Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Elife ; 112022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36205397

RESUMEN

We created a new nonhuman primate model of the genetic neurodegenerative disorder Huntington's disease (HD) by injecting a mixture of recombinant adeno-associated viral vectors, serotypes AAV2 and AAV2.retro, each expressing a fragment of human mutant HTT (mHTT) into the caudate and putamen of adult rhesus macaques. This modeling strategy results in expression of mutant huntingtin protein (mHTT) and aggregate formation in the injected brain regions, as well as dozens of other cortical and subcortical brain regions affected in human HD patients. We queried the disruption of cortico-basal ganglia circuitry for 30 months post-surgery using a variety of behavioral and imaging readouts. Compared to controls, mHTT-treated macaques developed working memory decline and progressive motor impairment. Multimodal imaging revealed circuit-wide white and gray matter degenerative processes in several key brain regions affected in HD. Taken together, we have developed a novel macaque model of HD that may be used to develop disease biomarkers and screen promising therapeutics.


Asunto(s)
Disfunción Cognitiva , Enfermedad de Huntington , Enfermedades Neurodegenerativas , Adulto , Animales , Biomarcadores , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/patología , Macaca mulatta
2.
Sci Rep ; 10(1): 6970, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32332773

RESUMEN

Recently, AAV2.retro, a new capsid variant capable of efficient retrograde transport in brain, was generated in mice using a directed evolution approach. However, it remains unclear to what degree transport will be recapitulated in the substantially larger and more complex nonhuman primate (NHP) brain. Here, we compared the biodistribution of AAV2.retro with its parent serotype, AAV2, in adult macaques following delivery into the caudate and putamen, brain regions which comprise the striatum. While AAV2 transduction was primarily limited to the injected brain regions, AAV2.retro transduced cells in the striatum and in dozens of cortical and subcortical regions with known striatal afferents. We then evaluated the capability of AAV2.retro to deliver disease-related gene cargo to biologically-relevant NHP brain circuits by packaging a fragment of human mutant HTT, the causative gene mutation in Huntington's disease. Following intra-striatal delivery, pathological mHTT-positive protein aggregates were distributed widely among cognitive, motor, and limbic cortico-basal ganglia circuits. Together, these studies demonstrate strong retrograde transport of AAV2.retro in NHP brain, highlight its utility in developing novel NHP models of brain disease and suggest its potential for querying circuit function and delivering therapeutic genes in the brain, particularly where treating dysfunctional circuits, versus single brain regions, is warranted.


Asunto(s)
Encéfalo/metabolismo , Parvovirinae/metabolismo , Animales , Anticuerpos Neutralizantes/metabolismo , Transporte Biológico/fisiología , Dependovirus , Modelos Animales de Enfermedad , Femenino , Humanos , Macaca mulatta , Masculino , Enfermedades Neurodegenerativas/metabolismo
3.
Mol Ther ; 27(11): 2018-2037, 2019 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-31420242

RESUMEN

The ability of recombinant adeno-associated virus (AAV) to deliver transgenes to the CNS has allowed for several advancements in the field of gene therapy to treat brain disorders. Although most AAVs do not readily cross the blood-brain barrier and transduce the CNS following peripheral administration, AAV-PHP.B has recently been shown to transduce brains of mice with higher efficiency compared with its parent serotype, AAV9, following injection into the retro-orbital sinus. Here, we extended this foundational work by comparing AAV-PHP.B transduction efficiency in wild-type C57BL/6J mice using four clinically applicable delivery strategies including two intravascular (intra-jugular vein and intra-carotid artery) and two intra-cerebral spinal fluid (CSF) routes (intra-cisterna magna and intra-lateral ventricle). We scaled up these comparisons in a larger-animal model and evaluated transduction efficiency of AAV-PHP.B in the rhesus macaque. We found widespread and largely equal CNS transduction in mice following all four injection strategies, whereas we observed a differential pattern of transduction in macaques with broad cortical and spinal cord transduction seen after intrathecal administration and only very low transduction following intravascular administration. Taken together, these results suggest that AAV-PHP.B may be a useful gene therapy vector for neurological disorders, particularly those stemming from broad cortical or spinal cord neuropathology.


Asunto(s)
Sistema Nervioso Central/metabolismo , Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Transducción Genética , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Expresión Génica , Genes Reporteros , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Macaca mulatta , Ratones , Neuronas/metabolismo , Médula Espinal/metabolismo , Distribución Tisular , Transgenes
4.
JCI Insight ; 3(22)2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30429376

RESUMEN

Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant or digenic disorder linked to derepression of the toxic DUX4 gene in muscle. There is currently no pharmacological treatment. The emergence of DUX4 enabled development of cell and animal models that could be used for basic and translational research. Since DUX4 is toxic, animal model development has been challenging, but progress has been made, revealing that tight regulation of DUX4 expression is critical for creating viable animals that develop myopathy. Here, we report such a model - the tamoxifen-inducible FSHD mouse model called TIC-DUX4. Uninduced animals are viable, born in Mendelian ratios, and overtly indistinguishable from WT animals. Induced animals display significant DUX4-dependent myopathic phenotypes at the molecular, histological, and functional levels. To demonstrate the utility of TIC-DUX4 mice for therapeutic development, we tested a gene therapy approach aimed at improving muscle strength in DUX4-expressing muscles using adeno-associated virus serotype 1.Follistatin (AAV1.Follistatin), a natural myostatin antagonist. This strategy was not designed to modulate DUX4 but could offer a mechanism to improve muscle weakness caused by DUX4-induced damage. AAV1.Follistatin significantly increased TIC-DUX4 muscle mass and strength even in the presence of DUX4 expression, suggesting that myostatin inhibition may be a promising approach to treat FSHD-associated weakness. We conclude that TIC-DUX4 mice are a relevant model to study DUX4 toxicity and, importantly, are useful in therapeutic development studies for FSHD.


Asunto(s)
Modelos Animales de Enfermedad , Folistatina/genética , Terapia Genética , Proteínas de Homeodominio/genética , Distrofia Muscular Facioescapulohumeral/terapia , Miostatina/antagonistas & inhibidores , Animales , Femenino , Folistatina/uso terapéutico , Masculino , Ratones Transgénicos , Distrofia Muscular Facioescapulohumeral/inducido químicamente , Distrofia Muscular Facioescapulohumeral/genética , Fenotipo , Tamoxifeno
5.
Neurobiol Dis ; 119: 65-78, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30048804

RESUMEN

We have identified a natural Japanese macaque model of the childhood neurodegenerative disorder neuronal ceroid lipofuscinosis, commonly known as Batten Disease, caused by a homozygous frameshift mutation in the CLN7 gene (CLN7-/-). Affected macaques display progressive neurological deficits including visual impairment, tremor, incoordination, ataxia and impaired balance. Imaging, functional and pathological studies revealed that CLN7-/- macaques have reduced retinal thickness and retinal function early in disease, followed by profound cerebral and cerebellar atrophy that progresses over a five to six-year disease course. Histological analyses showed an accumulation of cerebral, cerebellar and cardiac storage material as well as degeneration of neurons, white matter fragmentation and reactive gliosis throughout the brain of affected animals. This novel CLN7-/- macaque model recapitulates key behavioral and neuropathological features of human Batten Disease and provides novel insights into the pathophysiology linked to CLN7 mutations. These animals will be invaluable for evaluating promising therapeutic strategies for this devastating disease.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas de Transporte de Membrana/genética , Lipofuscinosis Ceroideas Neuronales/diagnóstico por imagen , Lipofuscinosis Ceroideas Neuronales/genética , Animales , Femenino , Técnicas de Inactivación de Genes/métodos , Locomoción/fisiología , Macaca , Masculino , Mutación Missense/genética , Lipofuscinosis Ceroideas Neuronales/fisiopatología , Equilibrio Postural/fisiología , Primates , Trastornos de la Visión/diagnóstico por imagen , Trastornos de la Visión/genética , Trastornos de la Visión/fisiopatología
6.
Mol Ther Methods Clin Dev ; 8: 121-130, 2018 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-29387734

RESUMEN

RNAi emerged as a prospective molecular therapy nearly 15 years ago. Since then, two major RNAi platforms have been under development: oligonucleotides and gene therapy. Oligonucleotide-based approaches have seen more advancement, with some promising therapies that may soon reach market. In contrast, vector-based approaches for RNAi therapy have remained largely in the pre-clinical realm, with limited clinical safety and efficacy data to date. We are developing a gene therapy approach to treat the autosomal-dominant disorder facioscapulohumeral muscular dystrophy. Our strategy involves silencing the myotoxic gene DUX4 using adeno-associated viral vectors to deliver targeted microRNA expression cassettes (miDUX4s). We previously demonstrated proof of concept for this approach in mice, and we are now taking additional steps here to assess safety issues related to miDUX4 overexpression and sequence-specific off-target silencing. In this study, we describe improvements in vector design and expansion of our miDUX4 sequence repertoire and report differential toxicity elicited by two miDUX4 sequences, of which one was toxic and the other was not. This study provides important data to help advance our goal of translating RNAi gene therapy for facioscapulohumeral muscular dystrophy.

7.
Hum Mol Genet ; 25(20): 4577-4589, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28173143

RESUMEN

D4Z4 repeats are present in at least 11 different mammalian species, including humans and mice. Each repeat contains an open reading frame encoding a double homeodomain (DUX) family transcription factor. Aberrant expression of the D4Z4 ORF called DUX4 is associated with the pathogenesis of Facioscapulohumeral muscular dystrophy (FSHD). DUX4 is toxic to numerous cell types of different species, and over-expression caused dysmorphism and developmental arrest in frogs and zebrafish, embryonic lethality in transgenic mice, and lesions in mouse muscle. Because DUX4 is a primate-specific gene, questions have been raised about the biological relevance of over-expressing it in non-primate models, as DUX4 toxicity could be related to non-specific cellular stress induced by over-expressing a DUX family transcription factor in organisms that did not co-evolve its regulated transcriptional networks. We assessed toxic phenotypes of DUX family genes, including DUX4, DUX1, DUX5, DUXA, DUX4-s, Dux-bl and mouse Dux. We found that DUX proteins were not universally toxic, and only the mouse Dux gene caused similar toxic phenotypes as human DUX4. Using RNA-seq, we found that 80% of genes upregulated by Dux were similarly increased in DUX4-expressing cells. Moreover, 43% of Dux-responsive genes contained ChIP-seq binding sites for both Dux and DUX4, and both proteins had similar consensus binding site sequences. These results suggested DUX4 and Dux may regulate some common pathways, and despite diverging from a common progenitor under different selective pressures for millions of years, the two genes maintain partial functional homology.


Asunto(s)
Redes Reguladoras de Genes , Proteínas de Homeodominio/metabolismo , Micotoxinas/metabolismo , Mioblastos/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Evolución Molecular , Proteínas de Homeodominio/genética , Humanos , Ratones , Ratones Transgénicos , Distrofia Muscular Facioescapulohumeral/metabolismo , Micotoxinas/genética , Alineación de Secuencia , Análisis de Secuencia de ARN
8.
PLoS One ; 10(3): e0118813, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25742305

RESUMEN

The DUX4 gene, encoded within D4Z4 repeats on human chromosome 4q35, has recently emerged as a key factor in the pathogenic mechanisms underlying Facioscapulohumeral muscular dystrophy (FSHD). This recognition prompted development of animal models expressing the DUX4 open reading frame (ORF) alone or embedded within D4Z4 repeats. In the first published model, we used adeno-associated viral vectors (AAV) and strong viral control elements (CMV promoter, SV40 poly A) to demonstrate that the DUX4 cDNA caused dose-dependent toxicity in mouse muscles. As a follow-up, we designed a second generation of DUX4-expressing AAV vectors to more faithfully genocopy the FSHD-permissive D4Z4 repeat region located at 4q35. This new vector (called AAV.D4Z4.V5.pLAM) contained the D4Z4/DUX4 promoter region, a V5 epitope-tagged DUX4 ORF, and the natural 3' untranslated region (pLAM) harboring two small introns, DUX4 exons 2 and 3, and the non-canonical poly A signal required for stabilizing DUX4 mRNA in FSHD. AAV.D4Z4.V5.pLAM failed to recapitulate the robust pathology of our first generation vectors following delivery to mouse muscle. We found that the DUX4.V5 junction sequence created an unexpected splice donor in the pre-mRNA that was preferentially utilized to remove the V5 coding sequence and DUX4 stop codon, yielding non-functional DUX4 protein with 55 additional residues on its carboxyl-terminus. Importantly, we further found that aberrant splicing could occur in any expression construct containing a functional splice acceptor and sequences resembling minimal splice donors. Our findings represent an interesting case study with respect to AAV.D4Z4.V5.pLAM, but more broadly serve as a note of caution for designing constructs containing V5 epitope tags and/or transgenes with downstream introns and exons.


Asunto(s)
Epítopos/genética , Exones , Intrones , Distrofia Muscular Facioescapulohumeral/genética , Sitios de Empalme de ARN , Transgenes , Animales , Modelos Animales de Enfermedad , Ratones
9.
Mol Ther ; 20(7): 1417-23, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22508491

RESUMEN

No treatment exists for facioscapulohumeral muscular dystrophy (FSHD), one of the most common inherited muscle diseases. Although FSHD can be debilitating, little effort has been made to develop targeted therapies. This lack of focus on targeted FSHD therapy perpetuated because the genes and pathways involved in the disorder were not understood. Now, more than 2 decades after efforts to decipher the root cause of FSHD began, this barrier to translation is finally lowering. Specifically, several recent studies support an FSHD pathogenesis model involving overexpression of the myopathic DUX4 gene. DUX4 inhibition has therefore emerged as a promising therapeutic strategy for FSHD. In this study, we tested a preclinical RNA interference (RNAi)-based DUX4 gene silencing approach as a prospective treatment for FSHD. We found that adeno-associated viral (AAV) vector-delivered therapeutic microRNAs corrected DUX4-associated myopathy in mouse muscle. These results provide proof-of-principle for RNAi therapy of FSHD through DUX4 inhibition.


Asunto(s)
Proteínas de Homeodominio/genética , Distrofia Muscular Facioescapulohumeral/genética , Distrofia Muscular Facioescapulohumeral/terapia , ARN Interferente Pequeño/uso terapéutico , Animales , Dependovirus/genética , Femenino , Terapia Genética , Vectores Genéticos , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/uso terapéutico , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapulohumeral/metabolismo , Interferencia de ARN
10.
Cell Mol Life Sci ; 68(17): 2951-60, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21152952

RESUMEN

Primary cilia are nearly ubiquitous cellular appendages that provide important sensory and signaling functions. Ciliary dysfunction underlies numerous human diseases, collectively termed ciliopathies. Primary cilia have distinct functions on different cell types and these functions are defined by the signaling proteins that localize to the ciliary membrane. Neurons throughout the mammalian brain possess primary cilia upon which certain G protein-coupled receptors localize. Yet, the precise signaling proteins present on the vast majority of neuronal cilia are unknown. Here, we report that dopamine receptor 1 (D1) localizes to cilia on mouse central neurons, thereby implicating neuronal cilia in dopamine signaling. Interestingly, ciliary localization of D1 is dynamic, and the receptor rapidly translocates to and from cilia in response to environmental cues. Notably, the translocation of D1 from cilia requires proteins mutated in the ciliopathy Bardet-Biedl syndrome (BBS), and we find that one of the BBS proteins, Bbs5, specifically interacts with D1.


Asunto(s)
Proteínas Portadoras/metabolismo , Cilios/metabolismo , Receptores de Dopamina D1/metabolismo , Animales , Síndrome de Bardet-Biedl/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas del Citoesqueleto , Humanos , Ratones , Ratones Noqueados , Neuronas/citología , Proteínas de Unión a Fosfato , Proteínas/metabolismo , Receptores de Dopamina D1/análisis , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
11.
Methods Cell Biol ; 91: 111-21, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20409783

RESUMEN

Primary cilia were first detected on neurons in the mammalian brain over 40 years ago using electron microscopy. However, this approach is very labor intensive and has inherent limitations that restrict its utility for studying neuronal cilia. While the study of cilia in other tissues was greatly facilitated by the identification of specific ciliary markers, historically there have been no markers for neuronal cilia. Fortunately, recent developments make the study of neuronal cilia more practical. First, specific proteins have been shown to selectively localize to neuronal cilia and can serve as markers by immunolabeling. Second, neurons have been shown to possess cilia in culture, which allows for the use of additional approaches, such as live-cell imaging of neuronal cilia. This chapter provides an overview of the current techniques for visualizing neuronal cilia in tissue as well as fixed and living cells. These approaches allow for the identification of additional neuronal ciliary proteins and provide a basis for future functional studies.


Asunto(s)
Biomarcadores/metabolismo , Cilios , Neuronas , Animales , Encéfalo/citología , Encéfalo/metabolismo , Cilios/metabolismo , Cilios/ultraestructura , Inmunohistoquímica/métodos , Ratones , Neuronas/citología , Neuronas/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Coloración y Etiquetado/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA