy of the 'survival of motor neuron' (Smn) gene, termed SMN1. Loss of SMN1 leads to reduced SMN protein levels, inducing degeneration of motor neurons (MN) and progressive muscle weakness and atrophy. Gene therapy, consisting of reintroducing SMN1 in the MNs, is an attractive approach for SMA. We showed the most efficient rescue of SMA mice to date after a single intravenous injection of an AAV9 expressing SMN1, highlighting the considerable potential of this method for the treatment of human SMA. Recently, a startup led by the Dr Kaspar decided to test this experimental approach in children with SMA type 1. Dr Mendell, in charge of this clinical project, showed a very significant increase of the lifespan and motor function of the patients (until 4 years) after a single injection of AAV9-SMN1 (named ZolgenSMA®) into an arm or leg vein. This gene therapy treatment obtained a marketing authorization by the FDA in May 24 and is now the first efficient therapy for neuromuscular disease.


TITLE: Amyotrophie spinale infantile - De la découverte du gène à la thérapie génique. ABSTRACT: L'amyotrophie spinale ou SMA est la maladie génétique la plus fréquente menant à la mortalité infantile. Cette maladie neuromusculaire est due à l'altération du gène SMN1. Cette anomalie génétique provoque la réduction des taux de protéine Smn, induisant la dégénérescence des neurones moteurs, la faiblesse et l'atrophie musculaire. La thérapie génique, consistant à réintroduire le gène SMN1 normal dans les motoneurones constitue une thérapie de choix pour la SMA. Nous avons montré l'efficacité sans précédent de cette approche chez la souris modèle de SMA après une simple injection intraveineuse d'un AAV9 exprimant SMN1. Une jeune société de biotechnologie, dirigée par le Dr Kaspar, a testé cette approche expérimentale chez de jeunes patients atteints de SMA type 1. Le Dr Mendell, en charge de ce projet clinique, a montré une augmentation significative de la survie et des fonctions motrices des patients (jusqu'à 4 ans) après une seule injection de l'AAV9-SMN (appelé ZolgenSMA) dans la veine du bras ou de la jambe. Cette thérapie, qui a obtenu l'AMM par la FDA le 24 mai 2019, est actuellement la première thérapie génique efficace dans les maladies neuromusculaires.


Asunto(s)
Estudios de Asociación Genética , Terapia Genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , Animales , Niño , Modelos Animales de Enfermedad , Estudios de Asociación Genética/métodos , Estudios de Asociación Genética/tendencias , Terapia Genética/métodos , Terapia Genética/tendencias , Humanos , Ratones , Neuronas Motoras/fisiología , Atrofia Muscular Espinal/diagnóstico , Mutación , Atrofias Musculares Espinales de la Infancia/diagnóstico , Atrofias Musculares Espinales de la Infancia/genética , Atrofias Musculares Espinales de la Infancia/terapia
19.
Transl Neurodegener ; 8: 35, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31827783

RESUMEN

BACKGROUND: We recently demonstrated an endolysosomal accumulation of the ß-secretase-derived APP C-terminal fragment (CTF) C99 in brains of Alzheimer disease (AD) mouse models. Moreover, we showed that the treatment with the γ-secretase inhibitor (D6) led to further increased endolysosomal APP-CTF levels, but also revealed extracellular APP-CTF-associated immunostaining. We here hypothesized that this latter staining could reflect extracellular vesicle (EV)-associated APP-CTFs and aimed to characterize these γ-secretase inhibitor-induced APP-CTFs. METHODS: EVs were purified from cell media or mouse brains from vehicle- or D6-treated C99 or APPswedish expressing cells/mice and analyzed for APP-CTFs by immunoblot. Combined pharmacological, immunological and genetic approaches (presenilin invalidation and C99 dimerization mutants (GXXXG)) were used to characterize vesicle-containing APP-CTFs. Subcellular APP-CTF localization was determined by immunocytochemistry. RESULTS: Purified EVs from both AD cell or mouse models were enriched in APP-CTFs as compared to EVs from control cells/brains. Surprisingly, EVs from D6-treated cells not only displayed increased C99 and C99-derived C83 levels but also higher molecular weight (HMW) APP-CTF-immunoreactivities that were hardly detectable in whole cell extracts. Accordingly, the intracellular levels of HMW APP-CTFs were amplified by the exosomal inhibitor GW4869. By combined pharmacological, immunological and genetic approaches, we established that these HMW APP-CTFs correspond to oligomeric APP-CTFs composed of C99 and/or C83. Immunocytochemical analysis showed that monomers were localized mainly to the trans-Golgi network, whereas oligomers were confined to endosomes and lysosomes, thus providing an anatomical support for the selective recovery of HMW APP-CTFs in EVs. The D6-induced APP-CTF oligomerization and subcellular mislocalization was indeed due to γ-secretase blockade, since it similarly occurred in presenilin-deficient fibroblasts. Further, our data proposed that besides favoring APP-CTF oligomerization by preventing C99 proteolysis, γ-secretase inhibiton also led to a defective SorLA-mediated retrograde transport of HMW APP-CTFs from endosomal compartments to the TGN. CONCLUSIONS: This is the first study to demonstrate the presence of oligomeric APP-CTFs in AD mouse models, the levels of which are selectively enriched in endolysosomal compartments including exosomes and amplified by γ-secretase inhibition. Future studies should evaluate the putative contribution of these exosome-associated APP-CTFs in AD onset, progression and spreading.

20.
Biol Psychiatry ; 83(5): 416-427, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28587718

RESUMEN

BACKGROUND: Mitophagy and mitochondrial dynamics alterations are two major hallmarks of neurodegenerative diseases. Dysfunctional mitochondria accumulate in Alzheimer's disease-affected brains by yet unexplained mechanisms. METHODS: We combined cell biology, molecular biology, and pharmacological approaches to unravel a novel molecular pathway by which presenilins control phosphatase and tensin homolog-induced kinase 1 (Pink-1) expression and transcription. In vivo approaches were carried out on various transgenic and knockout animals as well as in adeno-associated virus-infected mice. Functional readout and mitochondrial physiology (mitochondrial potential) were assessed by combined procedures including flow cytometry, live imaging analysis, and immunohistochemistry. RESULTS: We show that presenilins 1 and 2 trigger opposite effects on promoter transactivation, messenger RNA, and protein expression of Pink-1. This control is linked to γ-secretase activity and ß-amyloid precursor protein but is independent of phosphatase and tensin homolog. We show that amyloid precursor protein intracellular domain (AICD) accounts for presenilin-dependent phenotype and upregulates Pink-1 transactivation in cells as well as in vivo in a Forkhead box O3a-dependent manner. Interestingly, the modulation of γ-secretase activity or AICD expression affects Pink-1-related control of mitophagy and mitochondrial dynamics. Finally, we show that parkin acts upstream of presenilins to control Pink-1 promoter transactivation and protein expression. CONCLUSIONS: Overall, we delineate a molecular cascade presenilins-AICD-Forkhead box O3a linking parkin to Pink-1. Our study demonstrates AICD-mediated Pink-1-dependent control of mitochondrial physiology by presenilins. Furthermore, it unravels a parkin-Pink-1 feedback loop controlling mitochondrial physiology that could be disrupted in neurodegenerative conditions.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Proteína Forkhead Box O3/metabolismo , Hipocampo/metabolismo , Mitocondrias/metabolismo , Presenilinas/metabolismo , Proteínas Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Embrión de Mamíferos , Fibroblastos , Células HEK293 , Humanos , Espacio Intracelular/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA