Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Ann Neurol ; 94(2): 398-413, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37186119

RESUMO

OBJECTIVE: Facioscapulohumeral muscular dystrophy (FSHD) is caused by abnormal de-repression of the myotoxic transcription factor DUX4. Although the transcriptional targets of DUX4 are known, the regulation of DUX4 protein and the molecular consequences of this regulation are unclear. Here, we used in vitro models of FSHD to identify and characterize DUX4 post-translational modifications (PTMs) and their impact on the toxic function of DUX4. METHODS: We immunoprecipitated DUX4 protein and performed mass spectrometry to identify PTMs. We then characterized DUX4 PTMs and potential enzyme modifiers using mutagenesis, proteomics, and biochemical assays in HEK293 and human myoblast cell lines. RESULTS: We identified 17 DUX4 amino acids with PTMs, and generated 55 DUX4 mutants designed to prevent or mimic PTMs. Five mutants protected cells against DUX4-mediated toxicity and reduced the ability of DUX4 to transactivate FSHD biomarkers. These mutagenesis results suggested that DUX4 toxicity could be counteracted by serine/threonine phosphorylation and/or inhibition of arginine methylation. We therefore sought to identify modifying enzymes that could play a role in regulating DUX4 PTMs. We found several enzymes capable of modifying DUX4 protein in vitro, and confirmed that protein kinase A (PKA) and protein arginine methyltransferase (PRMT1) interact with DUX4. INTERPRETATION: These results support that DUX4 is regulated by PTMs and set a foundation for developing FSHD drug screens based mechanistically on DUX4 PTMs and modifying enzymes. ANN NEUROL 2023;94:398-413.


Assuntos
Distrofia Muscular Facioescapuloumeral , Humanos , Regulação da Expressão Gênica , Células HEK293 , Proteínas de Homeodomínio/genética , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Processamento de Proteína Pós-Traducional , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo
2.
RNA ; 25(9): 1211-1217, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31209064

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is among the most common forms of muscular dystrophy. FSHD is caused by aberrant expression of the toxic DUX4 gene in muscle. Detecting endogenous DUX4 in patient tissue using conventional methods can be challenging, due to the low level of DUX4 expression. Therefore, developing simple and trustworthy DUX4 detection methods is an important need in the FSHD field. Here, we describe such a method, which uses the RNAscope assay, an RNA in situ hybridization (ISH) technology. We show that a custom-designed RNAscope assay can detect overexpressed DUX4 mRNA in transfected HEK293 cells and endogenous DUX4 mRNA in FSHD patient-derived myotubes. The RNAscope assay was highly sensitive for tracking reductions in DUX4 mRNA following treatment with our therapeutic mi405 microRNA, suggesting that RNAscope-based DUX4 expression assays could be developed as a prospective outcome measure in therapy trials. This study could set the stage for optimizing and developing a new, rapid RNA ISH-based molecular diagnostic assay for future clinical use in the FSHD field.


Assuntos
Proteínas de Homeodomínio/genética , Hibridização In Situ/métodos , RNA/genética , Linhagem Celular , Perfilação da Expressão Gênica/métodos , Células HEK293 , Humanos , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Patologia Molecular/métodos , RNA Mensageiro/genética
3.
Mol Ther ; 28(7): 1706-1716, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32353324

RESUMO

Developmental and epileptic encephalopathy (DEE) associated with de novo variants in the gene encoding dynamin-1 (DNM1) is a severe debilitating disease with no pharmacological remedy. Like most genetic DEEs, the majority of DNM1 patients suffer from therapy-resistant seizures and comorbidities such as intellectual disability, developmental delay, and hypotonia. We tested RNAi gene therapy in the Dnm1 fitful mouse model of DEE using a Dnm1-targeted therapeutic microRNA delivered by a self-complementary adeno-associated virus vector. Untreated or control-injected fitful mice have growth delay, severe ataxia, and lethal tonic-clonic seizures by 3 weeks of age. These major impairments are mitigated following a single treatment in newborn mice, along with key underlying cellular features including gliosis, cell death, and aberrant neuronal metabolic activity typically associated with recurrent seizures. Our results underscore the potential for RNAi gene therapy to treat DNM1 disease and other genetic DEEs where treatment would require inhibition of the pathogenic gene product.


Assuntos
Dinamina I/genética , Síndromes Epilépticas/terapia , Terapia Genética/métodos , MicroRNAs/genética , Animais , Animais Recém-Nascidos , Dependovirus/genética , Modelos Animais de Doenças , Síndromes Epilépticas/genética , Síndromes Epilépticas/patologia , Vetores Genéticos/administração & dosagem , Humanos , Infusões Intraventriculares , Camundongos , MicroRNAs/administração & dosagem , Interferência de RNA , Resultado do Tratamento
4.
Hum Mol Genet ; 25(20): 4577-4589, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28173143

RESUMO

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.


Assuntos
Redes Reguladoras de Genes , Proteínas de Homeodomínio/metabolismo , Micotoxinas/metabolismo , Mioblastos/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Evolução Molecular , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Transgênicos , Distrofia Muscular Facioescapuloumeral/metabolismo , Micotoxinas/genética , Alinhamento de Sequência , Análise de Sequência de RNA
5.
Mol Ther ; 20(7): 1417-23, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22508491

RESUMO

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.


Assuntos
Proteínas de Homeodomínio/genética , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/terapia , RNA Interferente Pequeno/uso terapêutico , Animais , Dependovirus/genética , Feminino , Terapia Genética , Vetores Genéticos , Proteínas de Homeodomínio/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/uso terapêutico , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/metabolismo , Interferência de RNA
6.
Sci Adv ; 9(27): eadd9984, 2023 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-37418531

RESUMO

Macrophages are essential for skeletal muscle homeostasis, but how their dysregulation contributes to the development of fibrosis in muscle disease remains unclear. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six clusters and unexpectedly found that none corresponded to traditional definitions of M1 or M2 macrophages. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 (gal-3) and osteopontin (Spp1). Spatial transcriptomics, computational inferences of intercellular communication, and in vitro assays indicated that macrophage-derived Spp1 regulates stromal progenitor differentiation. Gal-3+ macrophages were chronically activated in dystrophic muscle, and adoptive transfer assays showed that the gal-3+ phenotype was the dominant molecular program induced within the dystrophic milieu. Gal-3+ macrophages were also elevated in multiple human myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining their transcriptional programs and reveal Spp1 as a major regulator of macrophage and stromal progenitor interactions.


Assuntos
Macrófagos , Transcriptoma , Camundongos , Animais , Humanos , Camundongos Endogâmicos C57BL , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Fibrose
7.
bioRxiv ; 2023 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-37131694

RESUMO

The monocytic/macrophage system is essential for skeletal muscle homeostasis, but its dysregulation contributes to the pathogenesis of muscle degenerative disorders. Despite our increasing knowledge of the role of macrophages in degenerative disease, it still remains unclear how macrophages contribute to muscle fibrosis. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six novel clusters. Unexpectedly, none corresponded to traditional definitions of M1 or M2 macrophage activation. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 and spp1. Spatial transcriptomics and computational inferences of intercellular communication indicated that spp1 regulates stromal progenitor and macrophage interactions during muscular dystrophy. Galectin-3 + macrophages were chronically activated in dystrophic muscle and adoptive transfer assays showed that the galectin-3 + phenotype was the dominant molecular program induced within the dystrophic milieu. Histological examination of human muscle biopsies revealed that galectin-3 + macrophages were also elevated in multiple myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining the transcriptional programs induced in muscle macrophages, and reveal spp1 as a major regulator of macrophage and stromal progenitor interactions.

8.
Ann Neurol ; 69(3): 540-52, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21446026

RESUMO

OBJECTIVE: Facioscapulohumeral muscular dystrophy (FSHD) is associated with D4Z4 repeat contraction on human chromosome 4q35. This genetic lesion does not result in complete loss or mutation of any gene. Consequently, the pathogenic mechanisms underlying FSHD have been difficult to discern. In leading FSHD pathogenesis models, D4Z4 contractions are proposed to cause epigenetic changes, which ultimately increase expression of genes with myopathic potential. Although no gene has been conclusively linked to FSHD development, recent evidence supports a role for the D4Z4-encoded DUX4 gene in FSHD. In this study, our objective was to test the in vivo myopathic potential of DUX4. METHODS: We delivered DUX4 to zebrafish and mouse muscle by transposon-mediated transgenesis and adeno-associated viral vectors, respectively. RESULTS: Overexpression of DUX4, which encodes a transcription factor, caused abnormalities associated with muscular dystrophy in zebrafish and mice. This toxicity required DNA binding, because a DUX4 DNA binding domain mutant produced no abnormalities. Importantly, we found the myopathic effects of DUX4 were p53 dependent, as p53 inhibition mitigated DUX4 toxicity in vitro, and muscles from p53 null mice were resistant to DUX4-induced damage. INTERPRETATION: Our work demonstrates the myopathic potential of DUX4 in animal muscle. Considering previous studies showed DUX4 was elevated in FSHD patient muscles, our data support the hypothesis that DUX4 overexpression contributes to FSHD development. Moreover, we provide a p53-dependent mechanism for DUX4 toxicity that is consistent with previous studies showing p53 pathway activation in FSHD muscles. Our work justifies further investigation of DUX4 and the p53 pathway in FSHD pathogenesis.


Assuntos
Proteínas de Homeodomínio/genética , Músculo Esquelético/patologia , Doenças Musculares/genética , Proteína Supressora de Tumor p53/genética , Animais , Feminino , Técnicas de Transferência de Genes , Força da Mão/fisiologia , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Força Muscular/fisiologia , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Doenças Musculares/patologia , Doenças Musculares/fisiopatologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Peixe-Zebra
9.
Mol Ther ; 19(11): 2048-54, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21730972

RESUMO

Muscular dystrophies, and other diseases of muscle, arise from recessive and dominant gene mutations. Gene replacement strategies may be beneficial for the former, while gene silencing approaches may provide treatment for the latter. In the last two decades, muscle-directed gene therapies were primarily focused on treating recessive disorders. This disparity at least partly arose because feasible mechanisms to silence dominant disease genes lagged behind gene replacement strategies. With the discovery of RNA interference (RNAi) and its subsequent development as a promising new gene silencing tool, the landscape has changed. In this study, our objective was to demonstrate proof-of-principle for RNAi therapy of a dominant myopathy in vivo. We tested the potential of adeno-associated viral (AAV)-delivered therapeutic microRNAs, targeting the human Facioscapulohumeral muscular dystrophy (FSHD) region gene 1 (FRG1), to correct myopathic features in mice expressing toxic levels of human FRG1 (FRG1(-high) mice). We found that FRG1 gene silencing improved muscle mass, strength, and histopathological abnormalities associated with muscular dystrophy in FRG1(-high) mice, thereby demonstrating therapeutic promise for treatment of dominantly inherited myopathies using RNAi. This approach potentially applies to as many as 29 different gene mutations responsible for myopathies inherited as dominant disorders.


Assuntos
Terapia Genética , MicroRNAs , Distrofias Musculares/terapia , Proteínas Nucleares/genética , Interferência de RNA , Animais , Dependovirus/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos , Músculos/metabolismo , Músculos/patologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Fenótipo , Proteínas de Ligação a RNA , Transdução Genética
10.
FEBS Lett ; 596(20): 2644-2658, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35662006

RESUMO

DUX4 is a transcription factor required during early embryonic development in placental mammals. In this work, we provide evidence that DUX4 is a co-repressor of nuclear receptors (NRs) of progesterone (PR) and glucocorticoids (GR). The DUX4 C-ter and N-ter regions, including the nuclear localization signals and homeodomain motifs, contribute to the co-repressor activity of DUX4 on PR and GR. Immunoprecipitation studies, using total protein extracts of cells expressing tagged versions of DUX4 and GR, support that these proteins are physically associated. Our studies suggest that DUX4 could modulate gene expression by co-regulating the activity of hormone NRs. This is the first report highlighting a potential endocrine role for DUX4.


Assuntos
Distrofia Muscular Facioescapuloumeral , Feminino , Gravidez , Animais , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/metabolismo , Glucocorticoides , Progesterona , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas Correpressoras , Receptores de Glucocorticoides/genética , Sinais de Localização Nuclear , Placenta/metabolismo , Fatores de Transcrição , Receptores Citoplasmáticos e Nucleares , Mamíferos
11.
J Clin Invest ; 132(13)2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35579942

RESUMO

Charcot-Marie-Tooth disease type 1A (CMT1A), the most common inherited demyelinating peripheral neuropathy, is caused by PMP22 gene duplication. Overexpression of WT PMP22 in Schwann cells destabilizes the myelin sheath, leading to demyelination and ultimately to secondary axonal loss and disability. No treatments currently exist that modify the disease course. The most direct route to CMT1A therapy will involve reducing PMP22 to normal levels. To accomplish this, we developed a gene therapy strategy to reduce PMP22 using artificial miRNAs targeting human PMP22 and mouse Pmp22 mRNAs. Our lead therapeutic miRNA, miR871, was packaged into an adeno-associated virus 9 (AAV9) vector and delivered by lumbar intrathecal injection into C61-het mice, a model of CMT1A. AAV9-miR871 efficiently transduced Schwann cells in C61-het peripheral nerves and reduced human and mouse PMP22 mRNA and protein levels. Treatment at early and late stages of the disease significantly improved multiple functional outcome measures and nerve conduction velocities. Furthermore, myelin pathology in lumbar roots and femoral motor nerves was ameliorated. The treated mice also showed reductions in circulating biomarkers of CMT1A. Taken together, our data demonstrate that AAV9-miR871-driven silencing of PMP22 rescues a CMT1A model and provides proof of principle for treating CMT1A using a translatable gene therapy approach.


Assuntos
Doença de Charcot-Marie-Tooth , Proteínas da Mielina , Animais , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/terapia , Terapia Genética , Camundongos , Proteínas da Mielina/genética , Bainha de Mielina/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Células de Schwann/patologia
12.
Skelet Muscle ; 12(1): 1, 2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-35039091

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is the second most common genetic myopathy, characterized by slowly progressing and highly heterogeneous muscle wasting with a typical onset in the late teens/early adulthood [1]. Although the etiology of the disease for both FSHD type 1 and type 2 has been attributed to gain-of-toxic function stemming from aberrant DUX4 expression, the exact pathogenic mechanisms involved in muscle wasting have yet to be elucidated [2-4]. The 2021 FSHD International Research Congress, held virtually on June 24-25, convened over 350 researchers and clinicians to share the most recent advances in the understanding of the disease mechanism, discuss the proliferation of interventional strategies and refinement of clinical outcome measures, including results from the ReDUX4 trial, a phase 2b clinical trial of losmapimod in FSHD [NCT04003974].


Assuntos
Distrofia Muscular Facioescapuloumeral , Adolescente , Adulto , Proteínas de Homeodomínio/genética , Humanos , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Distrofia Muscular Facioescapuloumeral/metabolismo
13.
J Biol Chem ; 285(45): 34493-502, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20807766

RESUMO

Protein phosphatase 2A (PP2A) is one of the most abundantly expressed serine/threonine protein phosphatases. A large body of evidence suggests that PP2A is a tumor suppressor and plays critical roles in regulating apoptosis. PP2A is a heterotrimeric protein complex. Its substrate specificity, localization, and activity are regulated by regulatory subunits of PP2A. A recent study has demonstrated that single nucleotide polymorphism in B56ε (PPP2R5E), a B56 family regulatory subunit of PP2A, is associated with human soft tissue sarcoma. This raises the possibility that B56ε is involved in tumorigenesis and plays important roles in regulating apoptosis. However, this hypothesis has not been tested experimentally. Our previous studies revealed that B56ε regulates a number of developmental signaling pathways during early embryonic patterning. Here we report novel functions of B56ε in regulating apoptosis. We provide evidence that B56ε has both anti- and pro-apoptotic functions. B56ε suppresses p53-independent apoptosis during neural development, but triggers p53-dependent apoptosis. Mechanistically, B56ε regulates the p53-dependent apoptotic pathway solely through controlling the stability of p53 protein. In addition to its function in regulating apoptosis, we show that B56ε undergoes proteolytic cleavage. The cleavage of B56ε is mediated by caspase-3 and occurs on the carboxyl side of an evolutionarily conserved N-terminal "DKXD" motif. These results demonstrate that B56ε, a substrate of caspase-3, is an essential regulator of apoptosis. So far, we have identified an alternative translation isoform and a caspase cleavage product of B56ε. The significance of post-transcriptional regulation of B56ε is discussed.


Assuntos
Apoptose/fisiologia , Padronização Corporal/fisiologia , Caspase 3/metabolismo , Embrião não Mamífero/embriologia , Tubo Neural/embriologia , Proteína Fosfatase 2/metabolismo , Transdução de Sinais/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Xenopus/metabolismo , Motivos de Aminoácidos , Animais , Caspase 3/genética , Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células HeLa , Humanos , Camundongos , Células NIH 3T3 , Polimorfismo de Nucleotídeo Único , Proteína Fosfatase 2/genética , Sarcoma/genética , Sarcoma/metabolismo , Proteína Supressora de Tumor p53/genética , Proteínas de Xenopus/genética , Xenopus laevis
14.
BMC Biotechnol ; 11: 107, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-22087765

RESUMO

BACKGROUND: RNA interference (RNAi) is a conserved gene silencing mechanism mediated by small inhibitory microRNAs (miRNAs).Promoter-driven miRNA expression vectors have emerged as important tools for delivering natural or artificially designed miRNAs to eukaryotic cells and organisms. Such systems can be used to query the normal or pathogenic functions of natural miRNAs or messenger RNAs, or to therapeutically silence disease genes. RESULTS: As with any molecular cloning procedure, building miRNA-based expression constructs requires a time investment and some molecular biology skills. To improve efficiency and accelerate the construction process, we developed a method to rapidly generate miRNA expression vectors using recombinases instead of more traditional cut-and-paste molecular cloning techniques. In addition to streamlining the construction process, our cloning strategy provides vectors with added versatility. In our system, miRNAs can be constitutively expressed from the U6 promoter, or inducibly expressed by Cre recombinase. We also engineered a built-in mechanism to destroy the vector with Flp recombinase, if desired. Finally, to further simplify the construction process, we developed a software package that automates the prediction and design of optimal miRNA sequences using our system. CONCLUSIONS: We designed and tested a modular system to rapidly clone miRNA expression cassettes. Our strategy reduces the hands-on time required to successfully generate effective constructs, and can be implemented in labs with minimal molecular cloning expertise. This versatile system provides options that permit constitutive or inducible miRNA expression, depending upon the needs of the end user. As such, it has utility for basic or translational applications.


Assuntos
Clonagem Molecular/métodos , DNA Nucleotidiltransferases/metabolismo , Engenharia Genética/métodos , Vetores Genéticos/genética , MicroRNAs/genética , Software , Northern Blotting , DNA Nucleotidiltransferases/genética , Primers do DNA/genética , Células HEK293 , Humanos , Microscopia de Fluorescência , Regiões Promotoras Genéticas/genética
15.
Nat Med ; 10(8): 816-20, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15235598

RESUMO

The dominant polyglutamine expansion diseases, which include spinocerebellar ataxia type 1 (SCA1) and Huntington disease, are progressive, untreatable, neurodegenerative disorders. In inducible mouse models of SCA1 and Huntington disease, repression of mutant allele expression improves disease phenotypes. Thus, therapies designed to inhibit expression of the mutant gene would be beneficial. Here we evaluate the ability of RNA interference (RNAi) to inhibit polyglutamine-induced neurodegeneration caused by mutant ataxin-1 in a mouse model of SCA1. Upon intracerebellar injection, recombinant adeno-associated virus (AAV) vectors expressing short hairpin RNAs profoundly improved motor coordination, restored cerebellar morphology and resolved characteristic ataxin-1 inclusions in Purkinje cells of SCA1 mice. Our data demonstrate in vivo the potential use of RNAi as therapy for dominant neurodegenerative disease.


Assuntos
Expressão Gênica , Degeneração Neural/genética , Degeneração Neural/terapia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/metabolismo , Interferência de RNA/fisiologia , RNA Mensageiro/metabolismo , Ataxias Espinocerebelares/patologia , Adenoviridae , Animais , Ataxina-1 , Ataxinas , Northern Blotting , Encéfalo/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Glutamina , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/farmacologia , Proteínas Nucleares/farmacologia , Plasmídeos/genética , Desempenho Psicomotor/efeitos dos fármacos , Células de Purkinje/efeitos dos fármacos , Células de Purkinje/metabolismo , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética
16.
Nat Med ; 8(3): 253-61, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11875496

RESUMO

Attempts to develop gene therapy for Duchenne muscular dystrophy (DMD) have been complicated by the enormous size of the dystrophin gene. We have performed a detailed functional analysis of dystrophin structural domains and show that multiple regions of the protein can be deleted in various combinations to generate highly functional mini- and micro-dystrophins. Studies in transgenic mdx mice, a model for DMD, reveal that a wide variety of functional characteristics of dystrophy are prevented by some of these truncated dystrophins. Muscles expressing the smallest dystrophins are fully protected against damage caused by muscle activity and are not morphologically different from normal muscle. Moreover, injection of adeno-associated viruses carrying micro-dystrophins into dystrophic muscles of immunocompetent mdx mice results in a striking reversal of histopathological features of this disease. These results demonstrate that the dystrophic pathology can be both prevented and reversed by gene therapy using micro-dystrophins.


Assuntos
Distrofina/genética , Terapia Genética/métodos , Músculo Esquelético/fisiologia , Distrofia Muscular de Duchenne/terapia , Análise de Variância , Animais , Dependovirus/genética , Dependovirus/metabolismo , Distrofina/química , Distrofina/metabolismo , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/fisiopatologia , Conformação Proteica
18.
Proc Natl Acad Sci U S A ; 105(15): 5868-73, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18398004

RESUMO

Huntington's disease (HD) is a fatal, dominant neurodegenerative disease caused by a polyglutamine repeat expansion in exon 1 of the HD gene, which encodes the huntingtin protein. We and others have shown that RNAi is a candidate therapy for HD because expression of inhibitory RNAs targeting mutant human HD transgenes improved neuropathology and behavioral deficits in HD mouse models. Here, we developed shRNAs targeting conserved sequences in human HD and mouse HD homolog (HDh) mRNAs to initiate preclinical testing in a knockin mouse model of HD. We screened 35 shRNAs in vitro and subsequently narrowed our focus to three candidates for in vivo testing. Unexpectedly, two active shRNAs induced significant neurotoxicity in mouse striatum, although HDh mRNA expression was reduced to similar levels by all three. Additionally, a control shRNA containing mismatches also induced toxicity, although it did not reduce HDh mRNA expression. Interestingly, the toxic shRNAs generated higher antisense RNA levels, compared with the nontoxic shRNA. These results demonstrate that the robust levels of antisense RNAs emerging from shRNA expression systems can be problematic in the mouse brain. Importantly, when sequences that were toxic in the context of shRNAs were placed into artificial microRNA (miRNA) expression systems, molecular and neuropathological readouts of neurotoxicity were significantly attenuated without compromising mouse HDh silencing efficacy. Thus, miRNA-based approaches may provide more appropriate biological tools for expressing inhibitory RNAs in the brain, the implications of which are crucial to the development of RNAi for both basic biological and therapeutic applications.


Assuntos
MicroRNAs/farmacologia , Síndromes Neurotóxicas/tratamento farmacológico , Interferência de RNA , RNA Interferente Pequeno/efeitos adversos , Animais , Encéfalo/efeitos dos fármacos , Corpo Estriado , Inativação Gênica , Terapia Genética/métodos , Humanos , Doença de Huntington/terapia , Camundongos , MicroRNAs/síntese química , MicroRNAs/uso terapêutico , Síndromes Neurotóxicas/etiologia
19.
Mol Ther Nucleic Acids ; 23: 476-486, 2021 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33510937

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) arises from epigenetic changes that de-repress the DUX4 gene in muscle. The full-length DUX4 protein causes cell death and muscle toxicity, and therefore we hypothesize that FSHD therapies should center on inhibiting full-length DUX4 expression. In this study, we developed a strategy to accomplish DUX4 inhibition using U7-small nuclear RNA (snRNA) antisense expression cassettes (called U7-asDUX4). These non-coding RNAs were designed to inhibit production or maturation of the full-length DUX4 pre-mRNA by masking the DUX4 start codon, splice sites, or polyadenylation signal. In so doing, U7-asDUX4 snRNAs operate similarly to antisense oligonucleotides. However, in contrast to oligonucleotides, which are limited by poor uptake in muscle and a requirement for lifelong repeated dosing, U7-asDUX4 snRNAs can be packaged within myotropic gene therapy vectors and may require only a single administration when delivered to post-mitotic cells in vivo. We tested several U7-asDUX4s that reduced DUX4 expression in vitro and improved DUX4-associated outcomes. Inhibition of DUX4 expression via U7-snRNAs could be a new prospective gene therapy approach for FSHD or be used in combination with other strategies, like RNAi therapy, to maximize DUX4 silencing in individuals with FSHD.

20.
Front Physiol ; 12: 633058, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33732165

RESUMO

Sarcolipin (SLN) is a regulator of sarco/endo plasmic reticulum Ca2+-ATPase (SERCA) pump and has been shown to be involved in muscle nonshivering thermogenesis (NST) and energy metabolism. Interestingly, SLN expression is significantly upregulated both during muscle development and in several disease states. However, the significance of altered SLN expression in muscle patho-physiology is not completely understood. We have previously shown that transgenic over-expression of SLN in skeletal muscle is not detrimental, and can promote oxidative metabolism and exercise capacity. In contrast, some studies have suggested that SLN upregulation in disease states is deleterious for muscle function and ablation of SLN can be beneficial. In this perspective article, we critically examine both published and some new data to determine the relevance of SLN expression to disease pathology. The new data presented in this paper show that SLN levels are induced in muscle during systemic bacterial (Salmonella) infection or lipopolysaccharides (LPS) treatment. We also present data showing that SLN expression is significantly upregulated in different types of muscular dystrophies including myotubular myopathy. These data taken together reveal that upregulation of SLN expression in muscle disease is progressive and increases with severity. Therefore, we suggest that increased SLN expression should not be viewed as the cause of the disease; rather, it is a compensatory response to meet the higher energy demand of the muscle. We interpret that higher SLN/SERCA ratio positively modulate cytosolic Ca2+ signaling pathways to promote mitochondrial biogenesis and oxidative metabolism to meet higher energy demand in muscle.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA