Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
EMBO Mol Med ; 15(11): e17683, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37724723

ABSTRACT

Spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality. The advent of approved treatments for this devastating condition has significantly changed SMA patients' life expectancy and quality of life. Nevertheless, these are not without limitations, and research efforts are underway to develop new approaches for improved and long-lasting benefits for patients. Protein arginine methyltransferases (PRMTs) are emerging as druggable epigenetic targets, with several small-molecule PRMT inhibitors already in clinical trials. From a screen of epigenetic molecules, we have identified MS023, a potent and selective type I PRMT inhibitor able to promote SMN2 exon 7 inclusion in preclinical SMA models. Treatment of SMA mice with MS023 results in amelioration of the disease phenotype, with strong synergistic amplification of the positive effect when delivered in combination with the antisense oligonucleotide nusinersen. Moreover, transcriptomic analysis revealed that MS023 treatment has minimal off-target effects, and the added benefit is mainly due to targeting neuroinflammation. Our study warrants further clinical investigation of PRMT inhibition both as a stand-alone and add-on therapy for SMA.


Subject(s)
Muscular Atrophy, Spinal , Quality of Life , Animals , Humans , Infant , Mice , Exons , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Oligonucleotides/pharmacology , Oligonucleotides/therapeutic use , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/therapeutic use
2.
JCI Insight ; 7(24)2022 12 22.
Article in English | MEDLINE | ID: mdl-36346674

ABSTRACT

Antisense oligonucleotides (ASOs) have emerged as one of the most innovative new genetic drug modalities. However, their high molecular weight limits their bioavailability for otherwise-treatable neurological disorders. We investigated conjugation of ASOs to an antibody against the murine transferrin receptor, 8D3130, and evaluated it via systemic administration in mouse models of the neurodegenerative disease spinal muscular atrophy (SMA). SMA, like several other neurological and neuromuscular diseases, is treatable with single-stranded ASOs that modulate splicing of the survival motor neuron 2 (SMN2) gene. Administration of 8D3130-ASO conjugate resulted in elevated levels of bioavailability to the brain. Additionally, 8D3130-ASO yielded therapeutic levels of SMN2 splicing in the central nervous system of adult human SMN2-transgenic (hSMN2-transgenic) mice, which resulted in extended survival of a severely affected SMA mouse model. Systemic delivery of nucleic acid therapies with brain-targeting antibodies offers powerful translational potential for future treatments of neuromuscular and neurodegenerative diseases.


Subject(s)
Muscular Atrophy, Spinal , Neurodegenerative Diseases , Mice , Animals , Humans , Oligonucleotides/pharmacology , Oligonucleotides/therapeutic use , Neurodegenerative Diseases/drug therapy , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Central Nervous System , Oligonucleotides, Antisense/therapeutic use , Mice, Transgenic , Disease Models, Animal
3.
Methods Mol Biol ; 2383: 491-513, 2022.
Article in English | MEDLINE | ID: mdl-34766309

ABSTRACT

Oligonucleotides (ONs) are therapeutic macromolecules with great potential for the treatment of neurological conditions, including spinal muscular atrophy (SMA), a neurodegenerative disease. However, the neurovascular unit severely limits their distribution to the neural parenchyma of the brain and the spinal cord. Cell-penetrating peptides (CPPs) can be conjugated to oligonucleotides to increase their delivery across biological barriers. In this chapter, we describe the synthesis and conjugation of CPPs to oligonucleotides, and the use of a severe SMA mouse model to test in vivo the efficacy of CPP-delivered oligonucleotides, using ELISA, western blot, and TaqMan™ RT-qPCR assays.


Subject(s)
Muscular Atrophy, Spinal , Animals , Cell-Penetrating Peptides , Disease Models, Animal , Mice , Muscular Atrophy, Spinal/drug therapy , Oligonucleotides , Oligonucleotides, Antisense
4.
JCI Insight ; 6(13)2021 07 08.
Article in English | MEDLINE | ID: mdl-34236053

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by loss of survival motor neuron (SMN) protein. While SMN restoration therapies are beneficial, they are not a cure. We aimed to identify potentially novel treatments to alleviate muscle pathology combining transcriptomics, proteomics, and perturbational data sets. This revealed potential drug candidates for repurposing in SMA. One of the candidates, harmine, was further investigated in cell and animal models, improving multiple disease phenotypes, including lifespan, weight, and key molecular networks in skeletal muscle. Our work highlights the potential of multiple and parallel data-driven approaches for the development of potentially novel treatments for use in combination with SMN restoration therapies.


Subject(s)
Harmine/pharmacology , Muscle, Skeletal , Muscular Atrophy, Spinal , Survival of Motor Neuron 1 Protein/metabolism , Animals , Cells, Cultured , Computational Biology , Disease Models, Animal , Drug Repositioning/methods , Gene Expression Profiling/methods , Humans , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Neuromuscular Agents/pharmacology , Proteomics/methods
5.
Nucleic Acid Ther ; 31(3): 190-200, 2021 06.
Article in English | MEDLINE | ID: mdl-33989066

ABSTRACT

A series of 2'-deoxy and novel 2'-O-methyl and 2'-O-(2-methoxyethyl) (2'-MOE) oligonucleotides with internucleotide methanesulfonyl (mesyl, µ) or 1-butanesulfonyl (busyl, ß) phosphoramidate groups has been synthesized for evaluation as potential splice-switching oligonucleotides. Evaluation of their splice-switching activity in spinal muscular atrophy patient-derived fibroblasts revealed no significant difference in splice-switching efficacy between 2'-MOE mesyl oligonucleotide and the corresponding phosphorothioate (nusinersen). Yet, a survival study with model neonatal mice has shown the antisense 2'-MOE mesyl oligonucleotide to be inferior to nusinersen at the highest dose of 40 mg/kg. A reason for their lower activity in vivo as ascertained by cellular uptake study by fluorescent confocal microscopy in HEK293 cell line could possibly be ascribed to compromised endosomal release and/or nuclear uptake of the 2'-OMe or 2'-MOE µ- and ß-oligonucleotides compared to their phosphorothioate analog.


Subject(s)
Muscular Atrophy, Spinal , Oligonucleotides , Amides , Animals , HEK293 Cells , Humans , Mice , Oligonucleotides/genetics , Oligonucleotides, Antisense/genetics , Phosphoric Acids
6.
EMBO Mol Med ; 13(4): e13243, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33821570

ABSTRACT

Nucleic acid-based therapeutics that regulate gene expression have been developed towards clinical use at a steady pace for several decades, but in recent years the field has been accelerating. To date, there are 11 marketed products based on antisense oligonucleotides, aptamers and small interfering RNAs, and many others are in the pipeline for both academia and industry. A major technology trigger for this development has been progress in oligonucleotide chemistry to improve the drug properties and reduce cost of goods, but the main hurdle for the application to a wider range of disorders is delivery to target tissues. The adoption of delivery technologies, such as conjugates or nanoparticles, has been a game changer for many therapeutic indications, but many others are still awaiting their eureka moment. Here, we cover the variety of methods developed to deliver nucleic acid-based therapeutics across biological barriers and the model systems used to test them. We discuss important safety considerations and regulatory requirements for synthetic oligonucleotide chemistries and the hurdles for translating laboratory breakthroughs to the clinic. Recent advances in the delivery of nucleic acid-based therapeutics and in the development of model systems, as well as safety considerations and regulatory requirements for synthetic oligonucleotide chemistries are discussed in this review on oligonucleotide-based therapeutics.


Subject(s)
Nanoparticles , Oligonucleotides , Gene Expression , Oligonucleotides, Antisense , RNA, Small Interfering
7.
Mol Ther Nucleic Acids ; 23: 731-742, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33575118

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by mutations in the survival motor neuron 1 (SMN1) gene. All patients have at least one copy of a paralog, SMN2, but a C-to-T transition in this gene results in exon 7 skipping in a majority of transcripts. Approved treatment for SMA involves promoting exon 7 inclusion in the SMN2 transcript or increasing the amount of full-length SMN by gene replacement with a viral vector. Increasing the pool of SMN2 transcripts and increasing their translational efficiency can be used to enhance splice correction. We sought to determine whether the 5' untranslated region (5' UTR) of SMN2 contains a repressive feature that can be targeted to increase SMN levels. We found that antisense oligonucleotides (ASOs) complementary to the 5' end of SMN2 increase SMN mRNA and protein levels and that this effect is due to inhibition of SMN2 mRNA decay. Moreover, use of the 5' UTR ASO in combination with a splice-switching oligonucleotide (SSO) increases SMN levels above those attained with the SSO alone. Our results add to the current understanding of SMN regulation and point toward a new therapeutic target for SMA.

8.
Methods Mol Biol ; 2036: 221-236, 2019.
Article in English | MEDLINE | ID: mdl-31410800

ABSTRACT

Antisense oligonucleotides (ASOs) are a widely used form of gene therapy, which is translatable to multiple disorders. A major obstacle for ASO efficacy is its bioavailability for in vivo and in vitro studies. To overcome this challenge we use cell-penetrating peptides (CPPs) for systemic delivery of ASOs. One of the most advanced clinical uses of ASOs is for the treatment of spinal muscular atrophy (SMA). In this chapter, we describe the techniques used for in vitro screening and analysing in vivo biodistribution of CPP-conjugated ASOs targeting the survival motor neuron 2, SMN2, the dose-dependent modifying gene for SMA.


Subject(s)
Cell-Penetrating Peptides/chemistry , Gene Transfer Techniques , Muscular Atrophy, Spinal/genetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Administration, Intravenous , Cell Line , Drug Delivery Systems , Fibroblasts/metabolism , Humans , Immunohistochemistry , Motor Neurons/metabolism , Muscular Atrophy, Spinal/therapy , Oligonucleotides, Antisense/chemistry , RNA Splicing
9.
Gene Ther ; 27(10-11): 505-515, 2019 11.
Article in English | MEDLINE | ID: mdl-32313099

ABSTRACT

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by loss of the survival motor neuron (SMN) gene. While there are currently two approved gene-based therapies for SMA, availability, high cost, and differences in patient response indicate that alternative treatment options are needed. Optimal therapeutic strategies will likely be a combination of SMN-dependent and -independent treatments aimed at alleviating symptoms in the central nervous system and peripheral muscles. Krüppel-like factor 15 (KLF15) is a transcription factor that regulates key metabolic and ergogenic pathways in muscle. We have recently reported significant downregulation of Klf15 in muscle of presymptomatic SMA mice. Importantly, perinatal upregulation of Klf15 via transgenic and pharmacological methods resulted in improved disease phenotypes in SMA mice, including weight and survival. In the current study, we designed an adeno-associated virus serotype 8 (AAV8) vector to overexpress a codon-optimized Klf15 cDNA under the muscle-specific Spc5-12 promoter (AAV8-Klf15). Administration of AAV8-Klf15 to severe Taiwanese Smn-/-;SMN2 or intermediate Smn2B/- SMA mice significantly increased Klf15 expression in muscle. We also observed significant activity of the AAV8-Klf15 vector in liver and heart. AAV8-mediated Klf15 overexpression moderately improved survival in the Smn2B/- model but not in the Taiwanese mice. An inability to specifically induce Klf15 expression at physiological levels in a time- and tissue-dependent manner may have contributed to this limited efficacy. Thus, our work demonstrates that an AAV8-Spc5-12 vector induces high gene expression as early as P2 in several tissues including muscle, heart, and liver, but highlights the challenges of achieving meaningful vector-mediated transgene expression of Klf15.


Subject(s)
Dependovirus , Muscular Atrophy, Spinal , Animals , Dependovirus/genetics , Disease Models, Animal , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Transgenic , Muscles , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Serogroup , Survival of Motor Neuron 1 Protein/genetics
10.
Hum Mol Genet ; 28(3): 396-406, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30281092

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by loss of dystrophin protein, leading to progressive muscle weakness and premature death due to respiratory and/or cardiac complications. Cardiac involvement is characterized by progressive dilated cardiomyopathy, decreased fractional shortening and metabolic dysfunction involving reduced metabolism of fatty acids-the major cardiac metabolic substrate. Several mouse models have been developed to study molecular and pathological consequences of dystrophin deficiency, but do not recapitulate all aspects of human disease pathology and exhibit a mild cardiac phenotype. Here we demonstrate that Cmah (cytidine monophosphate-sialic acid hydroxylase)-deficient mdx mice (Cmah-/-;mdx) have an accelerated cardiac phenotype compared to the established mdx model. Cmah-/-;mdx mice display earlier functional deterioration, specifically a reduction in right ventricle (RV) ejection fraction and stroke volume (SV) at 12 weeks of age and decreased left ventricle diastolic volume with subsequent reduced SV compared to mdx mice by 24 weeks. They further show earlier elevation of cardiac damage markers for fibrosis (Ctgf), oxidative damage (Nox4) and haemodynamic load (Nppa). Cardiac metabolic substrate requirement was assessed using hyperpolarized magnetic resonance spectroscopy indicating increased in vivo glycolytic flux in Cmah-/-;mdx mice. Early upregulation of mitochondrial genes (Ucp3 and Cpt1) and downregulation of key glycolytic genes (Pdk1, Pdk4, Ppara), also denote disturbed cardiac metabolism and shift towards glucose utilization in Cmah-/-;mdx mice. Moreover, we show long-term treatment with peptide-conjugated exon skipping antisense oligonucleotides (20-week regimen), resulted in 20% cardiac dystrophin protein restoration and significantly improved RV cardiac function. Therefore, Cmah-/-;mdx mice represent an appropriate model for evaluating cardiac benefit of novel DMD therapeutics.


Subject(s)
Cytidine Monophosphate/genetics , Dystrophin/deficiency , Morpholinos/therapeutic use , Animals , Cardiomyopathy, Dilated/genetics , Carnitine O-Palmitoyltransferase/genetics , Connective Tissue Growth Factor/analysis , Cytidine Monophosphate/physiology , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Exons , Genetic Therapy/methods , Heart/physiopathology , Male , Mice , Mice, Inbred mdx , Mixed Function Oxygenases/metabolism , Muscular Dystrophy, Duchenne/genetics , Myocardium/metabolism , NADPH Oxidase 4/analysis , Oligonucleotides, Antisense/genetics , Peptides/genetics , Phenotype , Stroke Volume , Uncoupling Protein 3/genetics , Ventricular Function, Right
11.
EBioMedicine ; 31: 226-242, 2018 May.
Article in English | MEDLINE | ID: mdl-29735415

ABSTRACT

The circadian glucocorticoid-Krüppel-like factor 15-branched-chain amino acid (GC-KLF15-BCAA) signaling pathway is a key regulatory axis in muscle, whose imbalance has wide-reaching effects on metabolic homeostasis. Spinal muscular atrophy (SMA) is a neuromuscular disorder also characterized by intrinsic muscle pathologies, metabolic abnormalities and disrupted sleep patterns, which can influence or be influenced by circadian regulatory networks that control behavioral and metabolic rhythms. We therefore set out to investigate the contribution of the GC-KLF15-BCAA pathway in SMA pathophysiology of Taiwanese Smn-/-;SMN2 and Smn2B/- mouse models. We thus uncover substantial dysregulation of GC-KLF15-BCAA diurnal rhythmicity in serum, skeletal muscle and metabolic tissues of SMA mice. Importantly, modulating the components of the GC-KLF15-BCAA pathway via pharmacological (prednisolone), genetic (muscle-specific Klf15 overexpression) and dietary (BCAA supplementation) interventions significantly improves disease phenotypes in SMA mice. Our study highlights the GC-KLF15-BCAA pathway as a contributor to SMA pathogenesis and provides several treatment avenues to alleviate peripheral manifestations of the disease. The therapeutic potential of targeting metabolic perturbations by diet and commercially available drugs could have a broader implementation across other neuromuscular and metabolic disorders characterized by altered GC-KLF15-BCAA signaling.


Subject(s)
Amino Acids, Branched-Chain/pharmacology , DNA-Binding Proteins , Dietary Supplements , Muscular Atrophy, Spinal , Prednisolone/pharmacology , Signal Transduction/drug effects , Transcription Factors , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Kruppel-Like Transcription Factors , Mice , Mice, Knockout , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Nucleic Acid Ther ; 27(3): 130-143, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28118087

ABSTRACT

Splice-switching antisense oligonucleotides are emerging treatments for neuromuscular diseases, with several splice-switching oligonucleotides (SSOs) currently undergoing clinical trials such as for Duchenne muscular dystrophy (DMD) and spinal muscular atrophy (SMA). However, the development of systemically delivered antisense therapeutics has been hampered by poor tissue penetration and cellular uptake, including crossing of the blood-brain barrier (BBB) to reach targets in the central nervous system (CNS). For SMA application, we have investigated the ability of various BBB-crossing peptides for CNS delivery of a splice-switching phosphorodiamidate morpholino oligonucleotide (PMO) targeting survival motor neuron 2 (SMN2) exon 7 inclusion. We identified a branched derivative of the well-known ApoE (141-150) peptide, which as a PMO conjugate was capable of exon inclusion in the CNS following systemic administration, leading to an increase in the level of full-length SMN2 transcript. Treatment of newborn SMA mice with this peptide-PMO (P-PMO) conjugate resulted in a significant increase in the average lifespan and gains in weight, muscle strength, and righting reflexes. Systemic treatment of adult SMA mice with this newly identified P-PMO also resulted in small but significant increases in the levels of SMN2 pre-messenger RNA (mRNA) exon inclusion in the CNS and peripheral tissues. This work provides proof of principle for the ability to select new peptide paradigms to enhance CNS delivery and activity of a PMO SSO through use of a peptide-based delivery platform for the treatment of SMA potentially extending to other neuromuscular and neurodegenerative diseases.


Subject(s)
Apolipoproteins E/pharmacokinetics , Morpholinos/pharmacology , Morpholinos/pharmacokinetics , Muscular Atrophy, Spinal/drug therapy , Peptides/pharmacokinetics , Animals , Animals, Newborn , Apolipoproteins E/chemical synthesis , Apolipoproteins E/chemistry , Biomarkers/blood , Blood-Brain Barrier/chemistry , Blood-Brain Barrier/metabolism , Brain/cytology , Cell Survival/drug effects , Disease Models, Animal , Exons , Fibroblasts/drug effects , Hepatocytes/drug effects , Humans , Kidney/chemistry , Mice , Morpholinos/chemistry , Morpholinos/therapeutic use , Nanoconjugates/analysis , Nanoconjugates/chemistry , Nanoconjugates/therapeutic use , Peptides/chemical synthesis , Peptides/chemistry , Phenotype , Quadriceps Muscle/chemistry , Survival of Motor Neuron 2 Protein/drug effects
13.
Proc Natl Acad Sci U S A ; 113(39): 10962-7, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27621445

ABSTRACT

The development of antisense oligonucleotide therapy is an important advance in the identification of corrective therapy for neuromuscular diseases, such as spinal muscular atrophy (SMA). Because of difficulties of delivering single-stranded oligonucleotides to the CNS, current approaches have been restricted to using invasive intrathecal single-stranded oligonucleotide delivery. Here, we report an advanced peptide-oligonucleotide, Pip6a-morpholino phosphorodiamidate oligomer (PMO), which demonstrates potent efficacy in both the CNS and peripheral tissues in severe SMA mice following systemic administration. SMA results from reduced levels of the ubiquitously expressed survival motor neuron (SMN) protein because of loss-of-function mutations in the SMN1 gene. Therapeutic splice-switching oligonucleotides (SSOs) modulate exon 7 splicing of the nearly identical SMN2 gene to generate functional SMN protein. Pip6a-PMO yields SMN expression at high efficiency in peripheral and CNS tissues, resulting in profound phenotypic correction at doses an order-of-magnitude lower than required by standard naked SSOs. Survival is dramatically extended from 12 d to a mean of 456 d, with improvement in neuromuscular junction morphology, down-regulation of transcripts related to programmed cell death in the spinal cord, and normalization of circulating insulin-like growth factor 1. The potent systemic efficacy of Pip6a-PMO, targeting both peripheral as well as CNS tissues, demonstrates the high clinical potential of peptide-PMO therapy for SMA.


