Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
1.
Cells ; 12(18)2023 09 15.
Article in English | MEDLINE | ID: mdl-37759507

ABSTRACT

Interleukin-6 (IL-6) is a pleiotropic cytokine that plays a crucial role in maintaining normal homeostatic processes under the pathogenesis of various inflammatory and autoimmune diseases. This context-dependent effect from a cytokine is due to two distinctive forms of signaling: cis-signaling and trans-signaling. IL-6 cis-signaling involves binding IL-6 to the membrane-bound IL-6 receptor and Glycoprotein 130 (GP130) signal-transducing subunit. By contrast, in IL-6 trans-signaling, complexes of IL-6 and the soluble form of the IL-6 receptor (sIL-6R) signal via membrane-bound GP130. Various strategies have been employed in the past decade to target the pro-inflammatory effect of IL-6 in numerous inflammatory disorders. However, their development has been hindered since these approaches generally target global IL-6 signaling, also affecting the anti-inflammatory effects of IL-6 signaling too. Therefore, novel strategies explicitly targeting the pro-inflammatory IL-6 trans-signaling without affecting the IL-6 cis-signaling are required and carry immense therapeutic potential. Here, we have developed a novel approach to specifically decoy IL-6-mediated trans-signaling by modulating alternative splicing in GP130, an IL-6 signal transducer, by employing splice switching oligonucleotides (SSO), to induce the expression of truncated soluble isoforms of the protein GP130. This isoform is devoid of signaling domains but allows for specifically sequestering the IL-6/sIL-6R receptor complex with high affinity in serum and thereby suppressing inflammation. Using the state-of-the-art Pip6a cell-penetrating peptide conjugated to PMO-based SSO targeting GP130 for efficient in vivo delivery, reduced disease phenotypes in two different inflammatory mouse models of systemic and intestinal inflammation were observed. Overall, this novel gene therapy platform holds great potential as a refined therapeutic intervention for chronic inflammatory diseases.


Subject(s)
Cytokines , Interleukin-6 , Animals , Mice , Cytokine Receptor gp130 , Inflammation , Oligonucleotides
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 ; 2434: 3-31, 2022.
Article in English | MEDLINE | ID: mdl-35213007

ABSTRACT

This introduction charts the history of the development of the major chemical modifications that have influenced the development of nucleic acids therapeutics focusing in particular on antisense oligonucleotide analogues carrying modifications in the backbone and sugar. Brief mention is made of siRNA development and other applications that have by and large utilized the same modifications. We also point out the pitfalls of the use of nucleic acids as drugs, such as their unwanted interactions with pattern recognition receptors, which can be mitigated by chemical modification or used as immunotherapeutic agents.


Subject(s)
Nucleic Acids , Nucleic Acids/genetics , Nucleic Acids/therapeutic use , Oligonucleotides/genetics , Oligonucleotides/therapeutic use , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use
4.
Life Sci Alliance ; 4(10)2021 10.
Article in English | MEDLINE | ID: mdl-34389686

ABSTRACT

Absence of dystrophin, an essential sarcolemmal protein required for muscle contraction, leads to the devastating muscle-wasting disease Duchenne muscular dystrophy. Dystrophin has an actin-binding domain, which binds and stabilises filamentous-(F)-actin, an integral component of the RhoA-actin-serum-response-factor-(SRF) pathway. This pathway plays a crucial role in circadian signalling, whereby the suprachiasmatic nucleus (SCN) transmits cues to peripheral tissues, activating SRF and transcription of clock-target genes. Given dystrophin binds F-actin and disturbed SRF-signalling disrupts clock entrainment, we hypothesised dystrophin loss causes circadian deficits. We show for the first time alterations in the RhoA-actin-SRF-signalling pathway, in dystrophin-deficient myotubes and dystrophic mouse models. Specifically, we demonstrate reduced F/G-actin ratios, altered MRTF levels, dysregulated core-clock and downstream target-genes, and down-regulation of key circadian genes in muscle biopsies from Duchenne patients harbouring an array of mutations. Furthermore, we show dystrophin is absent in the SCN of dystrophic mice which display disrupted circadian locomotor behaviour, indicative of disrupted SCN signalling. Therefore, dystrophin is an important component of the RhoA-actin-SRF pathway and novel mediator of circadian signalling in peripheral tissues, loss of which leads to circadian dysregulation.


Subject(s)
Dystrophin/metabolism , Serum Response Factor/metabolism , Signal Transduction , Actins/metabolism , Animals , Cell Line , Dystrophin/genetics , Mice , Myoblasts, Skeletal/metabolism , Utrophin/metabolism , rhoA GTP-Binding Protein/metabolism
5.
Nucleic Acid Ther ; 31(2): 172-181, 2021 04.
Article in English | MEDLINE | ID: mdl-33567244

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe muscle-wasting disease caused by frameshift or nonsense mutations in the DMD gene, resulting in the loss of dystrophin from muscle membranes. Exon skipping using splice-switching oligonucleotides (SSOs) restores the reading frame of DMD pre-mRNA by generating internally truncated but functional dystrophin protein. To potentiate effective tissue-specific targeting by functional SSOs, it is essential to perform accelerated and reliable in vitro screening-based assessment of novel oligonucleotides and drug delivery technologies, such as cell-penetrating peptides, before their in vivo pharmacokinetic and toxicity evaluation. We have established novel canine immortalized myoblast lines by transducing murine cyclin-dependent kinase-4 and human telomerase reverse transcriptase genes into myoblasts isolated from beagle-based wild-type or canine X-linked muscular dystrophy in Japan (CXMDJ) dogs. These myoblast lines exhibited improved myogenic differentiation and increased proliferation rates compared with passage-15 primary parental myoblasts, and their potential to differentiate into myotubes was maintained in later passages. Using these dystrophin-deficient immortalized myoblast lines, we demonstrate that a novel cell-penetrating peptide (Pip8b2)-conjugated SSO markedly improved multiexon skipping activity compared with the respective naked phosphorodiamidate morpholino oligomers. In vitro screening using immortalized canine cell lines will provide a basis for further pharmacological studies on drug delivery tools.


Subject(s)
Cyclin-Dependent Kinase 4/genetics , Dystrophin/genetics , Morpholinos/pharmacology , Muscular Dystrophy, Duchenne/therapy , Telomerase/genetics , Animals , Cell Line , Dogs , Exons/genetics , Genetic Therapy , Humans , Mice , Morpholinos/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Myoblasts/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Peptides/genetics , Peptides/pharmacology , RNA Splice Sites/genetics
6.
J Clin Invest ; 129(11): 4739-4744, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31479430

ABSTRACT

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.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Muscle, Skeletal/metabolism , Myotonic Dystrophy , Myotonin-Protein Kinase , Oligodeoxyribonucleotides, Antisense , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Mice , Muscle, Skeletal/pathology , Myotonic Dystrophy/drug therapy , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
7.
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
8.
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
9.
Nucleic Acid Ther ; 29(1): 1-12, 2019 02.
Article in English | MEDLINE | ID: mdl-30307373

ABSTRACT

The review starts with a historical perspective of the achievements of the Gait group in synthesis of oligonucleotides (ONs) and their peptide conjugates toward the award of the 2017 Oligonucleotide Therapeutic Society Lifetime Achievement Award. This acts as a prelude to the rewarding collaborative studies in the Gait and Wood research groups aimed toward the enhanced delivery of charge neutral ON drugs and the development of a series of Arg-rich cell-penetrating peptides called Pip (peptide nucleic acid/phosphorodiamidate morpholino oligonucleotide [PNA/PMO] internalization peptides) as conjugates of such ONs. In this review we concentrate on these developments toward the treatment of the neuromuscular diseases Duchenne muscular dystrophy and spinal muscular atrophy toward a platform technology for the enhancement of cellular and in vivo delivery suitable for widespread use as neuromuscular and neurodegenerative ON drugs.


Subject(s)
Cell-Penetrating Peptides/therapeutic use , Muscular Atrophy, Spinal/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Neuromuscular Diseases/drug therapy , Cell-Penetrating Peptides/genetics , Humans , Morpholinos/genetics , Morpholinos/therapeutic use , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Neuromuscular Diseases/genetics , Neuromuscular Diseases/pathology , Peptide Nucleic Acids/genetics , Peptide Nucleic Acids/therapeutic use
10.
Front Chem ; 5: 81, 2017.
Article in English | MEDLINE | ID: mdl-29094037

ABSTRACT

Antisense oligonucleotide (ASO)-based drug development is gaining significant momentum following the recent FDA approval of Eteplirsen (an ASO based on phosphorodiamidate morpholino) and Spinraza (2'-O-methoxyethyl-phosphorothioate) in late 2016. Their attractiveness is mainly due to the backbone modifications which have improved the in vivo characteristics of oligonucleotide drugs. Another class of ASO, based on peptide nucleic acid (PNA) chemistry, is also gaining popularity as a platform for development of gene-specific therapy for various disorders. However, the chemical synthesis of long PNAs, which are more target-specific, remains an ongoing challenge. Most of the reported methodology for the solid-phase synthesis of PNA suffer from poor coupling efficiency which limits production to short PNA sequences of less than 15 residues. Here, we have studied the effect of backbone modifications with Hmb (2-hydroxy-4-methoxybenzyl) and Dmb (2,4-dimethoxybenzyl) to ameliorate difficult couplings and reduce "on-resin" aggregation. We firstly synthesized a library of PNA dimers incorporating either Hmb or Dmb and identified that Hmb is superior to Dmb in terms of its ease of removal. Subsequently, we used Hmb backbone modification to synthesize a 22-mer purine-rich PNA, targeting dystrophin RNA splicing, which could not be synthesized by standard coupling methodology. Hmb backbone modification allowed this difficult PNA to be synthesized as well as to be continued to include a cell-penetrating peptide on the same solid support. This approach provides a novel and straightforward strategy for facile solid-phase synthesis of difficult purine-rich PNA sequences.

11.
Interface Focus ; 7(2): 20160117, 2017 Apr 06.
Article in English | MEDLINE | ID: mdl-28382203

ABSTRACT

There is an increasing interest in targeting molecules to the mitochondrial matrix. Many proteins are naturally imported through the translocase complexes found in the outer and inner mitochondrial membranes. One possible means for importing molecules is therefore to use a mitochondrial pre-protein as a vector and assess what forms of molecules can be attached to the pre-protein as cargo. A major difficulty with this approach is to ensure that any chimaeric molecule does indeed access the mitochondrial matrix and does not merely associate with the mitochondrial membranes. We have recently demonstrated that click chemistry can be used both to demonstrate convincingly mitochondrial import of a peptide-peptide nucleic acid conjugate and also to quantify the mitochondrial uptake for specific synthetic conjugates. We now report an adaptation of the synthesis to facilitate simple quantification of multiple molecules and hence to calculate the efficiency of their mitochondrial import.

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
14.
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
15.
Chembiochem ; 17(14): 1312-6, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27124570

ABSTRACT

Mitochondria are central to health and disease, hence there is considerable interest in developing mitochondria-targeted therapies that require the delivery of peptides or nucleic acid oligomers. However, progress has been impeded by the lack of a measure of mitochondrial import of these molecules. Here, we address this need by quantitatively detecting molecules within the mitochondrial matrix. We used a mitochondria- targeted cyclooctyne (MitoOct) that accumulates several- hundredfold in the matrix, driven by the membrane potential. There, MitoOct reacts through click chemistry with an azide on the target molecule to form a diagnostic product that can be quantified by mass spectrometry. Because the membrane potential-dependent MitoOct concentration in the matrix is essential for conjugation, we can now determine definitively whether a putative mitochondrion-targeted molecule reaches the matrix. This "ClickIn" approach will facilitate development of mitochondria-targeted therapies.


Subject(s)
Click Chemistry/methods , Drug Delivery Systems/methods , Mitochondria/metabolism , Azides/analysis , Azides/chemistry , Azides/pharmacokinetics , Cyclooctanes/chemistry , Cyclooctanes/pharmacokinetics , Drug Carriers/chemistry , Humans , Mass Spectrometry , Mitochondrial Membranes/metabolism , Molecular Targeted Therapy/methods
16.
Sci Rep ; 5: 17014, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26594036

ABSTRACT

There is currently an urgent need for biomarkers that can be used to monitor the efficacy of experimental therapies for Duchenne Muscular Dystrophy (DMD) in clinical trials. Identification of novel protein biomarkers has been limited due to the massive complexity of the serum proteome and the presence of a small number of very highly abundant proteins. Here we have utilised an aptamer-based proteomics approach to profile 1,129 proteins in the serum of wild-type and mdx (dystrophin deficient) mice. The serum levels of 96 proteins were found to be significantly altered (P < 0.001, q < 0.01) in mdx mice. Additionally, systemic treatment with a peptide-antisense oligonucleotide conjugate designed to induce Dmd exon skipping and recover dystrophin protein expression caused many of the differentially abundant serum proteins to be restored towards wild-type levels. Results for five leading candidate protein biomarkers (Pgam1, Tnni3, Camk2b, Cycs and Adamts5) were validated by ELISA in the mouse samples. Furthermore, ADAMTS5 was found to be significantly elevated in human DMD patient serum. This study has identified multiple novel, therapy-responsive protein biomarkers in the serum of the mdx mouse with potential utility in DMD patients.


Subject(s)
ADAM Proteins/genetics , Aptamers, Nucleotide/pharmacology , Biomarkers, Pharmacological/blood , Dystrophin/genetics , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/pharmacology , ADAM Proteins/blood , ADAMTS5 Protein , Animals , Blood Proteins/genetics , Blood Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/blood , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Disease Models, Animal , Dystrophin/agonists , Dystrophin/deficiency , Gene Expression Profiling , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Phosphoric Monoester Hydrolases/blood , Phosphoric Monoester Hydrolases/genetics , Protein Kinases/blood , Protein Kinases/genetics , Protein Serine-Threonine Kinases , Proteomics/methods
17.
Hum Mol Genet ; 24(23): 6756-68, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26385637

ABSTRACT

Duchenne muscular dystrophy (DMD) is a classical monogenic disorder, a model disease for genomic studies and a priority candidate for regenerative medicine and gene therapy. Although the genetic cause of DMD is well known, the molecular pathogenesis of disease and the response to therapy are incompletely understood. Here, we describe analyses of protein, mRNA and microRNA expression in the tibialis anterior of the mdx mouse model of DMD. Notably, 3272 proteins were quantifiable and 525 identified as differentially expressed in mdx muscle (P < 0.01). Therapeutic restoration of dystrophin by exon skipping induced widespread shifts in protein and mRNA expression towards wild-type expression levels, whereas the miRNome was largely unaffected. Comparison analyses between datasets showed that protein and mRNA ratios were only weakly correlated (r = 0.405), and identified a multitude of differentially affected cellular pathways, upstream regulators and predicted miRNA-target interactions. This study provides fundamental new insights into gene expression and regulation in dystrophic muscle.


Subject(s)
Dystrophin/genetics , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Animals , Disease Models, Animal , Gene Expression Profiling , Genetic Therapy , Male , Mice , Mice, Inbred mdx , MicroRNAs/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/therapy , Mutation , Proteomics , RNA, Messenger/metabolism
18.
Methods Mol Biol ; 1324: 317-29, 2015.
Article in English | MEDLINE | ID: mdl-26202278

ABSTRACT

Cell-penetrating peptide (CPP)-mediated delivery of phosphorodiamidate morpholino oligomers (PMO) results in efficient exon skipping and has shown great promise as a potential therapy for Duchenne muscular dystrophy (DMD). However, large differences in efficiency have been observed between CPPs and in delivery to different tissues. Cellular trafficking has appeared to be an important determinant of activity. This chapter provides details of experimental procedures to monitor exon skipping efficiency and cellular trafficking of Pip6a-PMO, a recently developed and particularly efficient conjugate, in skeletal H2k cells and in primary cardiomyocytes from mdx mice. Similar procedures may be used in principle to evaluate any free or vector-associated oligonucleotide for exon skipping.


Subject(s)
Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/metabolism , Exons , Morpholinos/administration & dosage , Muscular Dystrophy, Duchenne/therapy , Transfection/methods , Amino Acid Sequence , Animals , Cell Culture Techniques/methods , Cell Separation/methods , Cell-Penetrating Peptides/chemical synthesis , Cells, Cultured , Electrophoresis, Agar Gel/methods , Genetic Therapy/methods , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Microscopy, Fluorescence/methods , Molecular Sequence Data , Morpholinos/genetics , Morpholinos/therapeutic use , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Polymerase Chain Reaction/methods
19.
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
20.
Nano Lett ; 15(7): 4364-73, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26042553

ABSTRACT

Antisense oligonucleotides (ASOs) have the potential to revolutionize medicine due to their ability to manipulate gene function for therapeutic purposes. ASOs are chemically modified and/or incorporated within nanoparticles to enhance their stability and cellular uptake, however, a major challenge is the poor understanding of their uptake mechanisms, which would facilitate improved ASO designs with enhanced activity and reduced toxicity. Here, we study the uptake mechanism of three therapeutically relevant ASOs (peptide-conjugated phosphorodiamidate morpholino (PPMO), 2'Omethyl phosphorothioate (2'OMe), and phosphorothioated tricyclo DNA (tcDNA) that have been optimized to induce exon skipping in models of Duchenne muscular dystrophy (DMD). We show that PPMO and tcDNA have high propensity to spontaneously self-assemble into nanoparticles. PPMO forms micelles of defined size and their net charge (zeta potential) is dependent on the medium and concentration. In biomimetic conditions and at low concentrations, PPMO obtains net negative charge and its uptake is mediated by class A scavenger receptor subtypes (SCARAs) as shown by competitive inhibition and RNAi silencing experiments in vitro. In vivo, the activity of PPMO was significantly decreased in SCARA1 knockout mice compared to wild-type animals. Additionally, we show that SCARA1 is involved in the uptake of tcDNA and 2'OMe as shown by competitive inhibition and colocalization experiments. Surface plasmon resonance binding analysis to SCARA1 demonstrated that PPMO and tcDNA have higher binding profiles to the receptor compared to 2'OMe. These results demonstrate receptor-mediated uptake for a range of therapeutic ASO chemistries, a mechanism that is dependent on their self-assembly into nanoparticles.


Subject(s)
Nanoparticles/chemistry , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacokinetics , Scavenger Receptors, Class A/metabolism , Animals , Base Sequence , Cell Line , Exons , Genetic Therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Micelles , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Scavenger Receptors, Class A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...