Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Intervalo de año de publicación
1.
Pharmaceutics ; 16(6)2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38931878

RESUMEN

Rett syndrome (RTT) is a rare neurodevelopmental disorder caused by mutation in the X-linked gene methyl-CpG-binding protein 2 (Mecp2), a ubiquitously expressed transcriptional regulator. RTT results in mental retardation and developmental regression that affects approximately 1 in 10,000 females. Currently, there is no curative treatment for RTT. Thus, it is crucial to develop new therapeutic approaches for children suffering from RTT. Several studies suggested that RTT is linked with defects in cholesterol homeostasis, but for the first time, therapeutic evaluation is carried out by modulating this pathway. Moreover, AAV-based CYP46A1 overexpression, the enzyme involved in cholesterol pathway, has been demonstrated to be efficient in several neurodegenerative diseases. Based on these data, we strongly believe that CYP46A1 could be a relevant therapeutic target for RTT. Herein, we evaluated the effects of intravenous AAVPHP.eB-hCYP46A1-HA delivery in male and female Mecp2-deficient mice. The applied AAVPHP.eB-hCYP46A1 transduced essential neurons of the central nervous system (CNS). CYP46A1 overexpression alleviates behavioral alterations in both male and female Mecp2 knockout mice and extends the lifespan in Mecp2-deficient males. Several parameters related to cholesterol pathway are improved and correction of mitochondrial activity is demonstrated in treated mice, which highlighted the clear therapeutic benefit of CYP46A1 through the neuroprotection effect. IV delivery of AAVPHP.eB-CYP46A1 is perfectly well tolerated with no inflammation observed in the CNS of the treated mice. Altogether, our results strongly suggest that CYP46A1 is a relevant target and overexpression could alleviate the phenotype of Rett patients.

2.
Hum Gene Ther ; 32(7-8): 349-374, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33167739

RESUMEN

For more than 10 years, gene therapy for neurological diseases has experienced intensive research growth and more recently therapeutic interventions for multiple indications. Beneficial results in several phase 1/2 clinical studies, together with improved vector technology have advanced gene therapy for the central nervous system (CNS) in a new era of development. Although most initial strategies have focused on orphan genetic diseases, such as lysosomal storage diseases, more complex and widespread conditions like Alzheimer's disease, Parkinson's disease, epilepsy, or chronic pain are increasingly targeted for gene therapy. Increasing numbers of applications and patients to be treated will require improvement and simplification of gene therapy protocols to make them accessible to the largest number of affected people. Although vectors and manufacturing are a major field of academic research and industrial development, there is a growing need to improve, standardize, and simplify delivery methods. Delivery is the major issue for CNS therapies in general, and particularly for gene therapy. The blood-brain barrier restricts the passage of vectors; strategies to bypass this obstacle are a central focus of research. In this study, we present the different ways that can be used to deliver gene therapy products to the CNS. We focus on results obtained in large animals that have allowed the transfer of protocols to human patients and have resulted in the generation of clinical data. We discuss the different routes of administration, their advantages, and their limitations. We describe techniques, equipment, and protocols and how they should be selected for safe delivery and improved efficiency for the next generation of gene therapy trials for CNS diseases.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Técnicas de Transferencia de Gen , Animales , Sistema Nervioso Central , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/terapia , Terapia Genética , Vectores Genéticos/genética , Humanos
3.
BMC Res Notes ; 13(1): 210, 2020 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-32276655

RESUMEN

OBJECTIVE: Compromised brain cholesterol turnover and altered regulation of brain cholesterol metabolism have been allied with some neurodegenerative diseases, including Huntington's disease (HD). Following our previous studies in HD, in this study we aim to investigate in vitro in a neuroblastoma cellular model of HD, the effect of CYP46A1 overexpression, an essential enzyme in cholesterol metabolism, on huntingtin aggregation and levels. RESULTS: We found that CYP46A1 reduces the quantity and size of mutant huntingtin aggregates in cells, as well as the levels of mutant huntingtin protein. Additionally, our results suggest that the observed beneficial effects of CYP46A1 in HD cells are linked to the activation of autophagy. Taken together, our results further demonstrate that CYP46A1 is a pertinent target to counteract HD progression.


Asunto(s)
Autofagia , Colesterol 24-Hidroxilasa/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Neuroblastoma , Animales , Línea Celular Tumoral , Células Cultivadas , Enfermedad de Huntington/enzimología , Ratones , Proteínas Mutantes
4.
Mol Ther Methods Clin Dev ; 14: 237-251, 2019 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-31440523

RESUMEN

Exosomes represent a strategy for optimizing the adeno-associated virus (AAV) toward the development of novel therapeutic options for neurodegenerative disorders. However, in vivo spreading of exosomes and AAVs after intracerebral administration is poorly understood. This study provides an assessment and comparison of the spreading into the brain of exosome-enveloped AAVs (exo-AAVs) or unassociated AAVs (std-AAVs) through in vivo optical imaging techniques like probe-based confocal laser endomicroscopy (pCLE) and ex vivo fluorescence microscopy. The std-AAV serotypes (AAV6 and AAV9) encoding the GFP were enveloped in exosomes and injected into the ipsilateral hippocampus. At 3 months post-injection, pCLE detected enhanced GFP expression of both exo-AAV serotypes in contralateral hemispheres compared to std-AAVs. Although sparse GFP-positive astrocytes were observed using exo-AAVs, our results show that the enhancement of the transgene expression resulting from exo-AAVs was largely restricted to neurons and oligodendrocytes. Our results suggest (1) the possibility of combining gene therapy with an endoscopic approach to enable tracking of exo-AAV spread, and (2) exo-AAVs allow for widespread, long-term gene expression in the CNS, supporting the use of exo-AAVs as an efficient gene delivery tool.

5.
Acta Neuropathol ; 138(5): 837-858, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31197505

RESUMEN

Spinocerebellar ataxias (SCAs) are devastating neurodegenerative disorders for which no curative or preventive therapies are available. Deregulation of brain cholesterol metabolism and impaired brain cholesterol turnover have been associated with several neurodegenerative diseases. SCA3 or Machado-Joseph disease (MJD) is the most prevalent ataxia worldwide. We show that cholesterol 24-hydroxylase (CYP46A1), the key enzyme allowing efflux of brain cholesterol and activating brain cholesterol turnover, is decreased in cerebellar extracts from SCA3 patients and SCA3 mice. We investigated whether reinstating CYP46A1 expression would improve the disease phenotype of SCA3 mouse models. We show that administration of adeno-associated viral vectors encoding CYP46A1 to a lentiviral-based SCA3 mouse model reduces mutant ataxin-3 accumulation, which is a hallmark of SCA3, and preserves neuronal markers. In a transgenic SCA3 model with a severe motor phenotype we confirm that cerebellar delivery of AAVrh10-CYP46A1 is strongly neuroprotective in adult mice with established pathology. CYP46A1 significantly decreases ataxin-3 protein aggregation, alleviates motor impairments and improves SCA3-associated neuropathology. In particular, improvement in Purkinje cell number and reduction of cerebellar atrophy are observed in AAVrh10-CYP46A1-treated mice. Conversely, we show that knocking-down CYP46A1 in normal mouse brain impairs cholesterol metabolism, induces motor deficits and produces strong neurodegeneration with impairment of the endosomal-lysosomal pathway, a phenotype closely resembling that of SCA3. Remarkably, we demonstrate for the first time both in vitro, in a SCA3 cellular model, and in vivo, in mouse brain, that CYP46A1 activates autophagy, which is impaired in SCA3, leading to decreased mutant ataxin-3 deposition. More broadly, we show that the beneficial effect of CYP46A1 is also observed with mutant ataxin-2 aggregates. Altogether, our results confirm a pivotal role for CYP46A1 and brain cholesterol metabolism in neuronal function, pointing to a key contribution of the neuronal cholesterol pathway in mechanisms mediating clearance of aggregate-prone proteins. This study identifies CYP46A1 as a relevant therapeutic target not only for SCA3 but also for other SCAs.


Asunto(s)
Autofagia/fisiología , Encéfalo/metabolismo , Colesterol/metabolismo , Enfermedad de Machado-Joseph/metabolismo , Ataxias Espinocerebelosas/metabolismo , Adulto , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Enfermedad de Machado-Joseph/patología , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Células de Purkinje/metabolismo , Células de Purkinje/patología , Ataxias Espinocerebelosas/patología
6.
Acta Neuropathol Commun ; 7(1): 46, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30885273

RESUMEN

Recent evidences suggest the involvement of DYRK1A (dual specificity tyrosine phosphorylation-regulated kinase 1 A) in Alzheimer's disease (AD). Here we showed that DYRK1A undergoes a proteolytic processing in AD patients hippocampus without consequences on its kinase activity. Resulting truncated forms accumulate in astrocytes and exhibit increased affinity towards STAT3ɑ, a regulator of inflammatory process. These findings were confirmed in APP/PS1 mice, an amyloid model of AD, suggesting that this DYRK1A cleavage is a consequence of the amyloid pathology. We identified in vitro the Leucettine L41 as a compound able to prevent DYRK1A proteolysis in both human and mouse protein extracts. We then showed that intraperitoneal injections of L41 in aged APP/PS1 mice inhibit STAT3ɑ phosphorylation and reduce pro-inflammatory cytokines levels (IL1- ß, TNF-ɑ and IL-12) associated to an increased microglial recruitment around amyloid plaques and decreased amyloid-ß plaque burden. Importantly, L41 treatment improved synaptic plasticity and rescued memory functions in APP/PS1 mice. Collectively, our results suggest that DYRK1A may contribute to AD pathology through its proteolytic process, reducing its kinase specificity. Further evaluation of inhibitors of DYRK1A truncation promises a new therapeutic approach for AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Fenotipo , Presenilina-1/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteolisis , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Animales , Hipocampo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Quinasas DyrK
7.
Dis Model Mech ; 12(1)2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30559154

RESUMEN

Perturbation of protein homeostasis and aggregation of misfolded proteins is a major cause of many human diseases. A hallmark of the neurodegenerative disease spinocerebellar ataxia type 7 (SCA7) is the intranuclear accumulation of mutant, misfolded ataxin-7 (polyQ-ATXN7). Here, we show that endogenous ATXN7 is modified by SUMO proteins, thus also suggesting a physiological role for this modification under conditions of proteotoxic stress caused by the accumulation of polyQ-ATXN7. Co-immunoprecipitation experiments, immunofluorescence microscopy and proximity ligation assays confirmed the colocalization and interaction of polyQ-ATXN7 with SUMO2 in cells. Moreover, upon inhibition of the proteasome, both endogenous SUMO2/3 and the RNF4 ubiquitin ligase surround large polyQ-ATXN7 intranuclear inclusions. Overexpression of RNF4 and/or SUMO2 significantly decreased levels of polyQ-ATXN7 and, upon proteasomal inhibition, led to a marked increase in the polyubiquitination of polyQ-ATXN7. This provides a mechanism for the clearance of polyQ-ATXN7 from affected cells that involves the recruitment of RNF4 by SUMO2/3-modified polyQ-ATXN7, thus leading to its ubiquitination and proteasomal degradation. In a SCA7 knock-in mouse model, we similarly observed colocalization of SUMO2/3 with polyQ-ATXN7 inclusions in the cerebellum and retina. Furthermore, we detected accumulation of SUMO2/3 high-molecular-mass species in the cerebellum of SCA7 knock-in mice, compared with their wild-type littermates, and changes in SUMO-related transcripts. Immunohistochemical analysis showed the accumulation of SUMO proteins and RNF4 in the cerebellum of SCA7 patients. Taken together, our results show that the SUMO pathway contributes to the clearance of aggregated ATXN7 and suggest that its deregulation might be associated with SCA7 disease progression.


Asunto(s)
Ataxina-7/metabolismo , Proteínas Nucleares/metabolismo , Pliegue de Proteína , Proteolisis , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Ataxias Espinocerebelosas/metabolismo , Sumoilación , Factores de Transcripción/metabolismo , Animales , Cerebelo/metabolismo , Niño , Modelos Animales de Enfermedad , Células HEK293 , Células HeLa , Humanos , Cuerpos de Inclusión/metabolismo , Células MCF-7 , Ratones , Persona de Mediana Edad , Mutación/genética , Proteína de la Leucemia Promielocítica/metabolismo , Inhibidores de Proteasoma/farmacología , Agregado de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos , Ataxias Espinocerebelosas/patología , Sumoilación/efectos de los fármacos , Ubiquitina/metabolismo
9.
Neuropathology ; 38(2): 113-124, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29218765

RESUMEN

Autophagy, the major pathway for protein turnover, is critical to maintain cellular homeostasis and has been implicated in neurodegenerative diseases. The aim of this research was to analyze the expression of autophagy markers in postmortem brains from Machado-Joseph disease (MJD) patients. The expression of autophagy markers in the cerebellum and the oculomotor nucleus from MJD patients and age-matched controls with no signs of neuropathology was inspected postmortem by immunohistochemistry (IHC) and Western blot. Furthermore, autophagy was examined by means of transmission electron microscopy (TEM). Western blot and IHC revealed nuclear accumulation of misfolded ataxin-3 (ATXN3) and the presence of ubiquitin- and p62-positive aggregates in MJD patients as compared to controls. Moreover, the autophagic proteins, autophagy-related gene (Atg) protein (ATG)-7, ATG-12, ATG16L2 and autophagosomal microtubule-associated protein light chain 3 (LC3) were significantly increased in MJD brains relative to controls, while beclin-1 levels were reduced in MJD patients. Increase in the levels of lysosomal-associated membrane protein 2 (LAMP-2) and of the endosomal markers (Rab7 and Rab1A) were observed in MJD patients relatively to controls. In addition, these findings were further confirmed by TEM in brain tissue where large vesicles accumulating electron-dense materials were highly enriched in MJD patients. Postmortem brains with MJD exhibit increased markers of autophagy relative to age-matched control brains, therefore suggesting strong dysregulation of autophagy that may have an important role in the course of MJD pathogenesis.


Asunto(s)
Autofagia , Cerebelo/metabolismo , Enfermedad de Machado-Joseph/metabolismo , Complejo Nuclear Oculomotor/metabolismo , Adulto , Ataxina-3/metabolismo , Beclina-1/metabolismo , Biomarcadores/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Endosomas/metabolismo , Femenino , Proteínas Ligadas a GPI/metabolismo , Humanos , Lisosomas/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sirolimus/metabolismo , Ubiquitina/metabolismo
10.
Cereb Cortex ; 28(11): 3976-3993, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29048465

RESUMEN

The treatment of Alzheimer's disease (AD) remains challenging and requires a better in depth understanding of AD progression. Particularly, the link between amyloid protein precursor (APP) processing and Tau pathology development remains poorly understood. Growing evidences suggest that APP processing and amyloid-ß (Aß) release are upstream of Tau pathology but the lack of animal models mimicking the slow progression of human AD raised questions around this mechanism. Here, we described that an AD-like ßAPP processing in adults wild-type rats, yielding to human APP, ßCTF and Aß levels similar to those observed in AD patients, is sufficient to trigger gradual Tauopathy. The Tau hyperphosphorylation begins several months before the formation of both amyloid plaques and tangle-like aggregates in aged rats and without associated inflammation. Based on a longitudinal characterization over 30 months, we showed that extrasynaptic and emotional impairments appear before long-term potentiation deficits and memory decline and so before Aß and Tau aggregations. These compelling data allowed us to (1) experimentally confirm the causal relationship between ßAPP processing and Tau pathology in vivo and without Tau transgene overexpression, (2) support the amyloidogenic cascade and (3) propose a 4-step hypothesis of prodromal AD progression.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Péptidos beta-Amiloides/metabolismo , Animales , Progresión de la Enfermedad , Femenino , Vectores Genéticos , Humanos , Potenciación a Largo Plazo , Masculino , Fragmentos de Péptidos/metabolismo , Placa Amiloide/metabolismo , Presenilina-1/genética , Agregación Patológica de Proteínas/metabolismo , Ratas Wistar
11.
Hum Gene Ther ; 28(11): 988-1003, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29035118

RESUMEN

Clinical gene therapy has made important advances over the last decade. Among neurological diseases, severe genetic neurodegenerative conditions have been the focus of initial clinical applications. Gene therapy has also addressed complex neurodegenerative diseases, particularly Parkinson's disease, with encouraging results in human patients, demonstrating that specific targeting of central nervous system (CNS) cells is a relevant strategy for severe pathologies and that efficient access to the CNS with viral vectors is an achievable goal. The purpose of this review is to summarize the gene therapy clinical applications that have been conducted for neurodegenerative diseases. Limitations and hurdles to obtain and demonstrate benefit in patients, and the new developments that should allow new clinical applications with high beneficial potential are discussed.


Asunto(s)
Terapia Genética/tendencias , Vectores Genéticos/uso terapéutico , Enfermedades Neurodegenerativas/terapia , Enfermedad de Parkinson/terapia , Sistema Nervioso Central/patología , Dependovirus/genética , Humanos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología
12.
Front Mol Neurosci ; 10: 211, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28744197

RESUMEN

Impairment in cholesterol metabolism is associated with many neurodegenerative disorders including Alzheimer's disease (AD). However, the lipid alterations underlying neurodegeneration and the connection between altered cholesterol levels and AD remains not fully understood. We recently showed that cholesterol accumulation in hippocampal neurons, induced by silencing Cyp46a1 gene expression, leads to neurodegeneration with a progressive neuronal loss associated with AD-like phenotype in wild-type mice. We used a targeted and non-targeted lipidomics approach by liquid chromatography coupled to high-resolution mass spectrometry to further characterize lipid modifications associated to neurodegeneration and cholesterol accumulation induced by CYP46A1 inhibition. Hippocampus lipidome of normal mice was profiled 4 weeks after cholesterol accumulation due to Cyp46a1 gene expression down-regulation at the onset of neurodegeneration. We showed that major membrane lipids, sphingolipids and specific enzymes involved in phosphatidylcholine and sphingolipid metabolism, were rapidly increased in the hippocampus of AAV-shCYP46A1 injected mice. This lipid accumulation was associated with alterations in the lysosomal cargoe, accumulation of phagolysosomes and impairment of endosome-lysosome trafficking. Altogether, we demonstrated that inhibition of cholesterol 24-hydroxylase, key enzyme of cholesterol metabolism leads to a complex dysregulation of lipid homeostasis. Our results contribute to dissect the potential role of lipids in severe neurodegenerative diseases like AD.

13.
Sci Rep ; 7(1): 4955, 2017 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-28694463

RESUMEN

Gadolinium (Gd)-stained MRI is based on Gd contrast agent (CA) administration into the brain parenchyma. The strong signal increase induced by Gd CA can be converted into resolution enhancement to record microscopic MR images. Moreover, inhomogeneous distribution of the Gd CA in the brain improves the contrast between different tissues and provides new contrasts in MR images. Gd-stained MRI detects amyloid plaques, one of the microscopic lesions of Alzheimer's disease (AD), in APPSL/PS1M146L mice or in primates. Numerous transgenic mice with various plaque typologies have been developed to mimic cerebral amyloidosis and comparison of plaque detection between animal models and humans with new imaging methods is a recurrent concern. Here, we investigated detection of amyloid plaques by Gd-stained MRI in five mouse models of amyloidosis (APPSL/PS1M146L, APP/PS1dE9, APP23, APPSwDI, and 3xTg) presenting with compact, diffuse and intracellular plaques as well as in post mortem human-AD brains. The brains were then evaluated by histology to investigate the impact of size, compactness, and iron load of amyloid plaques on their detection by MRI. We show that Gd-stained MRI allows detection of compact amyloid plaques as small as 25 µm, independently of their iron load, in mice as well as in human-AD brains.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Amiloidosis/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Placa Amiloide/diagnóstico por imagen , Enfermedad de Alzheimer/metabolismo , Amiloidosis/metabolismo , Animales , Autopsia , Medios de Contraste/administración & dosificación , Modelos Animales de Enfermedad , Gadolinio/administración & dosificación , Humanos , Hierro/metabolismo , Ratones , Ratones Transgénicos , Placa Amiloide/metabolismo
15.
Brain ; 140(3): 826-842, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28003243

RESUMEN

Interleukin-2 (IL-2)-deficient mice have cytoarchitectural hippocampal modifications and impaired learning and memory ability reminiscent of Alzheimer's disease. IL-2 stimulates regulatory T cells whose role is to control inflammation. As neuroinflammation contributes to neurodegeneration, we investigated IL-2 in Alzheimer's disease. Therefore, we investigated IL-2 levels in hippocampal biopsies of patients with Alzheimer's disease relative to age-matched control individuals. We then treated APP/PS1ΔE9 mice having established Alzheimer's disease with IL-2 for 5 months using single administration of an AAV-IL-2 vector. We first found decreased IL-2 levels in hippocampal biopsies of patients with Alzheimer's disease. In mice, IL-2-induced systemic and brain regulatory T cells expansion and activation. In the hippocampus, IL-2 induced astrocytic activation and recruitment of astrocytes around amyloid plaques, decreased amyloid-ß42/40 ratio and amyloid plaque load, improved synaptic plasticity and significantly rescued spine density. Of note, this tissue remodelling was associated with recovery of memory deficits, as assessed in the Morris water maze task. Altogether, our data strongly suggest that IL-2 can alleviate Alzheimer's disease hallmarks in APP/PS1ΔE9 mice with established pathology. Therefore, this should prompt the investigation of low-dose IL-2 in Alzheimer's disease and other neuroinflammatory/neurodegenerative disorders.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Antipsicóticos/uso terapéutico , Interleucina-2/uso terapéutico , Trastornos de la Memoria/tratamiento farmacológico , Plasticidad Neuronal/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Antipsicóticos/farmacología , Estudios de Casos y Controles , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/genética , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Femenino , Regulación de la Expresión Génica/genética , Humanos , Interleucina-2/sangre , Interleucina-2/farmacología , Masculino , Trastornos de la Memoria/etiología , Ratones , Ratones Transgénicos , Plasticidad Neuronal/genética , Placa Amiloide/patología , Presenilina-1/genética , Sinapsis/efectos de los fármacos , Sinapsis/patología , Sinapsis/ultraestructura
16.
Mol Neurodegener ; 11(1): 58, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27465358

RESUMEN

BACKGROUND: We used lentiviral vectors (LVs) to generate a new SCA7 animal model overexpressing a truncated mutant ataxin-7 (MUT ATXN7) fragment in the mouse cerebellum, in order to characterize the specific neuropathological and behavioral consequences of the genetic defect in this brain structure. RESULTS: LV-mediated overexpression of MUT ATXN7 into the cerebellum of C57/BL6 adult mice induced neuropathological features similar to that observed in patients, such as intranuclear aggregates in Purkinje cells (PC), loss of synaptic markers, neuroinflammation, and neuronal death. No neuropathological changes were observed when truncated wild-type ataxin-7 (WT ATXN7) was injected. Interestingly, the local delivery of LV-expressing mutant ataxin-7 (LV-MUT-ATXN7) into the cerebellum of wild-type mice also mediated the development of an ataxic phenotype at 8 to 12 weeks post-injection. Importantly, our data revealed abnormal levels of the FUS/TLS, MBNL1, and TDP-43 RNA-binding proteins in the cerebellum of the LV-MUT-ATXN7 injected mice. MUT ATXN7 overexpression induced an increase in the levels of the pathological phosphorylated TDP-43, and a decrease in the levels of soluble FUS/TLS, with both proteins accumulating within ATXN7-positive intranuclear inclusions. MBNL1 also co-aggregated with MUT ATXN7 in most PC nuclear inclusions. Interestingly, no MBNL2 aggregation was observed in cerebellar MUT ATXN7 aggregates. Immunohistochemical studies in postmortem tissue from SCA7 patients and SCA7 knock-in mice confirmed SCA7-induced nuclear accumulation of FUS/TLS and MBNL1, strongly suggesting that these proteins play a physiopathological role in SCA7. CONCLUSIONS: This study validates a novel SCA7 mouse model based on lentiviral vectors, in which strong and sustained expression of MUT ATXN7 in the cerebellum was found sufficient to generate motor defects.


Asunto(s)
Ataxina-7/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ARN/metabolismo , Ataxias Espinocerebelosas/genética , Animales , Ataxina-7/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Lentivirus/genética , Ratones Endogámicos C57BL , Neuronas/metabolismo , Fenotipo
17.
Sci Rep ; 6: 28272, 2016 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-27320056

RESUMEN

Recombinant adeno-associated viral (AAV) vectors have advanced to the vanguard of gene therapy. Numerous naturally occurring serotypes have been used to target cells in various tissues. There is a strong need for fast and dynamic methods which efficiently unravel viral tropism in whole organs. Ultramicroscopy (UM) is a novel fluorescence microscopy technique that images optically cleared undissected specimens, achieving good resolutions at high penetration depths while being non-destructive. UM was applied to obtain high-resolution 3D analysis of AAV transduction in adult mouse brains, especially in the hippocampus, a region of interest for Alzheimer's disease therapy. We separately or simultaneously compared transduction efficacies for commonly used serotypes (AAV9 and AAVrh10) using fluorescent reporter expression. We provide a detailed comparative and quantitative analysis of the transduction profiles. UM allowed a rapid analysis of marker fluorescence expression in neurons with intact projections deep inside the brain, in defined anatomical structures. Major hippocampal neuronal transduction was observed with both vectors, with slightly better efficacy for AAV9 in UM. Glial response and synaptic marker expression did not change post transduction.We propose UM as a novel valuable complementary tool to efficiently and simultaneously unravel tropism of different viruses in a single non-dissected adult rodent brain.


Asunto(s)
Encéfalo/virología , Dependovirus/fisiología , Terapia Genética , Neuronas/virología , Transducción Genética , Tropismo Viral , Animales , Encéfalo/metabolismo , Encéfalo/patología , Masculino , Ratones , Microscopía Fluorescente , Neuronas/metabolismo , Neuronas/patología
18.
Brain ; 139(Pt 3): 953-70, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26912634

RESUMEN

Huntington's disease is an autosomal dominant neurodegenerative disease caused by abnormal polyglutamine expansion in huntingtin (Exp-HTT) leading to degeneration of striatal neurons. Altered brain cholesterol homeostasis has been implicated in Huntington's disease, with increased accumulation of cholesterol in striatal neurons yet reduced levels of cholesterol metabolic precursors. To elucidate these two seemingly opposing dysregulations, we investigated the expression of cholesterol 24-hydroxylase (CYP46A1), the neuronal-specific and rate-limiting enzyme for cholesterol conversion to 24S-hydroxycholesterol (24S-OHC). CYP46A1 protein levels were decreased in the putamen, but not cerebral cortex samples, of post-mortem Huntington's disease patients when compared to controls. Cyp46A1 mRNA and CYP46A1 protein levels were also decreased in the striatum of the R6/2 Huntington's disease mouse model and in SThdhQ111 cell lines. In vivo, in a wild-type context, knocking down CYP46A1 expression in the striatum, via an adeno-associated virus-mediated delivery of selective shCYP46A1, reproduced the Huntington's disease phenotype, with spontaneous striatal neuron degeneration and motor deficits, as assessed by rotarod. In vitro, CYP46A1 restoration protected SThdhQ111 and Exp-HTT-expressing striatal neurons in culture from cell death. In the R6/2 Huntington's disease mouse model, adeno-associated virus-mediated delivery of CYP46A1 into the striatum decreased neuronal atrophy, decreased the number, intensity level and size of Exp-HTT aggregates and improved motor deficits, as assessed by rotarod and clasping behavioural tests. Adeno-associated virus-CYP46A1 infection in R6/2 mice also restored levels of cholesterol and lanosterol and increased levels of desmosterol. In vitro, lanosterol and desmosterol were found to protect striatal neurons expressing Exp-HTT from death. We conclude that restoring CYP46A1 activity in the striatum promises a new therapeutic approach in Huntington's disease.


Asunto(s)
Colesterol/metabolismo , Enfermedad de Huntington/enzimología , Enfermedad de Huntington/prevención & control , Esteroide Hidroxilasas/biosíntesis , Anciano , Anciano de 80 o más Años , Animales , Células Cultivadas , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Colesterol 24-Hidroxilasa , Femenino , Humanos , Enfermedad de Huntington/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Persona de Mediana Edad
19.
Hum Gene Ther ; 27(2): 100-7, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26838997

RESUMEN

Key neuropathological hallmarks of Alzheimer's disease (AD) are extracellular amyloid plaques and intracellular accumulation of hyperphosphorylated Tau protein. The mechanisms underlying these neuropathological changes remain unclear. So far, research on AD therapy has had limited success in terms of symptomatic treatments although it has also had several failures for disease-modifying drugs. Gene transfer strategies to the brain have contributed to evaluate in animal models many interesting tracks, some of which should deserve clinical applications in AD patients in the future.


Asunto(s)
Enfermedad de Alzheimer/terapia , Terapia Genética/métodos , Placa Amiloide/terapia , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/administración & dosificación , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Anticuerpos Neutralizantes/uso terapéutico , Apolipoproteínas E/administración & dosificación , Apolipoproteínas E/genética , Ácido Aspártico Endopeptidasas/administración & dosificación , Ácido Aspártico Endopeptidasas/genética , Encéfalo/metabolismo , Encéfalo/patología , Factor Neurotrófico Derivado del Encéfalo/administración & dosificación , Factor Neurotrófico Derivado del Encéfalo/genética , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Lentivirus/genética , Factor 2 Relacionado con NF-E2/administración & dosificación , Factor 2 Relacionado con NF-E2/genética , Placa Amiloide/genética , Placa Amiloide/patología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Somatomedinas/administración & dosificación , Somatomedinas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...