Subject(s)
Muscular Atrophy, Spinal/drug therapy , Oligonucleotides/therapeutic use , Peptides/chemistry , Aging , Alleles , Amino Acid Sequence , Biomarkers/blood , Cell Line , Humans , Movement , Muscular Atrophy, Spinal/blood , Muscular Atrophy, Spinal/pathology , Neuromuscular Junction/drug effects , Neuromuscular Junction/metabolism , Oligonucleotides/administration & dosage , Oligonucleotides/pharmacology , Phenotype , RNA Splicing/genetics , Survival Analysis , Survival of Motor Neuron 2 Protein/genetics
14.
Eur J Hum Genet ; 24(2): 271-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25990798

ABSTRACT

Allele-specific gene therapy aims to silence expression of mutant alleles through targeting of disease-linked single-nucleotide polymorphisms (SNPs). However, SNP linkage to disease varies between populations, making such molecular therapies applicable only to a subset of patients. Moreover, not all SNPs have the molecular features necessary for potent gene silencing. Here we provide knowledge to allow the maximisation of patient coverage by building a comprehensive understanding of SNPs ranked according to their predicted suitability toward allele-specific silencing in 14 repeat expansion diseases: amyotrophic lateral sclerosis and frontotemporal dementia, dentatorubral-pallidoluysian atrophy, myotonic dystrophy 1, myotonic dystrophy 2, Huntington's disease and several spinocerebellar ataxias. Our systematic analysis of DNA sequence variation shows that most annotated SNPs are not suitable for potent allele-specific silencing across populations because of suboptimal sequence features and low variability (>97% in HD). We suggest maximising patient coverage by selecting SNPs with high heterozygosity across populations, and preferentially targeting SNPs that lead to purine:purine mismatches in wild-type alleles to obtain potent allele-specific silencing. We therefore provide fundamental knowledge on strategies for optimising patient coverage of therapeutics for microsatellite expansion disorders by linking analysis of population genetic variation to the selection of molecular targets.


Subject(s)
DNA Repeat Expansion/genetics , Genetic Diseases, Inborn/genetics , Genetic Therapy , Molecular Targeted Therapy , Alleles , Gene Silencing , Genetic Diseases, Inborn/therapy , Genetics, Population , Heterozygote , Humans , Polymorphism, Single Nucleotide/genetics
15.
Sci Rep ; 5: 11632, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26113184

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by absence of the integral structural protein, dystrophin, which renders muscle fibres susceptible to injury and degeneration. This ultimately results in cardiorespiratory dysfunction, which is the predominant cause of death in DMD patients, and highlights the importance of therapeutic targeting of the cardiorespiratory system. While there is some evidence to suggest that restoring dystrophin in the diaphragm improves both respiratory and cardiac function, the role of the diaphragm is not well understood. Here using exon skipping oligonucleotides we predominantly restored dystrophin in the diaphragm and assessed cardiac function by MRI. This approach reduced diaphragmatic pathophysiology and markedly improved diaphragm function but did not improve cardiac function or pathophysiology, with or without exercise. Interestingly, exercise resulted in a reduction of dystrophin protein and exon skipping in the diaphragm. This suggests that treatment regimens may require modification in more active patients. In conclusion, whilst the diaphragm is an important respiratory muscle, it is likely that dystrophin needs to be restored in other tissues, including multiple accessory respiratory muscles, and of course the heart itself for appropriate therapeutic outcomes. This supports the requirement of a body-wide therapy to treat DMD.


Subject(s)
Diaphragm/physiopathology , Disease Models, Animal , Heart/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Amino Acid Sequence , Animals , Atrial Natriuretic Factor , Blotting, Western , Diaphragm/diagnostic imaging , Diaphragm/metabolism , Dystrophin/genetics , Dystrophin/metabolism , Gene Expression/drug effects , Humans , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Mice, Inbred mdx , Morpholinos/chemistry , Morpholinos/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/genetics , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Natriuretic Peptide, C-Type/genetics , Natriuretic Peptide, C-Type/metabolism , Peptides/chemistry , Peptides/pharmacology , Physical Conditioning, Animal/physiology , Protein Precursors/genetics , Protein Precursors/metabolism , Radiography , Reverse Transcriptase Polymerase Chain Reaction
16.
Sci Rep ; 5: 8986, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25758104

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal neuromuscular disorder caused by mutations in the Dmd gene. In addition to skeletal muscle wasting, DMD patients develop cardiomyopathy, which significantly contributes to mortality. Antisense oligonucleotides (AOs) are a promising DMD therapy, restoring functional dystrophin protein by exon skipping. However, a major limitation with current AOs is the absence of dystrophin correction in heart. Pip peptide-AOs demonstrate high activity in cardiac muscle. To determine their therapeutic value, dystrophic mdx mice were subject to forced exercise to model the DMD cardiac phenotype. Repeated peptide-AO treatments resulted in high levels of cardiac dystrophin protein, which prevented the exercised induced progression of cardiomyopathy, normalising heart size as well as stabilising other cardiac parameters. Treated mice also exhibited significantly reduced cardiac fibrosis and improved sarcolemmal integrity. This work demonstrates that high levels of cardiac dystrophin restored by Pip peptide-AOs prevents further deterioration of cardiomyopathy and pathology following exercise in dystrophic DMD mice.


Subject(s)
Cardiomyopathies/etiology , Dystrophin/genetics , Morpholinos/administration & dosage , Muscular Dystrophy, Duchenne/complications , Physical Conditioning, Animal/adverse effects , Animals , Biomarkers , Cardiomyopathies/diagnosis , Cardiomyopathies/metabolism , Cardiomyopathies/prevention & control , Cardiomyopathies/therapy , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/prevention & control , Cardiomyopathy, Dilated/therapy , Disease Models, Animal , Dystrophin/metabolism , Fibrosis , Gene Expression , Humans , Magnetic Resonance Imaging, Cine , Mice , Mice, Inbred mdx , Myocardium/metabolism , Myocardium/pathology , Phenotype
17.
Nucleic Acid Ther ; 25(2): 65-77, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25594433

ABSTRACT

Oligonucleotide analogs have provided novel therapeutics targeting various disorders. However, their poor cellular uptake remains a major obstacle for their clinical development. Negatively charged oligonucleotides, such as 2'-O-Methyl RNA and locked nucleic acids have in recent years been delivered successfully into cells through complex formation with cationic polymers, peptides, liposomes, or similar nanoparticle delivery systems. However, due to the lack of electrostatic interactions, this promising delivery method has been unsuccessful to date using charge-neutral oligonucleotide analogs. We show here that lipid-functionalized cell-penetrating peptides can be efficiently exploited for cellular transfection of the charge-neutral oligonucleotide analog phosphorodiamidate morpholino. The lipopeptides form complexes with splice-switching phosphorodiamidate morpholino oligonucleotide and can be delivered into clinically relevant cell lines that are otherwise difficult to transfect while retaining biological activity. To our knowledge, this is the first study to show delivery through complex formation of biologically active charge-neutral oligonucleotides by cationic peptides.


Subject(s)
Agammaglobulinemia/drug therapy , Genetic Diseases, X-Linked/drug therapy , Morpholinos/administration & dosage , Muscular Atrophy, Spinal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Nanoparticles , Peptides/administration & dosage , Amino Acid Sequence , Animals , Cells, Cultured , Disease Models, Animal , Mice , Molecular Sequence Data , Morpholinos/therapeutic use , Peptides/chemistry
18.
Mol Ther Nucleic Acids ; 3: e212, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25423116

ABSTRACT

Splice switching oligonucleotides (SSOs) induce alternative splicing of pre-mRNA and typically employ chemical modifications to increase nuclease resistance and binding affinity to target pre-mRNA. Here we describe a new SSO non-base modifier (a naphthyl-azo group, "ZEN™") to direct exon exclusion in mutant dystrophin pre-mRNA to generate functional dystrophin protein. The ZEN modifier is placed near the ends of a 2'-O-methyl (2'OMe) oligonucleotide, increasing melting temperature and potency over unmodified 2'OMe oligonucleotides. In cultured H2K cells, a ZEN-modified 2'OMe phosphorothioate (PS) oligonucleotide delivered by lipid transfection greatly enhanced dystrophin exon skipping over the same 2'OMePS SSO lacking ZEN. However, when tested using free gymnotic uptake in vitro and following systemic delivery in vivo in dystrophin deficient mdx mice, the same ZEN-modified SSO failed to enhance potency. Importantly, we show for the first time that in vivo activity of anionic SSOs is modelled in vitro only when using gymnotic delivery. ZEN is thus a novel modifier that enhances activity of SSOs in vitro but will require improved delivery methods before its in vivo clinical potential can be realized.

19.
Nucleic Acid Ther ; 24(1): 13-24, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24506779

ABSTRACT

Targeting of pre-mRNA by short splice-switching oligonucleotides (SSOs) is increasingly being used as a therapeutic modality, one rationale being to disrupt splicing so as to remove exons containing premature termination codons, or to restore the translation reading frame around out-of-frame deletion mutations. The aim of this study was to investigate the effect of chemically linking individual SSOs so as to ascertain equimolar cellular uptake that would provide for more defined drug formulations. In contrast to conventional bispecific SSOs generated by conjugation in solution, here we describe a protocol for synthesis of bispecific SSOs on solid phase. These SSOs comprised of either a non-cleavable hydrocarbon linker or disulfide-based cleavable linkers. To assess the efficacy of these SSOs we have utilized splice switching to bypass a disease-causing mutation in the DMD gene concurrent with disruption of the reading frame of the myostatin gene (Mstn). The premise of this approach is that disruption of myostatin expression is known to induce muscle hypertrophy and so for Duchenne muscular dystrophy (DMD) could be expected to have a better outcome than dystrophin restoration alone. All tested SSOs mediated simultaneous robust exon removal from mature Dmd and Mstn transcripts in myotubes. Our results also demonstrate that using cleavable SSOs is preferred over the non-cleavable counterparts and that these are equally efficient at inducing exon skipping as cocktails of monospecific versions. In conclusion, we have developed a protocol for solid-phase synthesis of single molecule cleavable bispecific SSOs that can be efficiently exploited for targeting of multiple RNA transcripts.


Subject(s)
Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Targeted Gene Repair/methods , Animals , Base Sequence , Cell Line , Dystrophin/genetics , Exons , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Mutation , Myostatin/genetics , RNA Splicing/genetics
20.
Curr Gene Ther ; 12(3): 161-78, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22533378

ABSTRACT

The hydrophobic plasma membrane constitutes an indispensable barrier for cells, allowing influx of essential molecules while preventing access to other macromolecules. Although pivotal for the maintenance of cells, the inability to cross the plasma membrane is one of the major obstacles toward current drug development. Oligonucleotides (ONs) are a group of substances that display great therapeutic potential to interfere with gene expression. Several classes of ONs have emerged either based on double stranded RNAs, such as short interfering RNAs that are utilized to confer gene silencing, or single stranded ONs of various chemistries for antisense targeting of small regulatory micro RNAs or mRNAs. In particular the use of splice switching oligonucleotides (SSOs) to manipulate alternative splicing, by targeting pre-mRNA, has proven to be a highly promising therapeutic strategy to treat various genetic disorders, including Duchenne muscular dystrophy and spinal muscular atrophy. Despite being efficient compounds to alter splicing patterns, their hydrophilic macromolecular nature prohibits efficient cellular internalization.Various chemical drug delivery vehicles have been developed aiming at improving the bioavailability of nucleic acid-based drugs. In the context of SSOs, one group of peptidebased delivery vectors, i.e. cell-penetrating peptides (CPPs), display extremely high potency. CPPs have a remarkable ability to convey various, otherwise impermeable, macromolecules across the plasma membrane of cells in a relatively non-toxic fashion. This review provides insight into the application of CPPs and ONs in gene regulation with particular focus on CPP-assisted delivery of therapeutic SSOs.


Subject(s)
Alternative Splicing/genetics , Cell-Penetrating Peptides , Muscular Atrophy, Spinal , Muscular Dystrophy, Duchenne , Oligonucleotides , Cell Membrane/metabolism , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/therapeutic use , Gene Expression Regulation , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Targeted Therapy , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Oligonucleotides/genetics , Oligonucleotides/metabolism , Oligonucleotides/therapeutic use , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...