Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Ther Nucleic Acids ; 34: 102057, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37928442

RESUMO

Toxic gain-of-function mutations in superoxide dismutase 1 (SOD1) contribute to approximately 2%-3% of all amyotrophic lateral sclerosis (ALS) cases. Artificial microRNAs (amiRs) delivered by adeno-associated virus (AAV) have been proposed as a potential treatment option to silence SOD1 expression and mitigate disease progression. Primary microRNA (pri-miRNA) scaffolds are used in amiRs to shuttle a hairpin RNA into the endogenous miRNA pathway, but it is unclear whether different primary miRNA (pri-miRNA) scaffolds impact the potency and safety profile of the expressed amiR in vivo. In our process to develop an AAV amiR targeting SOD1, we performed a preclinical characterization of two pri-miRNA scaffolds, miR155 and miR30a, sharing the same guide strand sequence. We report that, while the miR155-based vector, compared with the miR30a-based vector, leads to a higher level of the amiR and more robust suppression of SOD1 in vitro and in vivo, it also presents significantly greater risks for CNS-related toxicities in vivo. Despite miR30a-based vector showing relatively lower potency, it can significantly delay the development of ALS-like phenotypes in SOD1-G93A mice and increase survival in a dose-dependent manner. These data highlight the importance of scaffold selection in the pursuit of highly efficacious and safe amiRs for RNA interference gene therapy.

2.
Gene Ther ; 30(5): 443-454, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36450833

RESUMO

CRISPR-based gene editing technology represents a promising approach to deliver therapies for inherited disorders, including amyotrophic lateral sclerosis (ALS). Toxic gain-of-function superoxide dismutase 1 (SOD1) mutations are responsible for ~20% of familial ALS cases. Thus, current clinical strategies to treat SOD1-ALS are designed to lower SOD1 levels. Here, we utilized AAV-PHP.B variants to deliver CRISPR-Cas9 guide RNAs designed to disrupt the human SOD1 (huSOD1) transgene in SOD1G93A mice. A one-time intracerebroventricular injection of AAV.PHP.B-huSOD1-sgRNA into neonatal H11Cas9 SOD1G93A mice caused robust and sustained mutant huSOD1 protein reduction in the cortex and spinal cord, and restored motor function. Neonatal treatment also reduced spinal motor neuron loss, denervation at neuromuscular junction (NMJ) and muscle atrophy, diminished axonal damage and preserved compound muscle action potential throughout the lifespan of treated mice. SOD1G93A treated mice achieved significant disease-free survival, extending lifespan by more than 110 days. Importantly, a one-time intrathecal or intravenous injection of AAV.PHP.eB-huSOD1-sgRNA in adult H11Cas9 SOD1G93A mice, immediately before symptom onset, also extended lifespan by at least 170 days. We observed substantial protection against disease progression, demonstrating the utility of our CRISPR editing preclinical approach for target evaluation. Our approach uncovered key parameters (e.g., AAV capsid, Cas9 expression) that resulted in improved efficacy compared to similar approaches and can also serve to accelerate drug target validation.


Assuntos
Esclerose Lateral Amiotrófica , Camundongos , Humanos , Animais , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/terapia , Superóxido Dismutase-1/genética , Edição de Genes , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Camundongos Transgênicos , Modelos Animais de Doenças
3.
Gene Ther ; 28(10-11): 646-658, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33558692

RESUMO

CRISPR-Cas systems have emerged as a powerful tool to generate genetic models for studying normal and diseased central nervous system (CNS). Targeted gene disruption at specific loci has been demonstrated successfully in non-dividing neurons. Despite its simplicity, high specificity and low cost, the efficiency of CRISPR-mediated knockout in vivo can be substantially impacted by many parameters. Here, we used CRISPR-Cas9 to disrupt the neuronal-specific gene, NeuN, and optimized key parameters to achieve effective gene knockout broadly in the CNS in postnatal mice. Three cell lines and two primary neuron cultures were used to validate the disruption of NeuN by single-guide RNAs (sgRNA) harboring distinct spacers and scaffold sequences. This triage identified an optimal sgRNA design with the highest NeuN disruption in in vitro and in vivo systems. To enhance CRISPR efficiency, AAV-PHP.B, a vector with superior neuronal transduction, was used to deliver this sgRNA in Cas9 mice via neonatal intracerebroventricular (ICV) injection. This approach resulted in 99.4% biallelic indels rate in the transduced cells, leading to greater than 70% reduction of total NeuN proteins in the cortex, hippocampus and spinal cord. This work contributes to the optimization of CRISPR-mediated knockout and will be beneficial for fundamental and preclinical research.


Assuntos
Sistemas CRISPR-Cas , RNA Guia de Cinetoplastídeos , Animais , Sistema Nervoso Central , Edição de Genes/métodos , Técnicas de Inativação de Genes , Camundongos , Neurônios/metabolismo , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo
4.
Gene Ther ; 28(7-8): 456-468, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33612827

RESUMO

Adeno-associated virus (AAV) transduction efficiency and tropism are conventionally determined by high expression of a fluorescent reporter gene. Emerging data has suggested that such conventional methods may underestimate AAV transduction for cells in which reporter expression from AAV vectors is undetectable. To explore an alternative method that captures AAV transduction in cells in which low expression of a cargo is sufficient for the intended activity, we sought after CRISPR/Cas9-mediated gene disruption. In this study, we use AAV to deliver CRISPR/guide RNA designed to abolish the genes NeuN, GFAP, or MOG expressed specifically in neurons, astrocytes, or oligodendrocytes respectively in the central nervous system (CNS) of mice. Abrogated expression of these cell-type-specific genes can be measured biochemically in CNS subregions and provides quantitative assessment of AAV transduction in these CNS cell types. By using this method, we compared CNS transduction of AAV9, AAV-PHP.B, and AAV-PHP.eB delivered via intracerebroventricular injection (ICV) in neonatal mice. We found both AAV-PHP.B and AAV-PHP.eB resulted in marked disruption of the NeuN gene by CRISPR/Cas9, significantly greater than AAV9 in several brain regions and spinal cord. In contrast, only modest disruption of the GFAP gene and the MOG gene was observed by all three AAV variants. Since the procedure of ICV circumvents the blood-brain barrier, our data suggests that, independent of their ability to cross the blood-brain barrier, AAV-PHP.B variants also exhibit remarkably improved neuronal transduction in the CNS. We anticipate this approach will facilitate profiling of AAV cellular tropism in murine CNS.


Assuntos
Dependovirus , Vetores Genéticos , Animais , Sistemas CRISPR-Cas , Sistema Nervoso Central , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Camundongos , Neurônios , Transdução Genética
5.
Sci Transl Med ; 8(347): 347ra93, 2016 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-27412785

RESUMO

C9ORF72 mutations are found in a significant fraction of patients suffering from amyotrophic lateral sclerosis and frontotemporal dementia, yet the function of the C9ORF72 gene product remains poorly understood. We show that mice harboring loss-of-function mutations in the ortholog of C9ORF72 develop splenomegaly, neutrophilia, thrombocytopenia, increased expression of inflammatory cytokines, and severe autoimmunity, ultimately leading to a high mortality rate. Transplantation of mutant mouse bone marrow into wild-type recipients was sufficient to recapitulate the phenotypes observed in the mutant animals, including autoimmunity and premature mortality. Reciprocally, transplantation of wild-type mouse bone marrow into mutant mice improved their phenotype. We conclude that C9ORF72 serves an important function within the hematopoietic system to restrict inflammation and the development of autoimmunity.


Assuntos
Doenças Autoimunes/etiologia , Doenças Autoimunes/genética , Proteína C9orf72/genética , Animais , Doenças Autoimunes/metabolismo , Autoimunidade/genética , Autoimunidade/fisiologia , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/fisiologia , Citocinas/metabolismo , Leucemia/genética , Leucemia/metabolismo , Camundongos , Mutação/genética , Esplenomegalia/genética , Esplenomegalia/imunologia , Trombocitopenia/genética , Trombocitopenia/imunologia
6.
Hum Mol Genet ; 25(9): 1814-23, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26976849

RESUMO

Riboflavin, also known as vitamin B2, is essential for cellular reduction-oxidation reactions, but is not readily synthesized by mammalian cells. It has been proposed that riboflavin absorption occurs through solute carrier family 52 members (SLC52) A1, A2 and A3. These transporters are also candidate genes for the childhood onset-neural degenerative syndrome Brown-Vialetto-Van Laere (BVVL). Although riboflavin is an essential nutrient, why mutations in its transporters result in a neural cell-specific disorder remains unclear. Here, we provide evidence that Slc52a3 is the mouse ortholog of SLC52A3 and show that Slc52a3 deficiency results in early embryonic lethality. Loss of mutant embryos was associated with both defects in placental formation and increased rates of apoptosis in embryonic cells. In contrast, Slc52a3 -/- embryonic stem cell lines could be readily established and differentiated into motor neurons, suggesting that this transporter is dispensable for neural differentiation and short-term maintenance. Consistent with this finding, examination of Slc52a3 gene products in adult tissues revealed expression in the testis and intestine but little or none in the brain and spinal cord. Our results suggest that BVVL patients with SCL52A3 mutations may be good candidates for riboflavin replacement therapy and suggests that either the mutations these individuals carry are hypomorphic, or that in these cases alternative transporters act during human embryogenesis to allow full-term development.


Assuntos
Paralisia Bulbar Progressiva/genética , Paralisia Bulbar Progressiva/patologia , Diferenciação Celular , Embrião de Mamíferos/citologia , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Proteínas de Membrana Transportadoras/metabolismo , Neurônios Motores/citologia , Mutação/genética , Animais , Células Cultivadas , Embrião de Mamíferos/metabolismo , Feminino , Humanos , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Knockout , Neurônios Motores/metabolismo , Neurogênese
7.
Sci Transl Med ; 6(248): 248ra104, 2014 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-25100738

RESUMO

Neurons produced from stem cells have emerged as a tool to identify new therapeutic targets for neurological diseases such as amyotrophic lateral sclerosis (ALS). However, it remains unclear to what extent these new mechanistic insights will translate to animal models, an important step in the validation of new targets. Previously, we found that glia from mice carrying the SOD1G93A mutation, a model of ALS, were toxic to stem cell-derived human motor neurons. We use pharmacological and genetic approaches to demonstrate that the prostanoid receptor DP1 mediates this glial toxicity. Furthermore, we validate the importance of this mechanism for neural degeneration in vivo. Genetic ablation of DP1 in SOD1G93A mice extended life span, decreased microglial activation, and reduced motor neuron loss. Our findings suggest that blocking DP1 may be a therapeutic strategy in ALS and demonstrate that discoveries from stem cell models of disease can be corroborated in vivo.


Assuntos
Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/terapia , Modelos Animais de Doenças , Terapia de Alvo Molecular , Animais , Técnicas de Cocultura , Citoproteção , Progressão da Doença , Humanos , Longevidade , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Mutação/genética , Neuroglia/patologia , Receptores de Prostaglandina/metabolismo , Reprodutibilidade dos Testes
8.
Stem Cell Reports ; 2(2): 119-26, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24527385

RESUMO

It has been suggested that the transcription factor Nanog is essential for the establishment of pluripotency during the derivation of embryonic stem cells and induced pluripotent stem cells (iPSCs). However, successful reprogramming to pluripotency with a growing list of divergent transcription factors, at ever-increasing efficiencies, suggests that there may be many distinct routes to a pluripotent state. Here, we have investigated whether Nanog is necessary for reprogramming murine fibroblasts under highly efficient conditions using the canonical-reprogramming factors Oct4, Sox2, Klf4, and cMyc. In agreement with prior results, the efficiency of reprogramming Nanog (-/-) fibroblasts was significantly lower than that of control fibroblasts. However, in contrast to previous findings, we were able to reproducibly generate iPSCs from Nanog (-/-) fibroblasts that effectively contributed to the germline of chimeric mice. Thus, whereas Nanog may be an important mediator of reprogramming, it is not required for establishing pluripotency in the mouse, even under standard conditions.


Assuntos
Reprogramação Celular/genética , Proteínas de Homeodomínio/genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Diferenciação Celular/genética , Quimera/genética , Análise por Conglomerados , Feminino , Fibroblastos/metabolismo , Expressão Gênica , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Fator 4 Semelhante a Kruppel , Masculino , Camundongos , Proteína Homeobox Nanog , Fenótipo , Fatores de Transcrição/genética , Transcriptoma , Transdução Genética
9.
Nat Neurosci ; 16(12): 1725-7, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24185425

RESUMO

Using transgenic mice harboring a targeted LacZ insertion, we studied the expression pattern of the C9ORF72 mouse ortholog (3110043O21Rik). Unlike most genes that are mutated in amyotrophic lateral sclerosis (ALS), which are ubiquitously expressed, the C9ORF72 ortholog was most highly transcribed in the neuronal populations that are sensitive to degeneration in ALS and frontotemporal dementia. Thus, our results provide a potential explanation for the cell type specificity of neuronal degeneration caused by C9ORF72 mutations.


Assuntos
Esclerose Lateral Amiotrófica/genética , Encéfalo/patologia , Demência Frontotemporal/genética , Regulação da Expressão Gênica/genética , Mutação/genética , Neurônios/patologia , Proteínas/genética , Acetilcolinesterase/metabolismo , Idoso , Esclerose Lateral Amiotrófica/patologia , Animais , Animais Recém-Nascidos , Proteína C9orf72 , Células Cultivadas , Embrião de Mamíferos , Demência Frontotemporal/patologia , Genótipo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição , Transfecção
10.
Cell Stem Cell ; 5(5): 491-503, 2009 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-19818703

RESUMO

The combined activity of three transcription factors can reprogram adult cells into induced pluripotent stem cells (iPSCs). However, the transgenic methods used for delivering reprogramming factors have raised concerns regarding the future utility of the resulting stem cells. These uncertainties could be overcome if each transgenic factor were replaced with a small molecule that either directly activated its expression from the somatic genome or in some way compensated for its activity. To this end, we have used high-content chemical screening to identify small molecules that can replace Sox2 in reprogramming. We show that one of these molecules functions in reprogramming by inhibiting Tgf-beta signaling in a stable and trapped intermediate cell type that forms during the process. We find that this inhibition promotes the completion of reprogramming through induction of the transcription factor Nanog.


Assuntos
Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Pirazóis/farmacologia , Piridinas/farmacologia , Fatores de Transcrição SOXB1/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Anticorpos Monoclonais , Benzamidas/farmacologia , Linhagem Celular , Transdiferenciação Celular , Dioxóis/farmacologia , Ensaios de Triagem em Larga Escala , Proteínas de Homeodomínio/genética , Células-Tronco Pluripotentes Induzidas/imunologia , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Proteína Homeobox Nanog , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Fatores de Transcrição SOXB1/genética , Transdução de Sinais , Transdução Genética , Fator de Crescimento Transformador beta/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA