Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res ; 76: 103375, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38490135

RESUMO

Myotonic dystrophy type 1 (DM1) is the most prevalent adult-onset muscular dystrophy affecting 1 in 8,000 individuals. It is characterized by multisystemic symptoms, primarily myopathy. The root cause of DM1 is a heterozygous CTG triplet expansion beyond the normal size threshold in the non-coding region of the DM1 protein kinase gene (DMPK). In our study, we generated and characterized three distinct DM1 induced pluripotent stem cell (iPSC) lines with CTG repeat expansions ranging from 900 to 2000 in the DMPK gene. These iPSC lines maintained normal karyotypes, exhibited distinctive colony morphology, robustly expressed pluripotency markers, differentiated into the three primary germ layers, and lacked residual viral vectors.


Assuntos
Células-Tronco Pluripotentes Induzidas , Distrofia Miotônica , Adulto , Humanos , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Expansão das Repetições de Trinucleotídeos , Experimentação Humana Terapêutica , Linhagem Celular , Miotonina Proteína Quinase/genética
2.
Stem Cell Res ; 75: 103308, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38232626

RESUMO

Dilated cardiomyopathy (DCM) is a prevalent cause of heart failure. We generated induced pluripotent stem cell (iPSC) lines from a DCM patient carrying a mutation in the SCN5A gene, with his healthy father serving as a control. Notably, we employed CRISPR-Cas9 to rectify the mutation in the patient's iPSC line. The resulting iPSC lines expressed pluripotency markers, underwent differentiation into all three embryonic germ layers, maintained a normal karyotype, and lacked reprogramming viral vectors. These iPSC lines serve as a model for delving into the mechanisms of DCM and hold promise for the development of personalized therapeutic approaches.


Assuntos
Cardiomiopatia Dilatada , Células-Tronco Pluripotentes Induzidas , Humanos , Masculino , Células-Tronco Pluripotentes Induzidas/metabolismo , Cardiomiopatia Dilatada/genética , Linhagem Celular , Mutação , Arritmias Cardíacas/metabolismo , Pai
3.
Stem Cell Res ; 72: 103234, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37871474

RESUMO

Congenital myotonic dystrophy (CDM) is an autosomal dominant multisystemic disorder attributed to a large expansion of CTG trinucleotide repeats within the myotonic dystrophy protein kinase (DMPK) gene. In this study, we successfully reprogrammed dermal fibroblasts derived from two pediatric CDM patients and two age-matched individuals into induced pluripotent stem cells (iPSCs) using a non-integrating viral vector. The resulting CDM iPSC lines harbored approximately 2000 CTG repeats in the mutated DMPK allele. These iPSC lines expressed pluripotency markers and exhibited the capacity to differentiate into cells representing all three germinal layers, confirming their reliability as a research tool for investigating CDM and therapeutic strategies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Distrofia Miotônica , Humanos , Criança , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Expansão das Repetições de Trinucleotídeos , Reprodutibilidade dos Testes , Miotonina Proteína Quinase/genética
4.
Stem Cell Res ; 71: 103168, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37473461

RESUMO

The generation of control human iPSC lines is important in fundamental research to understand the physiological and physiopathological mechanisms underlying human diseases. We generated and characterized two control hiPSC lines from lymphoblastoid cells collected from apparently healthy individuals. These hiPSCs display pluripotency markers, can differentiate into three embryonic germ layers, possess normal karyotypes and colony morphologies, and have no reprogramming viral vectors.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Linhagem Celular , Reprogramação Celular , Diferenciação Celular/fisiologia
5.
Stem Cell Res ; 71: 103148, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37352653

RESUMO

Myotonic dystrophy type 1 (DM1) is a genetic neuromuscular disorder that affects many organs, including the heart. DM1 is caused by a heterozygous CTG triplet expansion exceeding the normal size threshold in the non-coding region of the DM1 protein kinase gene (DMPK). We generated and characterized a DM1 iPSC line carrying a 700 CTG repeat expansion as well as a control iPSC line from a healthy individual. The two iPSC lines expressed several pluripotency markers, had the capacity to differentiate into the three primary germ layers, had no residual viral vectors, had normal karyotypes, and had a typical colony morphology.


Assuntos
Células-Tronco Pluripotentes Induzidas , Distrofia Miotônica , Humanos , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Linhagem Celular , Miotonina Proteína Quinase/genética
6.
Stem Cell Res ; 67: 103037, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36739767

RESUMO

Myotonic dystrophy Type 1 (DM1) is a severe inherited neuromuscular disease and is the most prevalent form of muscular dystrophy in adults. DM1 involves not only the striated muscles including skeletal, and cardiac but also other organs such as the eye, brain and gonads. We have generated and characterized 4 adult heterozygous DM1 iPSC lines carrying between 1300 and 1600 CTG repeat expansion in the DM1 protein kinase gene, and a control from an apparently healthy individual. They all show strong pluripotency markers, differentiation capacity, the absence of residual viral vectors as well as normal karyotypes and colony morphologies.


Assuntos
Células-Tronco Pluripotentes Induzidas , Distrofia Miotônica , Adulto , Humanos , Distrofia Miotônica/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Expansão das Repetições de Trinucleotídeos , Linhagem Celular , Músculo Esquelético/metabolismo , Miotonina Proteína Quinase/genética
7.
Int J Mol Sci ; 23(21)2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36362145

RESUMO

Myotonic dystrophy type 1 (DM1) is a dominant genetic disease in which the expansion of long CTG trinucleotides in the 3' UTR of the myotonic dystrophy protein kinase (DMPK) gene results in toxic RNA gain-of-function and gene mis-splicing affecting mainly the muscles, the heart, and the brain. The CUG-expanded transcripts are a suitable target for the development of antisense oligonucleotide (ASO) therapies. Various chemical modifications of the sugar-phosphate backbone have been reported to significantly enhance the affinity of ASOs for RNA and their resistance to nucleases, making it possible to reverse DM1-like symptoms following systemic administration in different transgenic mouse models. However, specific tissue delivery remains to be improved to achieve significant clinical outcomes in humans. Several strategies, including ASO conjugation to cell-penetrating peptides, fatty acids, or monoclonal antibodies, have recently been shown to improve potency in muscle and cardiac tissues in mice. Moreover, intrathecal administration of ASOs may be an advantageous complementary administration route to bypass the blood-brain barrier and correct defects of the central nervous system in DM1. This review describes the evolution of the chemical design of antisense oligonucleotides targeting CUG-expanded mRNAs and how recent advances in the field may be game-changing by forwarding laboratory findings into clinical research and treatments for DM1 and other microsatellite diseases.


Assuntos
Distrofia Miotônica , Camundongos , Humanos , Animais , Distrofia Miotônica/tratamento farmacológico , Distrofia Miotônica/genética , Miotonina Proteína Quinase/genética , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/uso terapêutico , Camundongos Transgênicos , Oligonucleotídeos/uso terapêutico , Regiões 3' não Traduzidas , Expansão das Repetições de Trinucleotídeos
8.
Hum Gene Ther ; 33(15-16): 810-820, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35794764

RESUMO

Myotonic dystrophy type 1 (DM1) is a neuromuscular disorder that affects many organs. It is caused by the expansion of a cytosine-thymine-guanine triplet repeat in the 3' untranslated region of the human dystrophia myotonica protein kinase (hDMPK) gene, which results in a toxic gain of function of mutant hDMPK RNA transcripts. Antisense oligonucleotides (ASOs) have emerged in recent years as a potential gene therapy to treat DM1. However, the clinical efficacy of the systemic administration of ASOs is limited by a combination of insufficient potency and poor tissue distribution. In the present study, we assessed the potential of a new ligand-conjugated ASO (IONIS-877864; C16-HA-ASO) to target mutant hDMPK mRNA transcripts in the DMSXL mouse model of DM1 carrying over 1000 CTG pathogenic repeats. DMSXL mice were treated subcutaneously for 9 weeks with either IONIS-877864 (12.5 or 25 mg/kg) or IONIS-486178 (12.5 or 25 mg/kg), an unconjugated ASO with the same sequence. At 25 mg/kg, IONIS-877864 significantly enhanced ASO delivery into the striated muscles of DMSXL mice following systemic administration compared with the unconjugated control. IONIS-877864 was also more efficacious than IONIS-486178, reducing mutant hDMPK transcripts by up to 92% in the skeletal muscles and 78% in the hearts of DMSXL mice. The decrease in mutant hDMPK transcripts in skeletal muscles caused by IONIS-877864 was associated with a significant improvement in muscle strength. IONIS-877864 was nontoxic in the DMSXL mouse model. The present study showed that the C16-HA-conjugated ASO is a powerful tool for the development of gene therapy for DM1.


Assuntos
Distrofia Miotônica , Animais , Modelos Animais de Doenças , Humanos , Ligantes , Camundongos , Músculo Esquelético/metabolismo , Distrofia Miotônica/genética , Distrofia Miotônica/patologia , Distrofia Miotônica/terapia , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/uso terapêutico , RNA/genética , Expansão das Repetições de Trinucleotídeos/genética
9.
Gene Ther ; 29(12): 698-709, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35075265

RESUMO

Myotonic dystrophy, or dystrophia myotonica type 1 (DM1), is a multi-systemic disorder and is the most common adult form of muscular dystrophy. It affects not only muscles but also many organs, including the brain. Cerebral impairments include cognitive deficits, daytime sleepiness, and loss of visuospatial and memory functions. The expression of mutated transcripts with CUG repeats results in a gain of toxic mRNA function. The antisense oligonucleotide (ASO) strategy to treat DM1 brain deficits is limited by the fact that ASOs do not cross the blood-brain barrier after systemic administration, indicating that other methods of delivery should be considered. ASO technology has emerged as a powerful tool for developing potential new therapies for a wide variety of human diseases, and its potential has been proven in a recent clinical trial. Targeting DMPK mRNA in neural cells derived from human induced pluripotent stem cells obtained from a DM1 patient with the IONIS 486178 ASO abolished CUG-expanded foci, enabled nuclear redistribution of MBNL1/2, and corrected aberrant splicing. Intracerebroventricular injection of the IONIS 486178 ASO in DMSXL mice decreased the levels of mutant DMPK mRNAs by up to 70% throughout different brain regions. It also reversed behavioral abnormalities following neonatal administration. The present study indicated that the IONIS 486178 ASO targets mutant DMPK mRNAs in the brain and strongly supports the feasibility of a therapy for DM1 patients based on the intrathecal injection of an ASO.


Assuntos
Células-Tronco Pluripotentes Induzidas , Distrofia Miotônica , Adulto , Humanos , Animais , Camundongos , Distrofia Miotônica/terapia , Distrofia Miotônica/tratamento farmacológico , Miotonina Proteína Quinase/genética , Miotonina Proteína Quinase/metabolismo , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/uso terapêutico , Expansão das Repetições de Trinucleotídeos , Proteínas de Ligação a RNA/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Oligonucleotídeos/uso terapêutico , Encéfalo/metabolismo
10.
J Clin Invest ; 129(11): 4739-4744, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31479430

RESUMO

Antisense oligonucleotides (ASOs) targeting pathologic RNAs have shown promising therapeutic corrections for many genetic diseases including myotonic dystrophy (DM1). Thus, ASO strategies for DM1 can abolish the toxic RNA gain-of-function mechanism caused by nucleus-retained mutant DMPK (DM1 protein kinase) transcripts containing CUG expansions (CUGexps). However, systemic use of ASOs for this muscular disease remains challenging due to poor drug distribution to skeletal muscle. To overcome this limitation, we test an arginine-rich Pip6a cell-penetrating peptide and show that Pip6a-conjugated morpholino phosphorodiamidate oligomer (PMO) dramatically enhanced ASO delivery into striated muscles of DM1 mice following systemic administration in comparison with unconjugated PMO and other ASO strategies. Thus, low-dose treatment with Pip6a-PMO-CAG targeting pathologic expansions is sufficient to reverse both splicing defects and myotonia in DM1 mice and normalizes the overall disease transcriptome. Moreover, treated DM1 patient-derived muscle cells showed that Pip6a-PMO-CAG specifically targets mutant CUGexp-DMPK transcripts to abrogate the detrimental sequestration of MBNL1 splicing factor by nuclear RNA foci and consequently MBNL1 functional loss, responsible for splicing defects and muscle dysfunction. Our results demonstrate that Pip6a-PMO-CAG induces long-lasting correction with high efficacy of DM1-associated phenotypes at both molecular and functional levels, and strongly support the use of advanced peptide conjugates for systemic corrective therapy in DM1.


Assuntos
Peptídeos Penetradores de Células/farmacologia , Músculo Esquelético/metabolismo , Distrofia Miotônica , Miotonina Proteína Quinase , Oligodesoxirribonucleotídeos Antissenso , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Humanos , Camundongos , Músculo Esquelético/patologia , Distrofia Miotônica/tratamento farmacológico , Distrofia Miotônica/genética , Distrofia Miotônica/metabolismo , Distrofia Miotônica/patologia , Miotonina Proteína Quinase/genética , Miotonina Proteína Quinase/metabolismo , Oligodesoxirribonucleotídeos Antissenso/genética , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
11.
Mol Ther Nucleic Acids ; 7: 465-474, 2017 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-28624222

RESUMO

Myotonic dystrophy type 1 (DM1), a dominant hereditary muscular dystrophy, is caused by an abnormal expansion of a (CTG)n trinucleotide repeat in the 3' UTR of the human dystrophia myotonica protein kinase (DMPK) gene. As a consequence, mutant transcripts containing expanded CUG repeats are retained in nuclear foci and alter the function of splicing regulatory factors members of the MBNL and CELF families, resulting in alternative splicing misregulation of specific transcripts in affected DM1 tissues. In the present study, we treated DMSXL mice systemically with a 2'-4'-constrained, ethyl-modified (ISIS 486178) antisense oligonucleotide (ASO) targeted to the 3' UTR of the DMPK gene, which led to a 70% reduction in CUGexp RNA abundance and foci in different skeletal muscles and a 30% reduction in the heart. Furthermore, treatment with ISIS 486178 ASO improved body weight, muscle strength, and muscle histology, whereas no overt toxicity was detected. This is evidence that the reduction of CUGexp RNA improves muscle strength in DM1, suggesting that muscle weakness in DM1 patients may be improved following elimination of toxic RNAs.

12.
J Pharmacol Exp Ther ; 355(2): 329-40, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26330536

RESUMO

Myotonic dystrophy type 1 (DM1) is the most common form of muscular dystrophy in adults. DM1 is caused by an expanded CTG repeat in the 3'-untranslated region of DMPK, the gene encoding dystrophia myotonica protein kinase (DMPK). Antisense oligonucleotides (ASOs) containing 2',4'-constrained ethyl-modified (cEt) residues exhibit a significantly increased RNA binding affinity and in vivo potency relative to those modified with other 2'-chemistries, which we speculated could translate to enhanced activity in extrahepatic tissues, such as muscle. Here, we describe the design and characterization of a cEt gapmer DMPK ASO (ISIS 486178), with potent activity in vitro and in vivo against mouse, monkey, and human DMPK. Systemic delivery of unformulated ISIS 486718 to wild-type mice decreased DMPK mRNA levels by up to 90% in liver and skeletal muscle. Similarly, treatment of either human DMPK transgenic mice or cynomolgus monkeys with ISIS 486178 led to up to 70% inhibition of DMPK in multiple skeletal muscles and ∼50% in cardiac muscle in both species. Importantly, inhibition of DMPK was well tolerated and was not associated with any skeletal muscle or cardiac toxicity. Also interesting was the demonstration that the inhibition of DMPK mRNA levels in muscle was maintained for up to 16 and 13 weeks post-treatment in mice and monkeys, respectively. These results demonstrate that cEt-modified ASOs show potent activity in skeletal muscle, and that this attractive therapeutic approach warrants further clinical investigation to inhibit the gain-of-function toxic RNA underlying the pathogenesis of DM1.


Assuntos
Distrofia Miotônica/tratamento farmacológico , Miotonina Proteína Quinase/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Oligonucleotídeos/farmacologia , Animais , Linhagem Celular , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Transgênicos , Músculo Esquelético/enzimologia , Miotonina Proteína Quinase/antagonistas & inibidores , Miotonina Proteína Quinase/genética , Oligonucleotídeos/química , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...