Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Neurosci ; 44(14)2024 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-38388424

RESUMEN

A missense mutation in the transcription repressor Nucleus accumbens-associated 1 (NACC1) gene at c.892C>T (p.Arg298Trp) on chromosome 19 causes severe neurodevelopmental delay ( Schoch et al., 2017). To model this disorder, we engineered the first mouse model with the homologous mutation (Nacc1+/R284W ) and examined mice from E17.5 to 8 months. Both genders had delayed weight gain, epileptiform discharges and altered power spectral distribution in cortical electroencephalogram, behavioral seizures, and marked hindlimb clasping; females displayed thigmotaxis in an open field. In the cortex, NACC1 long isoform, which harbors the mutation, increased from 3 to 6 months, whereas the short isoform, which is not present in humans and lacks aaR284 in mice, rose steadily from postnatal day (P) 7. Nuclear NACC1 immunoreactivity increased in cortical pyramidal neurons and parvalbumin containing interneurons but not in nuclei of astrocytes or oligodendroglia. Glial fibrillary acidic protein staining in astrocytic processes was diminished. RNA-seq of P14 mutant mice cortex revealed over 1,000 differentially expressed genes (DEGs). Glial transcripts were downregulated and synaptic genes upregulated. Top gene ontology terms from upregulated DEGs relate to postsynapse and ion channel function, while downregulated DEGs enriched for terms relating to metabolic function, mitochondria, and ribosomes. Levels of synaptic proteins were changed, but number and length of synaptic contacts were unaltered at 3 months. Homozygosity worsened some phenotypes including postnatal survival, weight gain delay, and increase in nuclear NACC1. This mouse model simulates a rare form of autism and will be indispensable for assessing pathophysiology and targets for therapeutic intervention.


Asunto(s)
Trastorno Autístico , Factores de Transcripción , Animales , Femenino , Humanos , Masculino , Ratones , Mutación/genética , Proteínas de Neoplasias/genética , Isoformas de Proteínas/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Aumento de Peso
2.
Hum Gene Ther ; 33(1-2): 25-36, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34376056

RESUMEN

Huntington's disease (HD) is a devastating, autosomal dominant neurodegenerative disease caused by a trinucleotide repeat expansion in the huntingtin (HTT) gene. Inactivation of the mutant allele by clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 based gene editing offers a possible therapeutic approach for this disease, but permanent disruption of normal HTT function might compromise adult neuronal function. Here, we use a novel HD mouse model to examine allele-specific editing of mutant HTT (mHTT), with a BAC97 transgene expressing mHTT and a YAC18 transgene expressing normal HTT. We achieve allele-specific inactivation of HTT by targeting a protein coding sequence containing a common, heterozygous single nucleotide polymorphism (SNP). The outcome is a marked and allele-selective reduction of mHTT protein in a mouse model of HD. Expression of a single CRISPR-Cas9 nuclease in neurons generated a high frequency of mutations in the targeted HD allele that included both small insertion/deletion (InDel) mutations and viral vector insertions. Thus, allele-specific targeting of InDel and insertion mutations to heterozygous coding region SNPs provides a feasible approach to inactivate autosomal dominant mutations that cause genetic disease.


Asunto(s)
Enfermedad de Huntington , Alelos , Animales , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Ratones , Polimorfismo de Nucleótido Simple
3.
Nat Methods ; 5(11): 951-3, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18931668

RESUMEN

Allele-specific silencing using small interfering RNAs targeting heterozygous single-nucleotide polymorphisms (SNPs) is a promising therapy for human trinucleotide repeat diseases such as Huntington's disease. Linking SNP identities to the two HTT alleles, normal and disease-causing, is a prerequisite for allele-specific RNA interference. Here we describe a method, SNP linkage by circularization (SLiC), to identify linkage between CAG repeat length and nucleotide identity of heterozygous SNPs using Huntington's disease patient peripheral blood samples.


Asunto(s)
Enfermedad de Huntington/genética , Polimorfismo de Nucleótido Simple/genética , Repeticiones de Trinucleótidos/genética , Humanos , Datos de Secuencia Molecular
4.
PLoS Genet ; 2(9): e140, 2006 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-16965178

RESUMEN

Small interfering RNAs (siRNAs), the guides that direct RNA interference (RNAi), provide a powerful tool to reduce the expression of a single gene in human cells. Ideally, dominant, gain-of-function human diseases could be treated using siRNAs that specifically silence the mutant disease allele, while leaving expression of the wild-type allele unperturbed. Previous reports suggest that siRNAs can be designed with single nucleotide specificity, but no rational basis for the design of siRNAs with single nucleotide discrimination has been proposed. We systematically identified siRNAs that discriminate between the wild-type and mutant alleles of two disease genes: the human Cu, Zn superoxide dismutase (SOD1) gene, which contributes to the progression of hereditary amyotrophic lateral sclerosis through the gain of a toxic property, and the huntingtin (HTT) gene, which causes Huntington disease when its CAG-repeat region expands beyond approximately 35 repeats. Using cell-free RNAi reactions in Drosophila embryo lysate and reporter assays and microarray analysis of off-target effects in cultured human cells, we identified positions within an siRNA that are most sensitive to mismatches. We also show that purine:purine mismatches imbue an siRNA with greater discriminatory power than other types of base mismatches. siRNAs in which either a G:U wobble or a mismatch is located in the "seed" sequence, the specialized siRNA guide region responsible for target binding, displayed lower levels of selectivity than those in which the mismatch was located 3' to the seed; this region of an siRNA is critical for target cleavage but not siRNA binding. Our data suggest that siRNAs can be designed to discriminate between the wild-type and mutant alleles of many genes that differ by just a single nucleotide.


Asunto(s)
Diseño Asistido por Computadora , Silenciador del Gen/fisiología , Nucleótidos/química , ARN Interferente Pequeño/síntesis química , Homología de Secuencia de Ácido Nucleico , Animales , Composición de Base , Disparidad de Par Base/fisiología , Emparejamiento Base , Secuencia de Bases , Sistema Libre de Células , Células Cultivadas , Drosophila/química , Embrión no Mamífero/química , Marcación de Gen/métodos , Células HeLa , Humanos , Análisis por Micromatrices , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Purinas/metabolismo , Sensibilidad y Especificidad , Superóxido Dismutasa/genética , Superóxido Dismutasa-1
5.
Hum Gene Ther ; 29(6): 663-673, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29207890

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disease caused by a genetic expansion of the CAG repeat region in the huntingtin (HTT) gene. Studies in HD mouse models have shown that artificial miRNAs can reduce mutant HTT, but evidence for their effectiveness and safety in larger animals is lacking. HD transgenic sheep express the full-length human HTT with 73 CAG repeats. AAV9 was used to deliver unilaterally to HD sheep striatum an artificial miRNA targeting exon 48 of the human HTT mRNA under control of two alternative promoters: U6 or CßA. The treatment reduced human mutant (m) HTT mRNA and protein 50-80% in the striatum at 1 and 6 months post injection. Silencing was detectable in both the caudate and putamen. Levels of endogenous sheep HTT protein were not affected. There was no significant loss of neurons labeled by DARPP32 or NeuN at 6 months after treatment, and Iba1-positive microglia were detected at control levels. It is concluded that safe and effective silencing of human mHTT protein can be achieved and sustained in a large-animal brain by direct delivery of an AAV carrying an artificial miRNA.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , MicroARNs/metabolismo , Proteínas Mutantes/metabolismo , Neostriado/metabolismo , Animales , Animales Modificados Genéticamente , Dependovirus/genética , Modelos Animales de Enfermedad , Electrólitos/metabolismo , Vectores Genéticos/metabolismo , Genoma Viral , Humanos , Inmunoensayo , Inyecciones , Riñón/fisiopatología , Hígado/fisiopatología , MicroARNs/genética , Microglía/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ovinos
6.
Mol Ther Nucleic Acids ; 7: 324-334, 2017 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-28624208

RESUMEN

Huntington's disease is a devastating, incurable neurodegenerative disease affecting up to 12 per 100,000 patients worldwide. The disease is caused by a mutation in the Huntingtin (Htt) gene. There is interest in reducing mutant Huntingtin by targeting it at the mRNA level, but the maximum tolerable dose and long-term effects of such a treatment are unknown. Using a self-complementary AAV9 vector, we delivered a mir-155-based artificial miRNA under the control of the chicken ß-actin or human U6 promoter. In mouse brain, the artificial miRNA reduced the human huntingtin mRNA by 50%. The U6, but not the CßA promoter, produced the artificial miRNA at supraphysiologic levels. Embedding the antisense strand in a U6-mir-30 scaffold reduced expression of the antisense strand but increased the sense strand. In mice treated with scAAV9-U6-mir-155-HTT or scAAV9-CßA-mir-155-HTT, activated microglia were present around the injection site 1 month post-injection. Six months post-injection, mice treated with scAAV9-CßA-mir-155-HTT were indistinguishable from controls. Those that received scAAV9-U6-mir-155-HTT showed behavioral abnormalities and striatal damage. In conclusion, miRNA backbone and promoter can be used together to modulate expression levels and strand selection of artificial miRNAs, and in brain, the CßA promoter can provide an effective and safe dose of a human huntingtin miRNA.

7.
Sci Rep ; 7: 46740, 2017 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-28436437

RESUMEN

Post-transcriptional gene silencing is a promising therapy for the monogenic, autosomal dominant, Huntington's disease (HD). However, wild-type huntingtin (HTT) has important cellular functions, so the ideal strategy would selectively lower mutant HTT while sparing wild-type. HD patients were genotyped for heterozygosity at three SNP sites, before phasing each SNP allele to wild-type or mutant HTT. Primary ex vivo myeloid cells were isolated from heterozygous patients and transfected with SNP-targeted siRNA, using glucan particles taken up by phagocytosis. Highly selective mRNA knockdown was achieved when targeting each allele of rs362331 in exon 50 of the HTT transcript; this selectivity was also present on protein studies. However, similar selectivity was not observed when targeting rs362273 or rs362307. Furthermore, HD myeloid cells are hyper-reactive compared to control. Allele-selective suppression of either wild-type or mutant HTT produced a significant, equivalent reduction in the cytokine response of HD myeloid cells to LPS, suggesting that wild-type HTT has a novel immune function. We demonstrate a sequential therapeutic process comprising genotyping and mutant HTT-linkage of SNPs, followed by personalised allele-selective suppression in a small patient cohort. We further show that allele-selectivity in ex vivo patient cells is highly SNP-dependent, with implications for clinical trial target selection.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Proteínas Mutantes/genética , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Alelos , Células Cultivadas , Estudios de Cohortes , Genotipo , Humanos , Enfermedad de Huntington/sangre , Persona de Mediana Edad , Células Mieloides/metabolismo , Interferencia de ARN
8.
J Huntingtons Dis ; 5(1): 33-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27003665

RESUMEN

BACKGROUND: Silencing mutant huntingtin mRNA by RNA interference (RNAi) is a therapeutic strategy for Huntington's disease. RNAi induces specific endonucleolytic cleavage of the target HTT mRNA, followed by exonucleolytic processing of the cleaved mRNA fragments. OBJECTIVES: We investigated the clearance of huntingtin mRNA cleavage products following RNAi, to find if particular huntingtin mRNA sequences persist. We especially wanted to find out if the expanded CAG increased production of a toxic mRNA species by impeding degradation of human mutant huntingtin exon 1 mRNA. METHODS: Mice expressing the human mutant HTT transgene with 128 CAG repeats (YAC128 mice) were injected in the striatum with self-complementary AAV9 vectors carrying a miRNA targeting exon 48 of huntingtin mRNA (scAAV-U6-miRNA-HTT-GFP). Transgenic huntingtin mRNA levels were measured in striatal lysates after two weeks. For qPCR, we used species specific primer-probe combinations that together spanned 6 positions along the open reading frame and untranslated regions of the human huntingtin mRNA. Knockdown was also measured in the liver following tail vein injection. RESULTS: Two weeks after intrastriatal administration of scAAV9-U6-miRNA-HTT-GFP, we measured transgenic mutant huntingtin in striatum using probes targeting six different sites along the huntingtin mRNA. Real time PCR showed a reduction of 29% to 36% in human HTT. There was no significant difference in knockdown measured at any of the six sites, including exon 1. In liver, we observed a more pronounced HTT mRNA knockdown of 70% to 76% relative to the untreated mice, and there were also no significant differences among sites. CONCLUSIONS: Our results demonstrate that degradation is equally distributed across the human mutant huntingtin mRNA following RNAi-induced cleavage.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Mutación/genética , Interferencia de ARN , ARN Mensajero/genética , Repeticiones de Trinucleótidos/genética , Animales , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Exones/genética , Técnicas de Silenciamiento del Gen , Proteína Huntingtina/análisis , Proteína Huntingtina/metabolismo , Hígado/metabolismo , Ratones , ARN Mensajero/análisis , ARN Mensajero/metabolismo
9.
Nucleic Acid Ther ; 26(2): 86-92, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26595721

RESUMEN

Preclinical development of RNA interference (RNAi)-based therapeutics requires a rapid, accurate, and robust method of simultaneously quantifying mRNA knockdown in hundreds of samples. The most well-established method to achieve this is quantitative real-time polymerase chain reaction (qRT-PCR), a labor-intensive methodology that requires sample purification, which increases the potential to introduce additional bias. Here, we describe that the QuantiGene(®) branched DNA (bDNA) assay linked to a 96-well Qiagen TissueLyser II is a quick and reproducible alternative to qRT-PCR for quantitative analysis of mRNA expression in vivo directly from tissue biopsies. The bDNA assay is a high-throughput, plate-based, luminescence technique, capable of directly measuring mRNA levels from tissue lysates derived from various biological samples. We have performed a systematic evaluation of this technique for in vivo detection of RNAi-based silencing. We show that similar quality data is obtained from purified RNA and tissue lysates. In general, we observe low intra- and inter-animal variability (around 10% for control samples), and high intermediate precision. This allows minimization of sample size for evaluation of oligonucleotide efficacy in vivo.


Asunto(s)
Técnicas de Silenciamiento del Gen , ARN Interferente Pequeño/genética , Animales , Expresión Génica , Silenciador del Gen , Ensayos Analíticos de Alto Rendimiento , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/uso terapéutico , Reproducibilidad de los Resultados
10.
J Huntingtons Dis ; 2(4): 491-500, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25062733

RESUMEN

BACKGROUND: Huntington's disease is caused by expansion of CAG trinucleotide repeats in the first exon of the huntingtin gene, which is essential for both development and neurogenesis. Huntington's disease is autosomal dominant. The normal allele contains 6 to 35 CAG triplets (average, 18) and the mutant, disease-causing allele contains >36 CAG triplets (average, 42). OBJECTIVE: We examined 279 postmortem brain samples, including 148 HD and 131 non-HD controls. A total of 108 samples from 87 HD patients that are heterozygous at SNP rs362307, with a normal allele (18 to 27 CAG repeats) and a mutant allele (39 to 73 CAG repeats) were used to measure relative abundance of mutant and wild-type huntingtin mRNA. METHODS: We used allele-specific, quantitative RT-PCR based on SNP heterozygosity to estimate the relative amount of mutant versus normal huntingtin mRNA in postmortem brain samples from patients with Huntington's disease. RESULTS: In the cortex and striatum, the amount of mRNA from the mutant allele exceeds that from the normal allele in 75% of patients. In the cerebellum, no significant difference between the two alleles was evident. Brain tissues from non-HD controls show no significant difference between two alleles of huntingtin mRNAs. Allelic differences were more pronounced at early neuropathological grades (grades 1 and 2) than at late grades (grades 3 and 4). CONCLUSION: More mutant HTT than normal could arise from increased transcription of mutant HTT allele, or decreased clearance of mutant HTT mRNA, or both. An implication is that equimolar silencing of both alleles would increase the mutant HTT to normal HTT ratio.


Asunto(s)
Encéfalo/metabolismo , Enfermedad de Huntington/genética , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , Adulto , Anciano , Anciano de 80 o más Años , Desequilibrio Alélico , Femenino , Humanos , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Expansión de Repetición de Trinucleótido
11.
Curr Biol ; 19(9): 774-8, 2009 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-19361997

RESUMEN

Among dominant neurodegenerative disorders, Huntington's disease (HD) is perhaps the best candidate for treatment with small interfering RNAs (siRNAs) [1-9]. Invariably fatal, HD is caused by expansion of a CAG repeat in the Huntingtin gene, creating an extended polyglutamine tract that makes the Huntingtin protein toxic [10]. Silencing mutant Huntingtin messenger RNA (mRNA) should provide therapeutic benefit, but normal Huntingtin likely contributes to neuronal function [11-13]. No siRNA strategy can yet distinguish among the normal and disease Huntingtin alleles and other mRNAs containing CAG repeats [14]. siRNAs targeting the disease isoform of a heterozygous single-nucleotide polymorphism (SNP) in Huntingtin provide an alternative [15-19]. We sequenced 22 predicted SNP sites in 225 human samples corresponding to HD and control subjects. We find that 48% of our patient population is heterozygous at a single SNP site; one isoform of this SNP is associated with HD. Several other SNP sites are frequently heterozygous. Consequently, five allele-specific siRNAs, corresponding to just three SNP sites, could be used to treat three-quarters of the United States and European HD patient populations. We have designed and validated selective siRNAs for the three SNP sites, laying the foundation for allele-specific RNA interference (RNAi) therapy for HD.


Asunto(s)
Terapia Genética/métodos , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Tamización de Portadores Genéticos , Humanos , Proteína Huntingtina , Análisis de Secuencia de ADN
12.
Kidney Int ; 66(5): 1890-900, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15496160

RESUMEN

BACKGROUND: In cultured renal epithelial cells, exposure to oxalate, a constituent of many kidney stones, elicits a cascade of responses that often leads to cell death. Oxalate toxicity is mediated via generation of reactive oxygen species (ROS) in a process that depends at least in part upon lipid signaling molecules that are generated through membrane events that culminate in phospholipase A2 (PLA2) activation. The present studies asked whether mitochondria, a major site of ROS production, were targets of oxalate toxicity, and if so, whether mitochondrial responses to oxalate were mediated by PLA2 activation. METHODS: Effects of oxalate and various lipids on mitochondrial membrane potential (DeltaPsim) were measured in Madin-Darby canine kidney (MDCK) cell monolayers using 5,5',6,6'-tetrachloro 1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1), a DeltaPsim-sensitive dye. Other studies assayed caspases, serine proteases activated during apoptosis, in response to oxalate or lipid signaling molecules. Additional studies asked whether oxalate or lipids produced by PLA2 activation promoted ROS formation in isolated renal mitochondria. RESULTS: Oxalate exposure decreased MDCK cell DeltaPsim within 30 minutes, a response attenuated by arachidonyl trifluoromethyl ketone (AACOCF3), an inhibitor of cytosolic PLA2 (cPLA2). Exposure to arachidonic acid or to lysophosphatidylcholine (lyso-PC), lipid products of PLA2 activation, or to ceramide, another lipid signal generated in MDCK cells following oxalate exposure, also depolarized MDCK cell DeltaPsim and increased the number of caspase-positive cells. Isolated renal mitochondria responded to oxalate, arachidonic acid, lyso-PC, and ceramide by increasing their accumulation of ROS, lipid peroxides, and oxidized thiol proteins. CONCLUSION: These studies suggest that lipid signaling molecules released after oxalate-induced PLA2 activation trigger marked, rapid changes in mitochondrial function that may mediate toxicity in renal epithelial cells.


Asunto(s)
Riñón/efectos de los fármacos , Riñón/fisiopatología , Mitocondrias/efectos de los fármacos , Oxalatos/envenenamiento , Esfingosina/análogos & derivados , Animales , Ácido Araquidónico/farmacología , Ácidos Araquidónicos/farmacología , Caspasas/metabolismo , Línea Celular , Perros , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Membranas Intracelulares/efectos de los fármacos , Riñón/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Lisofosfatidilcolinas/farmacología , Potenciales de la Membrana/efectos de los fármacos , Mitocondrias/metabolismo , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A2 , Especies Reactivas de Oxígeno/metabolismo , Esfingosina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA