Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 32(7): 2190-2206, 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-38796705

RESUMO

X-linked adrenoleukodystrophy (ALD), an inherited neurometabolic disorder caused by mutations in ABCD1, which encodes the peroxisomal ABC transporter, mainly affects the brain, spinal cord, adrenal glands, and testes. In ALD patients, very-long-chain fatty acids (VLCFAs) fail to enter the peroxisome and undergo subsequent ß-oxidation, resulting in their accumulation in the body. It has not been tested whether in vivo base editing or prime editing can be harnessed to ameliorate ALD. We developed a humanized mouse model of ALD by inserting a human cDNA containing the pathogenic variant into the mouse Abcd1 locus. The humanized ALD model showed increased levels of VLCFAs. To correct the mutation, we tested both base editing and prime editing and found that base editing using ABE8e(V106W) could correct the mutation in patient-derived fibroblasts at an efficiency of 7.4%. Adeno-associated virus (AAV)-mediated systemic delivery of NG-ABE8e(V106W) enabled robust correction of the pathogenic variant in the mouse brain (correction efficiency: ∼5.5%), spinal cord (∼5.1%), and adrenal gland (∼2%), leading to a significant reduction in the plasma levels of C26:0/C22:0. This established humanized mouse model and the successful correction of the pathogenic variant using a base editor serve as a significant step toward treating human ALD disease.


Assuntos
Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP , Adrenoleucodistrofia , Dependovirus , Modelos Animais de Doenças , Edição de Genes , Terapia Genética , Animais , Adrenoleucodistrofia/terapia , Adrenoleucodistrofia/genética , Camundongos , Humanos , Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP/genética , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/administração & dosagem , Adenina , Mutação , Fibroblastos/metabolismo , Ácidos Graxos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia
2.
Theranostics ; 12(5): 2465-2482, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35265220

RESUMO

Outer hair cell (OHC) degeneration is a major cause of progressive hearing loss and presbycusis. Despite the high prevalence of these disorders, targeted therapy is currently not available. Methods: We generated a mouse model harboring Kcnq4W276S/+ to recapitulate DFNA2, a common genetic form of progressive hearing loss accompanied by OHC degeneration. After comprehensive optimization of guide RNAs, Cas9s, vehicles, and delivery routes, we applied in vivo gene editing strategy to disrupt the dominant-negative allele in Kcnq4 and prevent progressive hearing loss. Results:In vivo gene editing using a dual adeno-associated virus package targeting OHCs significantly improved auditory thresholds in auditory brainstem response and distortion-product otoacoustic emission. In addition, we developed a new live-cell imaging technique using thallium ions to investigate the membrane potential of OHCs and successfully demonstrated that mutant allele disruption resulted in more hyperpolarized OHCs, indicating elevated KCNQ4 channel activity. Conclusion: These findings can facilitate the development of targeted therapies for DFNA2 and support the use of CRISPR-based gene therapy to rectify defects in OHCs.


Assuntos
Edição de Genes , Perda Auditiva , Animais , Modelos Animais de Doenças , Células Ciliadas Auditivas Externas/metabolismo , Perda Auditiva/genética , Perda Auditiva/metabolismo , Perda Auditiva/terapia , Canais de Potássio KCNQ/genética , Canais de Potássio KCNQ/metabolismo , Potenciais da Membrana , Camundongos
3.
Nat Biomed Eng ; 6(2): 181-194, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34446856

RESUMO

The use of prime editing-a gene-editing technique that induces small genetic changes without the need for donor DNA and without causing double strand breaks-to correct pathogenic mutations and phenotypes needs to be tested in animal models of human genetic diseases. Here we report the use of prime editors 2 and 3, delivered by hydrodynamic injection, in mice with the genetic liver disease hereditary tyrosinemia, and of prime editor 2, delivered by an adeno-associated virus vector, in mice with the genetic eye disease Leber congenital amaurosis. For each pathogenic mutation, we identified an optimal prime-editing guide RNA by using cells transduced with lentiviral libraries of guide-RNA-encoding sequences paired with the corresponding target sequences. The prime editors precisely corrected the disease-causing mutations and led to the amelioration of the disease phenotypes in the mice, without detectable off-target edits. Prime editing should be tested further in more animal models of genetic diseases.


Assuntos
Oftalmopatias , Edição de Genes , Animais , Edição de Genes/métodos , Fígado , Camundongos , Mutação , Fenótipo
5.
Am J Hum Genet ; 100(3): 454-472, 2017 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-28215400

RESUMO

Focal cortical dysplasia (FCD) is a major cause of the sporadic form of intractable focal epilepsies that require surgical treatment. It has recently been reported that brain somatic mutations in MTOR account for 15%-25% of FCD type II (FCDII), characterized by cortical dyslamination and dysmorphic neurons. However, the genetic etiologies of FCDII-affected individuals who lack the MTOR mutation remain unclear. Here, we performed deep hybrid capture and amplicon sequencing (read depth of 100×-20,012×) of five important mTOR pathway genes-PIK3CA, PIK3R2, AKT3, TSC1, and TSC2-by using paired brain and saliva samples from 40 FCDII individuals negative for MTOR mutations. We found that 5 of 40 individuals (12.5%) had brain somatic mutations in TSC1 (c.64C>T [p.Arg22Trp] and c.610C>T [p.Arg204Cys]) and TSC2 (c.4639G>A [p.Val1547Ile]), and these results were reproducible on two different sequencing platforms. All identified mutations induced hyperactivation of the mTOR pathway by disrupting the formation or function of the TSC1-TSC2 complex. Furthermore, in utero CRISPR-Cas9-mediated genome editing of Tsc1 or Tsc2 induced the development of spontaneous behavioral seizures, as well as cytomegalic neurons and cortical dyslamination. These results show that brain somatic mutations in TSC1 and TSC2 cause FCD and that in utero application of the CRISPR-Cas9 system is useful for generating neurodevelopmental disease models of somatic mutations in the brain.


Assuntos
Epilepsia/genética , Malformações do Desenvolvimento Cortical do Grupo I/genética , Proteínas Supressoras de Tumor/genética , Adolescente , Animais , Encéfalo/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Criança , Classe I de Fosfatidilinositol 3-Quinases , Clonagem Molecular , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Mutação , Neurônios , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Saliva/química , Análise de Sequência de DNA , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/genética , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa
6.
Genome Res ; 24(6): 1020-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24696462

RESUMO

RNA-guided endonucleases (RGENs) derived from the CRISPR/Cas system represent an efficient tool for genome editing. RGENs consist of two components: Cas9 protein and guide RNA. Plasmid-mediated delivery of these components into cells can result in uncontrolled integration of the plasmid sequence into the host genome, and unwanted immune responses and potential safety problems that can be caused by the bacterial sequences. Furthermore, this delivery method requires transfection tools. Here we show that simple treatment with cell-penetrating peptide (CPP)-conjugated recombinant Cas9 protein and CPP-complexed guide RNAs leads to endogenous gene disruptions in human cell lines. The Cas9 protein was conjugated to CPP via a thioether bond, whereas the guide RNA was complexed with CPP, forming condensed, positively charged nanoparticles. Simultaneous and sequential treatment of human cells, including embryonic stem cells, dermal fibroblasts, HEK293T cells, HeLa cells, and embryonic carcinoma cells, with the modified Cas9 and guide RNA, leads to efficient gene disruptions with reduced off-target mutations relative to plasmid transfections, resulting in the generation of clones containing RGEN-induced mutations. Our CPP-mediated RGEN delivery process provides a plasmid-free and additional transfection reagent-free method to use this tool with reduced off-target effects. We envision that our method will facilitate RGEN-directed genome editing.


Assuntos
Sistemas CRISPR-Cas , Peptídeos Penetradores de Células/química , RNA Guia de Cinetoplastídeos/genética , Transfecção/métodos , Linhagem Celular Tumoral , Genoma Humano , Células HEK293 , Humanos , Mutagênese Sítio-Dirigida/métodos , Nanopartículas/química , Edição de RNA , RNA Guia de Cinetoplastídeos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA