Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 231
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 148(6): 1085-8, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22424220

ABSTRACT

Dysregulation of splicing and alternative splicing underlies many genetic and acquired diseases. We present an overview of recent strategies and successes in modulating splicing therapeutically in clinical and preclinical contexts. Effective approaches include restoring open reading frames, influencing alternative splicing, and inducing exon inclusion to generate beneficial proteins and remove deleterious ones.


Subject(s)
Disease/genetics , Genetic Therapy , RNA Splicing/drug effects , Alternative Splicing , Animals , Humans , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Mutation , Neoplasms/genetics , Neoplasms/therapy , Oligonucleotides, Antisense/therapeutic use , Progeria/genetics , Progeria/therapy
2.
RNA ; 29(4): 446-454, 2023 04.
Article in English | MEDLINE | ID: mdl-36669889

ABSTRACT

Splice-modulating antisense oligonucleotides (ASOs) offer treatment options for rare neurological diseases, including those with very rare mutations, where patient-specific, individualized ASOs have to be developed. Inspired by the development of milasen, the 1 Mutation 1 Medicine (1M1M) and Dutch Center for RNA Therapeutics (DCRT) aim to develop patient-specific ASOs and treat eligible patients within Europe and the Netherlands, respectively. Treatment will be provided under a named patient setting. Our initiatives benefited from regulatory advice from the European Medicines Agency (EMA) with regard to preclinical proof-of-concept studies, safety studies, compounding and measuring benefit and safety in treated patients. We here outline the most important considerations from these interactions and how we implemented this advice into our plan to develop and treat eligible patients within Europe.


Subject(s)
Brain Diseases , Oligonucleotides , Humans , Oligonucleotides/genetics , Oligonucleotides/therapeutic use , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Brain , Europe , Brain Diseases/drug therapy
4.
Am J Med Genet C Semin Med Genet ; 193(1): 19-29, 2023 03.
Article in English | MEDLINE | ID: mdl-36453229

ABSTRACT

Rare genetic disorders affect as many as 3%-5% of all babies born. Approximately 10,000 such disorders have been identified or hypothesized to exist. Treatment is supportive except in a limited number of instances where specific therapies exist. Development of new therapies has been hampered by at least two major factors: difficulty in diagnosing diseases early enough to enable treatment before irreversible damage occurs, and the high cost of developing new drugs and getting them approved by regulatory agencies. Whole-genome sequencing (WGS) techniques have become exponentially less expensive and more rapid since the beginning of the human genome project, such that return of clinical data can now be achieved in days rather than years and at a cost that is comparable to other less expansive genetic testing. Thus, it is likely that WGS will ultimately become a mainstream, first-tier NBS technique at least for those disorders without appropriate high-throughput functional tests. However, there are likely to be several steps in the evolution to this end. The clinical implications of these advances are profound but highlight the bottlenecks in drug development that still limit transition to treatments. This article summarizes discussions arising from a recent National Institute of Health conference on nucleic acid therapy, with a focus on the impact of WGS in the identification of diagnosis and treatment of rare genetic disorders.


Subject(s)
Genetic Testing , Genetic Therapy , Humans , Genetic Testing/methods , Whole Genome Sequencing , Rare Diseases
5.
Genet Med ; 25(12): 100981, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37712502

ABSTRACT

PURPOSE: To summarize the results of first year implementation of pan-ethnic screening testing for Duchenne muscular dystrophy (DMD) and present the ensuing challenges. METHODS: Data acquisition for this study was performed by retrospective search of Ministry of Health registry for reports of all laboratories performing genetic screening tests. DMD testing was performed by multiplex ligation-dependent probe amplification technology. In case of single-exon deletion, sequencing of the specific exon was performed to rule out underlying single-nucleotide variant. RESULTS: Of overall 85,737 DMD tests, 82 clinically significant findings were noted (0.095%, or 1:1,046 women). In addition, 80 findings with uncertain clinical significance were detected (0.093%, or 1:1072), as well as 373 cases (0.4%, or 1:230) of single-exon deletions subsequently identified as false positives because of underlying single-nucleotide variant, mostly variants in exon 8 in North African Jewish population, and in exon 48 in Arab Muslim population. CONCLUSION: Interpretation of population-based DMD carrier screening is complex, occasionally requiring additional genetic testing methods and ethical considerations. Multicenter data registry, including ethnic origin and familial segregation in selected cases, is crucial for optimal definition of the results during genetic counseling and informed decisions regarding prenatal testing.


Subject(s)
Muscular Dystrophy, Duchenne , Female , Humans , Pregnancy , Dystrophin/genetics , Gene Deletion , Heterozygote , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/genetics , Mutation , Nucleotides , Retrospective Studies
6.
Clin Proteomics ; 20(1): 23, 2023 Jun 12.
Article in English | MEDLINE | ID: mdl-37308827

ABSTRACT

BACKGROUND: Molecular components in blood, such as proteins, are used as biomarkers to detect or predict disease states, guide clinical interventions and aid in the development of therapies. While multiplexing proteomics methods promote discovery of such biomarkers, their translation to clinical use is difficult due to the lack of substantial evidence regarding their reliability as quantifiable indicators of disease state or outcome. To overcome this challenge, a novel orthogonal strategy was developed and used to assess the reliability of biomarkers and analytically corroborate already identified serum biomarkers for Duchenne muscular dystrophy (DMD). DMD is a monogenic incurable disease characterized by progressive muscle damage that currently lacks reliable and specific disease monitoring tools. METHODS: Two technological platforms are used to detect and quantify the biomarkers in 72 longitudinally collected serum samples from DMD patients at 3 to 5 timepoints. Quantification of the biomarkers is achieved by detection of the same biomarker fragment either through interaction with validated antibodies in immuno-assays or through quantification of peptides by Parallel Reaction Monitoring Mass Spectrometry assay (PRM-MS). RESULTS: Five, out of ten biomarkers previously identified by affinity-based proteomics methods, were confirmed to be associated with DMD using the mass spectrometry-based method. Two biomarkers, carbonic anhydrase III and lactate dehydrogenase B, were quantified with two independent methods, sandwich immunoassays and PRM-MS, with Pearson correlations of 0.92 and 0.946 respectively. The median concentrations of CA3 and LDHB in DMD patients was elevated in comparison to those in healthy individuals by 35- and 3-fold, respectively. Levels of CA3 vary between 10.26 and 0.36 ng/ml in DMD patients whereas those of LDHB vary between 15.1 and 0.8 ng/ml. CONCLUSIONS: These results demonstrate that orthogonal assays can be used to assess the analytical reliability of biomarker quantification assays, providing a means to facilitate the translation of biomarkers to clinical practice. This strategy also warrants the development of the most relevant biomarkers, markers that can be reliably quantified with different proteomics methods.

7.
RNA Biol ; 20(1): 693-702, 2023 01.
Article in English | MEDLINE | ID: mdl-37667454

ABSTRACT

Mutations in the DMD gene are causative for Duchenne muscular dystrophy (DMD). Antisense oligonucleotide (AON) mediated exon skipping to restore disrupted dystrophin reading frame is a therapeutic approach that allows production of a shorter but functional protein. As DMD causing mutations can affect most of the 79 exons encoding dystrophin, a wide variety of AONs are needed to treat the patient population. Design of AONs is largely guided by trial-and-error, and it is yet unclear what defines the skippability of an exon. Here, we use a library of phosphorodiamidate morpholino oligomer (PMOs) AONs of similar physical properties to test the skippability of a large number of DMD exons. The DMD transcript is non-sequentially spliced, meaning that certain introns are retained longer in the transcript than downstream introns. We tested whether the relative intron retention time has a significant effect on AON efficiency, and found that targeting an out-of-frame exon flanked at its 5'-end by an intron that is retained in the transcript longer ('slow' intron) leads to overall higher exon skipping efficiency than when the 5'-end flanking intron is 'fast'. Regardless of splicing speed of flanking introns, we find that positioning an AON closer to the 5'-end of the target exon leads to higher exon skipping efficiency opposed to targeting an exons 3'-end. The data enclosed herein can be of use to guide future target selection and preferential AON binding sites for both DMD and other disease amenable by exon skipping therapies.


Subject(s)
Muscular Dystrophy, Duchenne , Oligonucleotides, Antisense , Humans , Oligonucleotides, Antisense/genetics , Introns , Dystrophin/genetics , Exons , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy
8.
Nucleic Acids Res ; 49(21): 12089-12105, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34850138

ABSTRACT

Antisense oligonucleotides are small pieces of modified DNA or RNA, which offer therapeutic potential for many diseases. We report on the synthesis of 7',5'-α-bc-DNA phosphoramidite building blocks, bearing the A, G, T and MeC nucleobases. Solid-phase synthesis was performed to construct five oligodeoxyribonucleotides containing modified thymidine residues, as well as five fully modified oligonucleotides. Incorporations of the modification inside natural duplexes resulted in strong destabilizing effects. However, fully modified strands formed very stable duplexes with parallel RNA complements. In its own series, 7',5'-α-bc-DNA formed duplexes with a surprising high thermal stability. CD spectroscopy and extensive molecular modeling indicated the adoption by the homo-duplex of a ladder-like structure, while hetero-duplexes with DNA or RNA still form helical structure. The biological properties of this new modification were investigated in animal models for Duchenne muscular dystrophy and spinal muscular atrophy, where exon splicing modulation can restore production of functional proteins. It was found that the 7',5'-α-bc-DNA scaffold confers a high biostability and a good exon splicing modulation activity in vitro and in vivo.


Subject(s)
DNA/therapeutic use , Genetic Therapy/methods , Muscular Atrophy, Spinal/therapy , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides , Animals , Cell Line , Male , Mice , Oligonucleotides/chemistry , Oligonucleotides/therapeutic use
9.
Proc Natl Acad Sci U S A ; 117(28): 16456-16464, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32616572

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene leading to the presence of premature termination codons (PTC). Previous transcriptional studies have shown reduced DMD transcript levels in DMD patient and animal model muscles when PTC are present. Nonsense-mediated decay (NMD) has been suggested to be responsible for the observed reduction, but there is no experimental evidence supporting this claim. In this study, we aimed to investigate the mechanism responsible for the drop in DMD expression levels in the presence of PTC. We observed that the inhibition of NMD does not normalize DMD gene expression in DMD. Additionally, in situ hybridization showed that DMD messenger RNA primarily localizes in the nuclear compartment, confirming that a cytoplasmic mechanism like NMD indeed cannot be responsible for the observed reduction. Sequencing of nascent RNA to explore DMD transcription dynamics revealed a lower rate of DMD transcription in patient-derived myotubes compared to healthy controls, suggesting a transcriptional mechanism involved in reduced DMD transcript levels. Chromatin immunoprecipitation in muscle showed increased levels of the repressive histone mark H3K9me3 in mdx mice compared to wild-type mice, indicating a chromatin conformation less prone to transcription in mdx mice. In line with this finding, treatment with the histone deacetylase inhibitor givinostat caused a significant increase in DMD transcript expression in mdx mice. Overall, our findings show that transcription dynamics across the DMD locus are affected by the presence of PTC, hinting at a possible epigenetic mechanism responsible for this process.


Subject(s)
Codon, Nonsense/genetics , Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , RNA, Messenger/genetics , Animals , Codon, Nonsense/metabolism , Disease Models, Animal , Dystrophin/metabolism , Humans , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/metabolism , Nonsense Mediated mRNA Decay , RNA, Messenger/metabolism
10.
Hum Mol Genet ; 29(5): 745-755, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32025735

ABSTRACT

Duchenne muscular dystrophy is a severe pediatric neuromuscular disorder caused by the lack of dystrophin. Identification of biomarkers is needed to support and accelerate drug development. Alterations of metabolites levels in muscle and plasma have been reported in pre-clinical and clinical cross-sectional comparisons. We present here a 7-month longitudinal study comparing plasma metabolomic data in wild-type and mdx mice. A mass spectrometry approach was used to study metabolites in up to five time points per mouse at 6, 12, 18, 24 and 30 weeks of age, providing an unprecedented in depth view of disease trajectories. A total of 106 metabolites were studied. We report a signature of 31 metabolites able to discriminate between healthy and disease at various stages of the disease, covering the acute phase of muscle degeneration and regeneration up to the deteriorating phase. We show how metabolites related to energy production and chachexia (e.g. glutamine) are affected in mdx mice plasma over time. We further show how the signature is connected to molecular targets of nutraceuticals and pharmaceutical compounds currently in development as well as to the nitric oxide synthase pathway (e.g. arginine and citrulline). Finally, we evaluate the signature in a second longitudinal study in three independent mouse models carrying 0, 1 or 2 functional copies of the dystrophin paralog utrophin. In conclusion, we report an in-depth metabolomic signature covering previously identified associations and new associations, which enables drug developers to peripherally assess the effect of drugs on the metabolic status of dystrophic mice.


Subject(s)
Disease Models, Animal , Metabolome , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/blood , Muscular Dystrophy, Duchenne/pathology , Animals , Cross-Sectional Studies , Disease Progression , Longitudinal Studies , Mice , Mice, Inbred mdx
11.
Hum Mol Genet ; 29(11): 1853-1863, 2020 07 21.
Article in English | MEDLINE | ID: mdl-31960911

ABSTRACT

CADASIL is a vascular protein aggregation disorder caused by cysteine-altering NOTCH3 variants, leading to mid-adult-onset stroke and dementia. Here, we report individuals with a cysteine-altering NOTCH3 variant that induces exon 9 skipping, mimicking therapeutic NOTCH3 cysteine correction. The index came to our attention after a coincidental finding on a commercial screening MRI, revealing white matter hyperintensities. A heterozygous NOTCH3 c.1492G>T, p.Gly498Cys variant, was identified using a gene panel, which was also present in four first- and second-degree relatives. Although some degree of white matter hyperintensities was present on MRI in all family members with the NOTCH3 variant, the CADASIL phenotype was mild, as none had lacunes on MRI and there was no disability or cognitive impairment above the age of 60 years. RT-PCR and Sanger sequencing analysis on patient fibroblast RNA revealed that exon 9 was absent from the majority of NOTCH3 transcripts of the mutant allele, effectively excluding the mutation. NOTCH3 aggregation was assessed in skin biopsies using electron microscopy and immunohistochemistry and did not show granular osmiophilic material and only very mild NOTCH3 staining. For purposes of therapeutic translatability, we show that, in cell models, exon 9 exclusion can be obtained using antisense-mediated exon skipping and CRISPR/Cas9-mediated genome editing. In conclusion, this study provides the first in-human evidence that cysteine corrective NOTCH3 exon skipping is associated with less NOTCH3 aggregation and an attenuated phenotype, justifying further therapeutic development of NOTCH3 cysteine correction for CADASIL.


Subject(s)
CADASIL/genetics , Cysteine/genetics , Protein Aggregation, Pathological/genetics , Receptor, Notch3/genetics , White Matter/metabolism , Adult , Aged , Biopsy , CADASIL/diagnostic imaging , CADASIL/metabolism , CADASIL/physiopathology , CRISPR-Cas Systems/genetics , Exons/genetics , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Protein Aggregation, Pathological/diagnostic imaging , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Severity of Illness Index , Skin/chemistry , Skin/diagnostic imaging , White Matter/diagnostic imaging , White Matter/pathology
12.
J Inherit Metab Dis ; 44(1): 72-87, 2021 01.
Article in English | MEDLINE | ID: mdl-32391605

ABSTRACT

Antisense oligonucleotide (AON) therapies involve short strands of modified nucleotides that target RNA in a sequence-specific manner, inducing targeted protein knockdown or restoration. Currently, 10 AON therapies have been approved in the United States and Europe. Nucleotides are chemically modified to protect AONs from degradation, enhance bioavailability and increase RNA affinity. Whereas single stranded AONs can efficiently be delivered systemically, delivery of double stranded AONs requires capsulation in lipid nanoparticles or binding to a conjugate as the uptake enhancing backbone is hidden in this conformation. With improved chemistry, delivery vehicles and conjugates, doses can be lowered, thereby reducing the risk and occurrence of side effects. AONs can be used to knockdown or restore levels of protein. Knockdown can be achieved by single stranded or double stranded AONs binding the RNA transcript and activating RNaseH-mediated and RISC-mediated degradation respectively. Transcript binding by AONs can also prevent translation, hence reducing protein levels. For protein restoration, single stranded AONs are used to modulate pre-mRNA splicing and either include or skip an exon to restore protein production. Intervening at a genetic level, AONs provide therapeutic options for inherited metabolic diseases as well. This review provides an overview of the different AON approaches, with a focus on AONs developed for inborn errors of metabolism.


Subject(s)
Exons , Oligonucleotides, Antisense/pharmacology , RNA Splicing/genetics , RNA, Messenger/chemistry , Animals , Gene Knockdown Techniques , Humans , Nucleic Acid Conformation
13.
Mol Cell Neurosci ; 105: 103484, 2020 06.
Article in English | MEDLINE | ID: mdl-32240725

ABSTRACT

This study investigates changes with respect to increasing protein levels in dystrophic nerves of two mdx mouse models of Duchenne muscular dystrophy (DMD). We propose that these nerve changes result from progressive ongoing damage to neuromuscular junctions (NMJs) due to repeated intrinsic bouts of necrosis in dystrophic muscles. We compared sciatic nerves from classic mdx mice aged 13, 15 and 18 months (M), with D2.mdx mice (on DBA2 background) aged 9 and 13 M, using immunoblotting to quantify levels of 7 proteins. The neuronal proteins S100ß and Tau5 were increased by 13 M in mdx nerves (compared with WT), indicating ongoing myonecrosis in this strain. In striking contrast there was no difference in levels of these neuronal proteins for D2.mdx and D2.WT sciatic nerves at 13 M, indicating reduced myonecrosis over this time in D2.mdx mice compared with mdx. These novel changes in mdx sciatic nerves by 13 M, suggest early denervation or neurodegeneration of dystrophic nerves that is likely irreversible and progressive. This neuronal readout of persistent myonecrosis may provide a useful new long-term biomarker for preclinical studies that aim to reduce myonecrosis, plus such neuronal changes present potential new drug targets to help maintain the function of DMD muscles.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , tau Proteins/metabolism , Animals , Disease Models, Animal , Mice, Inbred mdx , Neuromuscular Junction/metabolism
14.
FASEB J ; 33(7): 8110-8124, 2019 07.
Article in English | MEDLINE | ID: mdl-30933664

ABSTRACT

The C57BL/10ScSn-Dmdmdx/J (BL10-mdx) mouse has been the most commonly used model for Duchenne muscular dystrophy (DMD) for decades. Their muscle dysfunction and pathology is, however, less severe than in patients with DMD, which complicates preclinical studies. Recent discoveries indicate that disease severity is exacerbated when muscular dystrophy mouse models are generated on a DBA2/J genetic background. Knowledge on the natural history of animal models is pivotal for high-quality preclinical testing. However, for BL10-mdx mice on a DBA2/J background (D2-mdx), limited data are available. We addressed this gap in the natural history knowledge. First, we compared histopathological aspects in skeletal muscles of young D2-mdx, BL10-mdx, and wild-type mice. Pathology was more pronounced in D2-mdx mice and differed in severity between muscles within individuals. Secondly, we subjected D2-mdx mice to a functional test regime for 34 weeks and identified that female D2-mdx mice outperform severely impaired males, making females less useful for functional preclinical studies. Direct comparisons between 10- and 34-wk-old D2-mdx mice revealed that disease pathology ameliorates with age. Heart pathology was progressive, with some features already evident at a young age. This natural history study of the D2-mdx mouse will be instrumental for experimental design of future preclinical studies.-Van Putten, M., Putker, K., Overzier, M., Adamzek, W. A., Pasteuning-Vuhman, S., Plomp, J. J., Aartsma-Rus, A. Natural disease history of the D2-mdx mouse model for Duchenne muscular dystrophy.


Subject(s)
Muscular Dystrophy, Animal , Muscular Dystrophy, Duchenne , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred mdx , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology
15.
Hum Mutat ; 40(10): 1630-1633, 2019 10.
Article in English | MEDLINE | ID: mdl-31356707

ABSTRACT

Variations in the DMD gene that affect dystrophin production underlie both the severe Duchenne and the milder Becker muscular dystrophies (DMD and BMD, respectively). Depending on their location, deletions and duplications involving one or more exons of a gene can have a range of consequences. This overview, summarizing the important points to consider, was drafted in response to frequent questions we receive about deletions/duplications involving the dystrophin encoding DMD gene. Although directed at DMD, the observations made can be applied to many other genes. The overview is meant primarily for healthcare professionals involved with interpreting the results of genetic analyses in clinical practice.


Subject(s)
Exons , Gene Deletion , Gene Duplication , Genetic Association Studies , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/genetics , Phenotype , Genetic Predisposition to Disease , Humans
16.
Am J Hum Genet ; 99(5): 1163-1171, 2016 Nov 03.
Article in English | MEDLINE | ID: mdl-27745838

ABSTRACT

The expressivity of Mendelian diseases can be influenced by factors independent from the pathogenic mutation: in Duchenne muscular dystrophy (DMD), for instance, age at loss of ambulation (LoA) varies between individuals whose DMD mutations all abolish dystrophin expression. This suggests the existence of trans-acting variants in modifier genes. Common single nucleotide polymorphisms (SNPs) in candidate genes (SPP1, encoding osteopontin, and LTBP4, encoding latent transforming growth factor ß [TGFß]-binding protein 4) have been established as DMD modifiers. We performed a genome-wide association study of age at LoA in a sub-cohort of European or European American ancestry (n = 109) from the Cooperative International Research Group Duchenne Natural History Study (CINRG-DNHS). We focused on protein-altering variants (Exome Chip) and included glucocorticoid treatment as a covariate. As expected, due to the small population size, no SNPs displayed an exome-wide significant p value (< 1.8 × 10-6). Subsequently, we prioritized 438 SNPs in the vicinities of 384 genes implicated in DMD-related pathways, i.e., the nuclear-factor-κB and TGFß pathways. The minor allele at rs1883832, in the 5'-untranslated region of CD40, was associated with earlier LoA (p = 3.5 × 10-5). This allele diminishes the expression of CD40, a co-stimulatory molecule for T cell polarization. We validated this association in multiple independent DMD cohorts (United Dystrophinopathy Project, Bio-NMD, and Padova, total n = 660), establishing this locus as a DMD modifier. This finding points to cell-mediated immunity as a relevant pathogenetic mechanism and potential therapeutic target in DMD.


Subject(s)
CD40 Antigens/genetics , Muscular Dystrophy, Duchenne/genetics , NF-kappa B/genetics , Polymorphism, Single Nucleotide , Transforming Growth Factor beta/genetics , Adolescent , Alleles , CD40 Antigens/metabolism , Case-Control Studies , Child , Dystrophin/genetics , Dystrophin/metabolism , Exons , Genes, Modifier , Genome-Wide Association Study , Glucocorticoids/pharmacology , Humans , Latent TGF-beta Binding Proteins/genetics , Latent TGF-beta Binding Proteins/metabolism , Mutation , NF-kappa B/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Transforming Growth Factor beta/metabolism , White People/genetics
17.
Mol Ther ; 26(1): 132-147, 2018 01 03.
Article in English | MEDLINE | ID: mdl-29103911

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe, progressive muscle wasting disorder caused by reading frame disrupting mutations in the DMD gene. Exon skipping is a therapeutic approach for DMD. It employs antisense oligonucleotides (AONs) to restore the disrupted open reading frame, allowing the production of shorter, but partly functional dystrophin protein as seen in less severely affected Becker muscular dystrophy patients. To be effective, AONs need to be delivered and effectively taken up by the target cells, which can be accomplished by the conjugation of tissue-homing peptides. We performed phage display screens using a cyclic peptide library combined with next generation sequencing analyses to identify candidate muscle-homing peptides. Conjugation of the lead peptide to 2'-O-methyl phosphorothioate AONs enabled a significant, 2-fold increase in delivery and exon skipping in all analyzed skeletal and cardiac muscle of mdx mice and appeared well tolerated. While selected as a muscle-homing peptide, uptake was increased in liver and kidney as well. The homing capacity of the peptide may have been overruled by the natural biodistribution of the AON. Nonetheless, our results suggest that the identified peptide has the potential to facilitate delivery of AONs and perhaps other compounds to skeletal and cardiac muscle.


Subject(s)
Alternative Splicing , Gene Transfer Techniques , Genetic Therapy , Muscular Dystrophy, Duchenne/genetics , Oligonucleotides, Antisense/genetics , Peptides, Cyclic , Amino Acid Sequence , Animals , Disease Models, Animal , Dystrophin/genetics , Exons , Humans , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/chemistry , Peptide Library , Peptides, Cyclic/chemistry
18.
J Mol Cell Cardiol ; 125: 29-38, 2018 12.
Article in English | MEDLINE | ID: mdl-30336143

ABSTRACT

Duchenne muscular dystrophy is a severe muscle wasting disease, characterized by a severely reduced lifespan in which cardiomyopathy is one of the leading causes of death. Multiple therapies aiming at dystrophin restoration have been approved. It is anticipated that these therapies will maintain muscle function for longer and extend the ambulatory period, which in turn will increase the cardiac workload which could be detrimental for cardiac function. We investigated the effects of voluntary running exercise in combination with low dystrophin levels on function and pathology of skeletal muscle and heart. We divided 15.5-month old female mdx (no dystrophin), mdx-XistΔhs (varying low dystrophin levels) and wild type mice (BL10-WT and XistΔhs-WT) to either a sedentary or voluntary wheel running regime and assessed muscle function at 17.5 months of age. Thereafter, a cardiac MRI was obtained, and muscle and heart histopathology were assessed. We show that voluntary exercise is beneficial to skeletal muscle and heart function in dystrophic mice while not affecting muscle pathology. Low amounts of dystrophin further improve skeletal muscle and cardiac function. These findings suggest that voluntary exercise may be beneficial for skeletal muscle and heart in DMD patients, especially in conjunction with low amounts of dystrophin.


Subject(s)
Heart/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Myocardium/metabolism , Physical Conditioning, Animal/physiology , Animals , Blotting, Western , Female , Male , Motor Activity/physiology , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism
19.
J Cell Mol Med ; 22(4): 2442-2448, 2018 04.
Article in English | MEDLINE | ID: mdl-29441734

ABSTRACT

Muscular dystrophies are characterized by a progressive loss of muscle tissue and/or muscle function. While metabolic alterations have been described in patients'-derived muscle biopsies, non-invasive readouts able to describe these alterations are needed in order to objectively monitor muscle condition and response to treatment targeting metabolic abnormalities. We used a metabolomic approach to study metabolites concentration in serum of patients affected by multiple forms of muscular dystrophy such as Duchenne and Becker muscular dystrophies, limb-girdle muscular dystrophies type 2A and 2B, myotonic dystrophy type 1 and facioscapulohumeral muscular dystrophy. We show that 15 metabolites involved in energy production, amino acid metabolism, testosterone metabolism and response to treatment with glucocorticoids were differentially expressed between healthy controls and Duchenne patients. Five metabolites were also able to discriminate other forms of muscular dystrophy. In particular, creatinine and the creatine/creatinine ratio were significantly associated with Duchenne patients performance as assessed by the 6-minute walk test and north star ambulatory assessment. The obtained results provide evidence that metabolomics analysis of serum samples can provide useful information regarding muscle condition and response to treatment, such as to glucocorticoids treatment.


Subject(s)
Metabolomics , Muscles/metabolism , Muscular Dystrophies/blood , Adolescent , Adult , Female , Humans , Male , Middle Aged , Muscles/pathology , Muscular Dystrophies/classification , Muscular Dystrophies/pathology , Muscular Dystrophies, Limb-Girdle/blood , Muscular Dystrophies, Limb-Girdle/pathology , Muscular Dystrophy, Duchenne/blood , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Facioscapulohumeral/blood , Muscular Dystrophy, Facioscapulohumeral/pathology , Myotonic Dystrophy/blood , Myotonic Dystrophy/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL