Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell ; 167(5): 1144, 2016 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-27863231

RESUMO

Exondys 51 is the first therapy for Duchenne muscular dystrophy (DMD) to have been granted accelerated approval by the FDA. Approval was granted based on using dystrophin expression as a surrogate marker. Exondys 51 targets DMD exon 51 for skipping to restore the reading frame for 13% of Duchenne patients.


Assuntos
Distrofina/genética , Terapia Genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Éxons , Humanos , Fases de Leitura , Estados Unidos , United States Food and Drug Administration
2.
J Nanobiotechnology ; 21(1): 303, 2023 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-37641124

RESUMO

Skeletal muscle disease severity can often progress asymmetrically across muscle groups and heterogeneously within tissues. An example is Duchenne Muscular Dystrophy (DMD) in which lack of dystrophin results in devastating skeletal muscle wasting in some muscles whereas others are spared or undergo hypertrophy. An efficient, non-invasive approach to identify sites of asymmetry and degenerative lesions could enable better patient monitoring and therapeutic targeting of disease. In this study, we utilized a versatile intravenously injectable mesoporous silica nanoparticle (MSNP) based nanocarrier system to explore mechanisms of biodistribution in skeletal muscle of mdx mouse models of DMD including wildtype, dystrophic, and severely dystrophic mice. Moreover, MSNPs could be imaged in live mice and whole muscle tissues enabling investigation of how biodistribution is altered by different types of muscle pathology such as inflammation or fibrosis. We found MSNPs were tenfold more likely to aggregate within select mdx muscles relative to wild type, such as gastrocnemius and quadriceps. This was accompanied by decreased biodistribution in off-target organs. We found the greatest factor affecting preferential delivery was the regenerative state of the dystrophic skeletal muscle with the highest MSNP abundance coinciding with the regions showing the highest level of embryonic myosin staining and intramuscular macrophage uptake. To demonstrate, muscle regeneration regulated MSNP distribution, we experimentally induced regeneration using barium chloride which resulted in a threefold increase of intravenously injected MSNPs to sites of regeneration 7 days after injury. These discoveries provide the first evidence that nanoparticles have selective biodistribution to skeletal muscle in DMD to areas of active regeneration and that nanoparticles could enable diagnostic and selective drug delivery in DMD skeletal muscle.


Assuntos
Distrofina , Músculo Esquelético , Animais , Camundongos , Distribuição Tecidual , Camundongos Endogâmicos mdx , Regeneração
3.
Physiology (Bethesda) ; 34(5): 341-353, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31389773

RESUMO

This is a review describing advances in CRISPR/Cas-mediated therapies for neuromuscular disorders (NMDs). We explore both CRISPR-mediated editing and dead Cas approaches as potential therapeutic strategies for multiple NMDs. Last, therapeutic considerations, including delivery and off-target effects, are also discussed.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Doenças Neuromusculares/genética , Animais , Edição de Genes/métodos , Humanos
4.
Mol Ther ; 24(9): 1561-9, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27406980

RESUMO

Targeted genome editing technology can correct the sickle cell disease mutation of the ß-globin gene in hematopoietic stem cells. This correction supports production of red blood cells that synthesize normal hemoglobin proteins. Here, we demonstrate that Transcription Activator-Like Effector Nucleases (TALENs) and the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 nuclease system can target DNA sequences around the sickle-cell mutation in the ß-globin gene for site-specific cleavage and facilitate precise correction when a homologous donor template is codelivered. Several pairs of TALENs and multiple CRISPR guide RNAs were evaluated for both on-target and off-target cleavage rates. Delivery of the CRISPR/Cas9 components to CD34+ cells led to over 18% gene modification in vitro. Additionally, we demonstrate the correction of the sickle cell disease mutation in bone marrow derived CD34+ hematopoietic stem and progenitor cells from sickle cell disease patients, leading to the production of wild-type hemoglobin. These results demonstrate correction of the sickle mutation in patient-derived CD34+ cells using CRISPR/Cas9 technology.


Assuntos
Anemia Falciforme/genética , Sistemas CRISPR-Cas , Edição de Genes , Células-Tronco Hematopoéticas/metabolismo , Mutação , Reparo Gênico Alvo-Dirigido , Globinas beta/genética , Anemia Falciforme/terapia , Sequência de Bases , Linhagem Celular , Clivagem do DNA , Marcação de Genes , Loci Gênicos , Humanos , Ligação Proteica , RNA Guia de Cinetoplastídeos , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/metabolismo
5.
Mol Ther Methods Clin Dev ; 30: 90-102, 2023 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-37746243

RESUMO

High systemic doses of adeno-associated viruses (AAVs) have been associated with immune-related serious adverse events (SAEs). Although AAV was well tolerated in preclinical models, SAEs were observed in clinical trials, indicating the need for improved preclinical models to understand AAV-induced immune responses. Here, we show that mice dual-dosed with AAV9 at 4-week intervals better recapitulate aspects of human immunity to AAV. In the model, anti-AAV9 immunoglobulin G (IgGs) increased in a linear fashion between the first and second AAV administrations. Complement activation was only observed in the presence of high levels of both AAV and anti-AAV IgG. Myeloid-derived pro-inflammatory cytokines were significantly induced in the same pattern as complement activation, suggesting that myeloid cell activation to AAV may rely on the presence of both AAV and anti-AAV IgG complexes. Single-cell RNA sequencing of peripheral blood mononuclear cells confirmed that activated monocytes were a primary source of pro-inflammatory cytokines and chemokines, which were significantly increased after a second AAV9 exposure. The same activated monocyte clusters expressed both Fcγ and complement receptors, suggesting that anti-AAV-mediated activation of myeloid cells through Fcγ receptors and/or complement receptors is one mechanism by which anti-AAV antigen complexes may prime antigen-presenting cells and amplify downstream immunity.

6.
Pharm Res ; 27(5): 841-54, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20224990

RESUMO

PURPOSE: Interleukin-10 (IL-10) is an anti-inflammatory molecule that has achieved interest as a therapeutic for neuropathic pain. In this work, the potential of plasmid DNA-encoding IL-10 (pDNA-IL-10) slowly released from biodegradable microparticles to provide long-term pain relief in an animal model of neuropathic pain was investigated. METHODS: PLGA microparticles encapsulating pDNA-IL-10 were developed and assessed both in vitro and in vivo. RESULTS: In vitro, pDNA containing microparticles activated macrophages, enhanced the production of nitric oxide, and increased the production of IL-10 protein relative to levels achieved with unencapsulated pDNA-IL-10. In vivo, intrathecally administered microparticles embedded in meningeal tissue, induced phagocytic cell recruitment to the cerebrospinal fluid, and relieved neuropathic pain for greater than 74 days following a single intrathecal administration, a feat not achieved with unencapsulated pDNA. Therapeutic effects of microparticle-delivered pDNA-IL-10 were blocked in the presence of IL-10-neutralizing antibody, and elevated levels of plasmid-derived IL-10 were detected in tissues for a prolonged time period post-injection (>28 days), demonstrating that therapeutic effects are dependent on IL-10 protein production. CONCLUSIONS: These studies demonstrate that microparticle encapsulation significantly enhances the potency of intrathecally administered pDNA, which may be extended to treat other disorders that require intrathecal gene therapy.


Assuntos
DNA/administração & dosagem , DNA/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Interleucina-10/genética , Doenças do Sistema Nervoso Periférico/terapia , Plasmídeos/genética , Animais , Comportamento Animal/fisiologia , Células Cultivadas , Imuno-Histoquímica , Injeções Espinhais , Interleucina-10/biossíntese , Ácido Láctico , Macrófagos/metabolismo , Masculino , Nanopartículas , Óxido Nítrico/metabolismo , Tamanho da Partícula , Doenças do Sistema Nervoso Periférico/líquido cefalorraquidiano , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
STAR Protoc ; 1(3): 100158, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33377052

RESUMO

This protocol describes the use of CRISPR/Cas9-mediated homology-directed recombination to construct a PAX7-GFP reporter in human pluripotent stem cells (hPSCs). PAX7 is a key transcription factor and regulator of skeletal muscle stem/progenitor cells. We obtained heterozygous knockin reporter cells and validated their PAX7 expression using both artificial activation by the CRISPR/dCas9-VPR system and physiological activation during hPSC myogenic differentiation. These cells can serve as tools for better understanding of in vitro hPSC myogenesis and enriching myogenic cells for downstream analysis. For complete details on the use and execution of this protocol, please refer to Xi et al. (2017) and Xi et al. (2020).


Assuntos
Genes Reporter , Desenvolvimento Muscular , Fator de Transcrição PAX7/metabolismo , Células-Tronco Pluripotentes/metabolismo , Regiões 3' não Traduzidas/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Contagem de Células , Diferenciação Celular , Sequência Conservada , Resistência Microbiana a Medicamentos , Genótipo , Humanos , Mamíferos , Mesoderma/embriologia , MicroRNAs/genética , MicroRNAs/metabolismo , Fator de Transcrição PAX7/química , Plasmídeos/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , RNA Guia de Cinetoplastídeos/genética , Reprodutibilidade dos Testes , Somitos/embriologia
8.
Cell Stem Cell ; 27(1): 158-176.e10, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32396864

RESUMO

The developmental trajectory of human skeletal myogenesis and the transition between progenitor and stem cell states are unclear. We used single-cell RNA sequencing to profile human skeletal muscle tissues from embryonic, fetal, and postnatal stages. In silico, we identified myogenic as well as other cell types and constructed a "roadmap" of human skeletal muscle ontogeny across development. In a similar fashion, we also profiled the heterogeneous cell cultures generated from multiple human pluripotent stem cell (hPSC) myogenic differentiation protocols and mapped hPSC-derived myogenic progenitors to an embryonic-to-fetal transition period. We found differentially enriched biological processes and discovered co-regulated gene networks and transcription factors present at distinct myogenic stages. This work serves as a resource for advancing our knowledge of human myogenesis. It also provides a tool for a better understanding of hPSC-derived myogenic progenitors for translational applications in skeletal muscle-based regenerative medicine.


Assuntos
Desenvolvimento Muscular , Células-Tronco Pluripotentes , Diferenciação Celular , Humanos , Músculo Esquelético , Fatores de Transcrição
9.
Nat Cell Biol ; 20(1): 46-57, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29255171

RESUMO

Human pluripotent stem cells (hPSCs) can be directed to differentiate into skeletal muscle progenitor cells (SMPCs). However, the myogenicity of hPSC-SMPCs relative to human fetal or adult satellite cells remains unclear. We observed that hPSC-SMPCs derived by directed differentiation are less functional in vitro and in vivo compared to human satellite cells. Using RNA sequencing, we found that the cell surface receptors ERBB3 and NGFR demarcate myogenic populations, including PAX7 progenitors in human fetal development and hPSC-SMPCs. We demonstrated that hPSC skeletal muscle is immature, but inhibition of transforming growth factor-ß signalling during differentiation improved fusion efficiency, ultrastructural organization and the expression of adult myosins. This enrichment and maturation strategy restored dystrophin in hundreds of dystrophin-deficient myofibres after engraftment of CRISPR-Cas9-corrected Duchenne muscular dystrophy human induced pluripotent stem cell-SMPCs. The work provides an in-depth characterization of human myogenesis, and identifies candidates that improve the in vivo myogenic potential of hPSC-SMPCs to levels that are equal to directly isolated human fetal muscle cells.


Assuntos
Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/genética , Mioblastos/metabolismo , Proteínas do Tecido Nervoso/genética , Receptor ErbB-3/genética , Receptores de Fator de Crescimento Neural/genética , Adulto , Idoso , Sistemas CRISPR-Cas , Diferenciação Celular , Distrofina/genética , Distrofina/metabolismo , Feminino , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/citologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Mioblastos/citologia , Miosinas/genética , Miosinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição PAX7/genética , Fator de Transcrição PAX7/metabolismo , Receptor ErbB-3/metabolismo , Receptores de Fator de Crescimento Neural/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
10.
J Neuromuscul Dis ; 5(4): 407-417, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30198876

RESUMO

A new line of dystrophic mdx mice on the DBA/2J (D2) background has emerged as a candidate to study the efficacy of therapeutic approaches for Duchenne muscular dystrophy (DMD). These mice harbor genetic polymorphisms that appear to increase the severity of the dystropathology, with disease modifiers that also occur in DMD patients, making them attractive for efficacy studies and drug development. This workshop aimed at collecting and consolidating available data on the pathological features and the natural history of these new D2/mdx mice, for comparison with classic mdx mice and controls, and to identify gaps in information and their potential value. The overall aim is to establish guidance on how to best use the D2/mdx mouse model in preclinical studies.


Assuntos
Modelos Animais de Doenças , Distrofia Muscular Animal , Distrofia Muscular de Duchenne , Animais , Camundongos , Camundongos Endogâmicos DBA , Camundongos Endogâmicos mdx
11.
J Neuromuscul Dis ; 4(2): 139-145, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28505980

RESUMO

Duchenne muscular dystrophy is caused by mutations in DMD which disrupt the reading frame. Therapeutic strategies that restore DMD's reading frame, such as exon skipping and CRISPR/Cas9, need to be tested in the context of the human DMD sequence in vivo. We have developed a novel dystrophic mouse model by using CRISPR/Cas9 to delete exon 45 in the human DMD gene in hDMD mice, which places DMD out-of-frame. We have utilized this model to demonstrate that our clinically-relevant CRISPR/Cas9 platform, which targets deletion of human DMD exons 45-55, can be directly applied in vivo to restore dystrophin.


Assuntos
Modelos Animais de Doenças , Distrofina/genética , Terapia Genética , Distrofia Muscular de Duchenne/terapia , Animais , Sistemas CRISPR-Cas , Distrofina/metabolismo , Éxons , Edição de Genes/métodos , Terapia Genética/métodos , Células HEK293 , Humanos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia
12.
Cell Stem Cell ; 18(4): 533-40, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26877224

RESUMO

Mutations in DMD disrupt the reading frame, prevent dystrophin translation, and cause Duchenne muscular dystrophy (DMD). Here we describe a CRISPR/Cas9 platform applicable to 60% of DMD patient mutations. We applied the platform to DMD-derived hiPSCs where successful deletion and non-homologous end joining of up to 725 kb reframed the DMD gene. This is the largest CRISPR/Cas9-mediated deletion shown to date in DMD. Use of hiPSCs allowed evaluation of dystrophin in disease-relevant cell types. Cardiomyocytes and skeletal muscle myotubes derived from reframed hiPSC clonal lines had restored dystrophin protein. The internally deleted dystrophin was functional as demonstrated by improved membrane integrity and restoration of the dystrophin glycoprotein complex in vitro and in vivo. Furthermore, miR31 was reduced upon reframing, similar to observations in Becker muscular dystrophy. This work demonstrates the feasibility of using a single CRISPR pair to correct the reading frame for the majority of DMD patients.


Assuntos
Sistemas CRISPR-Cas/genética , Distrofina/metabolismo , Deleção de Genes , Edição de Genes/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Animais , Distrofina/deficiência , Distrofina/genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Camundongos SCID , Músculo Esquelético/citologia , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa